CD4+ T cell production of IL-10 and regulation of immune responses in aging

A dissertation submitted to the Graduate School of the

University of Cincinnati

in partial fulfillment of the requirements for the degree of

Doctor of Philosophy

Immunology Graduate Program, College of Medicine

2018 by

Maha Almanan M.D., Faculty of Medicine, University of Khartoum, Khartoum, Sudan

Committee Chair: David Hildeman, Ph.D. Abstract

The immune system plays a vital role in the protection against invading pathogens. A successful immune response is dictated by the fine balance between pro- and anti-

inflammatory mediators. With age the immune system undergoes a progressive

dysregulation due to the dramatic reduction in the naïve T cell pool, a relative clonal

expansion of memory T cells as well as accumulation of regulatory T cells. Another

aspect of this immune dysregulation is the persistent, low grade inflammation that

develops with age and is referred to as (inflammaging). Together, dysfunctional immune

responses and inflammaging are a great risk for serious age-related pathologies. Thus,

understanding the mechanisms regulating this age-related immune dysfunction is

critical for healthy aging.

Viral infections including latent cytomegalovirus (CMV) are thought to be one of driving forces of age-related alterations in immune function and inflammaging. While T cell responses are critical against CMV infection, the regulatory mechanisms that control latency are not well understood. Regulatory T cells are known to accumulate with age,

likely due to their enhanced survival. While Treg accumulated with age, their per cell

functionality remains controversial and possibly model dependent. Treg cells are important regulators of acute infections (including CMV). However, the impact of Treg on latent CMV is unclear. In chapter 3 we aimed at investigating the role of Foxp3+ regulatory T cells (Treg) in latent murine cytomegalovirus (MCMV) infection. We show

that Treg played divergent roles in the control of MCMV infection. In the spleen, Treg

antagonize CD8+ T cell effector function and promote viral persistence, while in the

II salivary gland Treg prevent IL-10 production from Foxp3- CD4+ T cells and limit viral

reactivation and replication. Together, work in chapter 3 broadens our understanding of

the homeostasis and functions of regulatory T cells and IL-10 with age.

Our discovery that Treg control IL-10 during latent CMV infection was intriguing. IL-10 is

a known regulatory cytokine, playing a major role in shaping immune responses and

regulating excessive inflammation. Thus, IL-10 may contribute to counter-regulation of

age-driven inflammation. In chapter 4 of this dissertation, we aimed to investigate the

role of IL-10 with age and further understand the cellular and molecular mechanisms

underlying its production. We provide evidence that aging favors the accrual of CD4+

Foxp3- IL-10+ T cells. Additionally, T follicular helper cells (Tfh) were major producers of

IL-10 in aged humans and mice which we have designated as Tfh10 cells. Interestingly, both IL-21 and IL-6 were indispensable for the accumulation of Tfh10 cells with age.

Finally, the loss of BCL6 in Foxp3- CD4+ T cells enhanced the production of IL-10 with

age. Results from this study highlight the dynamic regulatory/counter-regulatory balance

controlling age-related inflammation. More importantly, we show that despite age-driven

intrinsic defects in adaptive immune responses, blockade of IL-10 signaling is largely

sufficient to restore germinal center (GC) B cell responses in aged mice. Together work

from chapter 3 and 4 underscores the complexity of the unique age-related regulatory

networks that shape immune responses to latent infections, as well as vaccines.

III IV Acknowledgments

I would like to express my sincere gratitude to my mentor, Dr. David Hildeman. Thank you for your endless support and patience over the last five years, sense of humor, immense scientific knowledge and for challenging me to think independently. Thank you for believing in me at times when I didn't believe in myself, I cannot think of a better mentor to have. I don’t know how you do it, but I always looked up to you and I aspire to become a great role model and pass on what you've taught me to my own students one day. I just want you to know that I am a much better scientist because of you and for that I will always be grateful for the rest of my life.

I would like to acknowledge members of my committee who provided endless support and guidance throughout my PhD study. Dr. Claire Chougnet, I am grateful for all the invaluable scientific discussions which has greatly impacted my PhD training and I am also thankful for the time you have taken to offer career and life advice. Dr. Kasper

Hoebe, you have always challenged my scientific curiosity and for that I thank you. Dr.

Kris Steinbrecher, thank you for investing your time and personally assisting with my experiments. Dr. Rhonda Cardin, thank you for your patience while introducing me to the world of virology and assisting with experiments, I am truly grateful for all your support.

I would like to thank my lab members Allyson Sholl, Jana Raynor, Kun-Po Li, Matthew

Alder and Pulak Tripathi, who have grown to become my second family. You all were very helpful at various times at the beginning of me joining the lab and I have continued to succeed because of it. Cyd Castro, Sarah Jones, Rachel Walters, and Sharmilia

V Shanmuganad, thank you all for your contributions to my work at different times.

Especially within the last 2 years, we have grown to become one sisterhood in science and I am forever grateful.

I would also like to thank the Immunology Program who accepted me and gave me one of the best opportunities of my life and allowed me to meet some of the most wonderful people and some of my best friends. I especially thank Hesham Shihata, Kate Carroll,

Carolyn Rydyznski, and Courtney Jackson, for always caring, listening and being a shoulder to lean on throughout the years.

Lastly, I would like to thank my family for their continued love and support, especially my mother who was my biggest motivation to pursue my graduate studies. Even though you are no longer in the present, I hope that I have made you proud. Dad, you have taught me to continue to keep my head up and gave me your blessings and because of the way you have raised me, I feel I have become a better parent. To my children,

Yasmeen and Mosaab, at times it was tough to juggle being a student and being your mother. At times people have asked how do I manage, I just say to myself because of you. I am truly blessed to have you in my life.

VI Table Of Contents

Abstract ...... II Acknowledgments ...... V

Table of Contents ...... VII

List of Abbreviations ...... X Chapter 1: Introduction ...... 1 1. Age-dependent immune dysregulation ...... 1

1.1. Defects in innate immune-responses with age ...... 2

1.2. Defects in adaptive immune-responses with age ...... 6

1.2.1. B cells ...... 6

1.2.2. T cells ...... 8

(i) Homeostatic defects in T cells with age ...... 8

(ii) Functional defects in T cells with age ...... 9

1.3. Aging and its impact on vaccine responsiveness ...... 10

2. Regulatory T cells ...... 11

2.1. Thymus derived Treg (tTreg) ...... 12

2.2. Peripherally derived Treg (pTreg) ...... 12

2.3. Treg functionality and specialization ...... 14

2.4. Treg in aging ...... 15

3. Cytomegalovirus (CMV) 16

3.1. Acute CMV ...... 17

3.2. Latent CMV ...... 20

3.2.1. Cellular and molecular mechanisms controlling CMV reactivation

from latency ...... 21

3.2.2. CMV in aging ...... 22

3.2.3. Regulatory mechanisms controlling CMV ...... 23

VII (i) Treg in CMV ...... 23

(ii)IL-10 in CMV ...... 24

4. Interleukin-10 (IL-10) ...... 25

4.1. Transcriptional regulation of IL-10 production in CD4 T cells ...... 26 4.1.1. TCR mediated IL-10 expression ...... 26

4.1.2. Cytokine mediated IL-10 expression ...... 27

4.2. IL-10/IL-10R and signaling pathway ...... 29

4.3. Biological effects of IL-10 ...... 31

4.4. Aging and IL-10 ...... 33

5. T follicular helper (Tfh) cells ...... 34

5.1. Aging and Tfh cells ...... 35

6. Interleukin-6 (IL-6) ...... 37

6.1. IL-6 signaling pathway ...... 37

6.1.1. Classical pathway ...... 37

6.1.2. Trans-signaling pathway ...... 38

6.2. Biological effects of IL-6 ...... 39

6.3. Aging and IL-6 ...... 40

7. Interleukin-21 (IL-21) ...... 41

7.1. IL-21 signaling pathway ...... 41

7.2. Biological effects of IL-21 ...... 42

7.3. Aging and IL-21 ...... 43

Summary ...... 44

References ...... 46

Chapter 2: T-reg Homeostasis and Functions in Ageing ...... 81

VIII References ...... 97

Chapter 3: Tissue-specific control of latent CMV reactivation by regulatory T cells ...... 104

References ...... 126

Figures ...... 128

Chapter 4: IL-10 producing Tfh cells counter inflammaging but suppress humoral responses ...... 151

References ...... 172

Figures ...... 180

Chapter 5: Summary and future directions 200

Figures ...... 220

References ...... 224

IX List of Abbreviations

AID activation-induced cytidine deaminase

APCs antigen presenting cells

Bim Bcl-2 interacting mediator of cell death

CMV cytomegalovirus

CSR class switch recombination cTreg central Treg

DC Dendritic cell eTreg effector Treg

FoxP3 Fork-head box P3

GC germinal center

G-CSF granulocyte colony-stimulating factor

GM-CSF granulocyte-monocyte colony-stimulating factor

ICOS Inducible co-stimulator

IL-10 Interleukin 10

IL-21 Interleukin 21

IL-6 Interleukin 6

LPS lipopolysaccharide

NK natural killer cells

PAMPs pathogen-associated molecular patterns

PRRs pattern recognition receptors pTreg peripherally derived Treg

RTEs recent thymic emigrants

X STAT3 signal transduced and activator of transcription 3

TCR T cell receptor

Tfh T follicular helper cell

TLRs Toll-like receptors

Treg tTreg thymus derived regulatory T cell

XI Chapter 1

Introduction

1. Age-related immune dysregulation

The global population aged 60 years and over numbered 962 million in 2017, more than

twice as large as it was in 1980, with predictions of higher rates by 2050. More

importantly, the number of persons aged 80 years and over is projected to increase

more than threefold between 2017 and 2050, rising from 137 million to 425 million (1).

This demographic shift is expected to have a huge economic impact, particularly on health care.

One of the profound consequences of aging is the functional dysregulation in innate and adaptive immune responses (2). Importantly, a significant change favored by this immune dysregulation is the enhanced production of pro-inflammatory mediators resulting in inflammaging (3-6). Together, the immune dysregulation and inflammaging are suggested to be the underlying causes of most of the diseases of the elderly such as cancer, Alzheimer's disease, Parkinson's disease, multiple sclerosis and atherosclerosis (7-10). A major focus of aging research is to understand the cellular and molecular mechanisms underlying immune defects with age with an ultimate goal of improving the quality of life for older individuals and decreasing the costs of health care.

In this introductory section we will summarize the current understanding of the age- driven defects in the immune system and their relevance toward susceptibility to disease.

1 1.1. Defects in innate immune-responses with age

The effect of aging on the innate immune system in humans and mice is an area of

rapidly advancing research. Interestingly, accumulating evidence indicates that aging

has a profound impact on innate immune cell function and homeostasis, which in turn

greatly impacts the adaptive immune response. Innate immunity is mediated by various

cell types such as natural killer (NK) cells, neutrophils, macrophages, dendritic cells

(DCs) and others. In this section, we will summarize the recent knowledge on the

effects of aging on innate immunity.

NK cells are critical for eliminating cancerous and virally infected cells. Several defects

in NK cell maturation and function have been reported with age. For example, previous

studies provided evidence for the defective response of aged NK cell to cytokines like

IL-2, IL-12, IFN-α, IFN-β, and IFN-γ (11). More importantly, cytotoxic activity of human

and murine NK cells declined with age (12-14). Thus, both defects involoving NK- mediated cytotoxicity and/or cytokine production result in lack of functionality with age.

Other studies investigated the homeostasis of NK cells based on their maturation status. Interestingly, there is strong evidence for reduced proportion of mature NK cells with age, which appears to be due to the aged non-hematopoietic environment (15).

Together, these data suggest that aging has profound effects on NK cell functionality and maturation which might compromise immune responses to tumors and viral infections in elderly.

Neutrophils are another type of innate immune cell that plays a critical role in host defense. Several studies have investigated whether there are changes in neutrophil

2

function or homeostasis with age. The data remain controversial. For example, prior

work has shown no difference in neutrophil tissue adherence, migration, and

phagocytosis (16), suggesting marginal defects in neutrophil function with age.

However, another study comparing neutrophil function in young (23–35 years) and elderly (>65 years) individuals reported a reduction in the phagocytic index of neutrophils with age as well as a defect in neutrophil maturation (17).

In addition to their function, neutrophil survival and persistence at sites of inflammation

are also important controllers of infection and immunopathology. Given the increased

inflammation with age, several groups have investigated the survival of aged

neutrophils. Interestingly, aged neutrophils failed to survive under the effect of pro-

inflammatory mediators, such as granulocyte-monocyte colony-stimulating factor (GM-

CSF), granulocyte colony-stimulating factor (G-CSF ), and bacterial lipopolysaccharide

(LPS) (18). Combined, these data suggest that neutrophil function per se is minimally disturbed in aging; however, survival of aged neutrophils is shortened significantly, likely due to their altered response to inflammatory environment. Future studies will be

necessary to better understand the seemingly context-dependent functional changes in

neutrophils with age.

Macrophages are another integral component of the innate immune system. Several studies investigated the phenotype of macrophages and their activity in response to stimuli as readout for their functionality. While some studies reported functional impairment of macrophages with age, this phenotype seems to be reversible. For

example, aged macrophages were defective in their response to IFN-γ stimuli leading to

3

reduced nitric oxide production and increased susceptibility to parasitic infections as well as tumors (19-21). In contrast, other studies have argued that this age-related dysfunction in macrophages is reversible in response to in vitro treatment with multiple cytokines, suggesting that functional defects in macrophages with age are due to the environmental exposure (22). Interestingly, the aging environment favors a shift in adipose tissue macrophages towards an inflammatory phenotype with enhanced production of IL-6 and TNF-α (23). These latter data implicate adipose tissue

macrophages as key contributors to the enhanced inflammation observed with age.

Altogether, aged macrophages seem to acquire substantial plasticity that can shape

their functionality and phenotype in response to the aging environment.

Dendritic cells (DCs) are key participants in the generation of immunity and

maintenance of tolerance. Aging has a profound negative effect on DCs. For example, aged human DCs are less able to prime CD4+ and CD8+ T cell responses relative to young DCs (24, 25). Additionally, aged human DCs showed a reduction in phagocytosis as well as a decline in migration ability (26). Notably, activation of T cells requires the cooperation of DCs expressing MHC class I/II and costimulatory molecules including

CD80, CD86. Interestingly, several studies reported reduced expression of CD80,

CD86, and MHC II on murine DCs with age (27-29), suggesting defective functionality with age. In agreement, recent work has shown that a decline in the phagocytic activity of aged DCs is due to their mitochondrial metabolic dysfunction (30), suggesting that targeting metabolic pathways in DCs is a key for improving their functionality with age.

Together, age-associated functional deficits in DCs might play a substantial role in

increased susceptibility to disease with age.

4 The innate immune response relies on a large family of pattern recognition receptors

(PRRs) expressed on innate immune cells that target pathogen-associated molecular patterns (PAMPs) (31). Three major classes of innate immune receptors have been identified to date. 1) Toll-like receptors, which activate downstream signaling pathways such as (NF-κB) (32). 2) NOD-like receptors mediated by the inflammasome complex activation (33). 3) RIG-I which is critical for the regulation of an antiviral response (34).

Stimulation of innate cells (macrophages, polymorphonuclear cells, natural killer cells and dendritic cells) via these receptors is a critical step for subsequent activation of adaptive immune responses. Indeed, ligation of these receptors by their ligands results in the secretion of cytokines and chemokines that promotes the recruitment and activation/differentiation of adaptive immune cells. Interaction of T cells with antigen presenting cells (APCs) is a key factor for initiating the immune response that normally culminates in the eradication of pathogen and the generation of memory responses for future protection.

There are age-related defects reported in Toll-like receptor signaling and downstream cytokine production which have been mainly attributed to their altered expression. For example, in mice, reduction in the expression of almost all TLRs on macrophages was observed with age. Further, in vitro stimulated splenic macrophages from aged mice secreted lower levels of IL-6 and TNF-α compared to their young counterparts (35).

Similarly, in humans, age-associated decreases in surface expression of TLR1 was associated with deficits in TLR1-induced cytokine production from aged monocytes (36).

Additionally, a study by Panda et al. showed a substantial decrease in old, compared to young individuals in TNF-α production in myeloid (mDCs) and plasmacytoid (pDCs) in

5 response to TLR1/2, TLR2/6, TLR3, TLR5 stimulation. Findings from the study also indicated that the defects in TLR-induced cytokine production with age correlated with poor antibody responses to influenza immunization in elderly individuals (37), suggesting TLR-targeted therapies as a strategy to enhance vaccination efficacy.

Interestingly, poly I:C (TLR3 ligand) given as an adjuvant was sufficient to increase pro-inflammatory cytokines and improve the responsiveness of CD4+ T cells in aged mice (38), suggesting that age-related defects in CD4+ T cell responses are reversible.

Collectively, data emerging from human and mouse studies provide strong evidence for age-related immune dysregulation intrinsic to innate immune cells including aberrant expression and signaling events downstream of TLRs.

1.2. Defects in adaptive immune-responses with age

1.2.1. B cells

B cells and antibody-mediated immunity play an important role against pathogens.

Several studies have suggested defects in B cell homeostasis and function with age

(39). Age-related defects in B cells homeostasis begin with B cell development and persist throughout B cell lifespan. In the periphery, human studies reported that the percentage and absolute number of total B cells, defined by expression of the lineage marker CD19, decrease with age (40). Additionally, mouse studies have reported limited number of naive B cells in the periphery with reduced repertoire diversity due to the clonal expansion of memory B cells with age (41, 42), dominated by antigen- experienced CD5+ B cells (43).

6 A sizable amount of studies suggest that multiple factors during B cell development

contribute to the lack of naïve B cells in aged mice and humans. For example, studies

utilizing mixed bone marrow chimeras suggested that the reduction in B cells with age is

due to reduced hematopoietic stem cell (HSC) commitment to the B lineage, as well as

changes to the bone marrow (BM) microenvironment(44). Additionally, defective B cell

differentiation with age resulted also from reduction in B lineage transcription factors

(E2A and EBF1) as well as reduction in pre-BCR surrogate light chain (45). In contrast, another study showed that the proportions of early B cell precursors are maintained with age in humans (46). Thus, although it is likely that defects during B cell development contribute to altered B cell homeostasis and function with age, the data remain controversial and require verification in subsequent studies.

In addition to their homeostasis, B cell production of antibodies is essential for effective

humoral responses. Functionally, the quality of antibody production from B cells decline

with age resulting in lower-affinity antibodies and reduction in vaccination responses

(47). Indeed, a study by Agematsu et al. provided evidence for a reduction in plasma

cell differentiation and function of IgM memory B cells in response to pneumococcal

vaccine in elderly individuals compared to adults (48). Additionally, in vitro stimulated B

cells from elderly with anti-CD40/IL-4 were profoundly impaired in their capacity to

undergo class switch recombination (CSR) compared to their counterparts (49).

Altogether, B cells encounter major changes in their phenotype and function which contribute to increased risk of infections and loss of vaccination responses with age.

7

1.2.2. T cells

(i) Homeostatic defects in T cells with age

As T cells are key players of the adaptive immune system, extensive research in aging

has focused on their development in the thymus and their homeostasis and function in

the periphery. It is well established that, with age, the size of the thymus declines,

reaching a peak at puberty, followed by a progressive reduction in the output of naïve T

cells (50, 51). Thus, production of naïve T cells is significantly reduced with age (52).

Age-dependent decline in the production of naïve T cells is thought to impair the

response of elderly individuals to new antigens, likely putting the elderly at risk for

infections even with vaccination (53).

With age, both cortical and medullary regions decrease in volume with disorganization

of epithelial cell architecture and of the corticomedullary junction, accompanied by

replacement of lost stromal volume with adipose tissue (54). Furthermore, work from

Hale et al. using GFP to tag recent thymic emigrants (RTEs) in aged mice reported age-

dependent decline in RTE numbers (55). Interestingly, a previous study suggested that reduced thymic output of T cells might be compensated for by a reduction in the pro- apoptotic molecule Bim which enhanced naïve T cell life span in aged mice (56). These

data suggest that the progressive decline in naïve T cell production from the thymus

with age is balanced by enhanced survival in the periphery.

Another concept that has emerged to explain defective T cell responses with age is the

impingement of the naïve T cell repertoire by clonally expanded memory T cells.

8 Although CD4 T cells clonal expansion was observed with age, CD8 T cell clonal expansion is the most prominent (57). One explanation that was suggested for this oligoclonal expansion with age is the repeated exposure to specific viral antigens.

Indeed, expansion of terminally differentiated CD8+ effector cell (TEMRA) populations

has been largely linked to cytomegalovirus (CMV) and Epstein-Barr virus (EBV)

infection (58, 59). Another explanation that was proposed is the bystander proliferation

of CD8 T cell clones in response to Type I IFN production (60). Together, the reduction

in naive T cells in the peripheral pool and predominance of specific memory T cell

clones is thought to contribute to poor responsiveness to new antigens with age and

increased the risk of infections.

(ii) Functional defects in T cells with age

In addition to defects in development and homeostasis, aging also affects T cell

functionality. Indeed, aged T cells experience signaling defects, reduced proliferation

capacity, loss of synapse formation and altered trafficking (61). For example, early work

from Miller et al. documented several defects in TCR signaling including decline in the

activation of the Raf-1/MEK/ERK kinases and in JNK protein kinase with age (62).

Additionally, alteration of lipid raft composition with aging has been shown to contribute

to defective T cell responses (63). Importantly, decreased calcium flux after T cell

receptor engagement or mitogen stimulation along with decreased translocation of the

transcription factor NFATc to the nucleus with age (64), likely contributes to a decline in

T cell functionality with age. Together, aging adversely affect T and B cell homeostasis

9 and function resulting in the loss of ability to fight infections and decline in responses to

immunization.

1.3. Aging and its impact on vaccine responsiveness

Notably, both B and T cell changes with age are associated with a decline in the

antibody response to influenza vaccination (65, 66). Indeed, in young individuals the influenza vaccine provides 65-80% protection while the protection afforded in aged individuals is only 30-50% (67). As a result, 90% of the 10,000–40,000 deaths related to

influenza annually in the United States occur in persons aged ≥65 years. Successful

immunization against influenza is mediated mainly via B cells and T follicular helper

(Tfh) cells that act in concert to produce germinal centers (GCs) where activated B cells

undergo class switch recombination (CSR) and somatic hypermutation (SHM),

producing the high-affinity class-switched antibodies.

Several factors have been attributed to the loss of immunization responses in old

people. Some reports implicated defects in T cells, other reports implicated age-related

B cell defects. For example, work from Lefebvre et al. indicated that aged mice

experienced loss of vaccination responses and protection from influenza infection

compared to young mice. The authors attributed the loss of protection to a shift in aged

CD4 T cells toward Tfh cells rather than Th1 cells in the lungs which are required for

protection from lung infection (68).

B cells also accumulate intrinsic defects that correlate with the reduction in generation

of antibody responses to vaccines with age (65, 69, 70). For example, reduced

10 expression of activation-induced cytidine deaminase (AID) with age results in

diminished class switch recombination (CSR) and loss of production of higher affinity

antibodies in response to vaccination (49). Altogether, the effects of aging on CD4 T

cells and B cells significantly impact vaccination responsiveness. While age-related

intrinsic defects in T and B cell responses is well established in the literature, recent

data has suggested the existence of a suppressive environment that also contributes to

depressed adaptive immune responses with age.

2. Regulatory T cells

Regulatory T cells (Treg), classically defined by expression of high levels of interleukin 2

receptor-α (IL-2Rα/CD25) and transcription factor forkhead winged helix transcription

factor 3 (Foxp3), are indispensable for maintaining immune homeostasis (71). In

humans, mutations in the Foxp3 gene result in profound autoimmune diseases. Indeed,

the immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX)

is a direct consequence for an X-linked mutation in Foxp3 gene and Treg dysfunction in

humans. The disease was described for the first time in a large family with 19 affected

males across five generations (72). A similar discovery for the disease was made

simultaneously in mice deficient in Treg cells when it was found that these mice also

had defects in the Foxp3 gene (73, 74). Later, it was shown that depletion of Treg cells in mice expressing diphtheria toxin receptor under the control of the gene via administration of diphtheria toxin resulted in excessive myelo-proliferative disease and profound inflammation, providing strong evidence that Treg are indispensable for

maintaining peripheral immune homeostasis (75).

11

2.1. Thymus derived Treg (tTreg)

Following their discovery, extensive research was directed towards understanding the development and origin of Treg. Treg cells develop primarily in the thymus (thymus derived Treg, tTreg), or they can also be differentiated in the periphery (peripherally derived Treg, pTreg). During Treg differentiation, thymocytes bearing TCRs with high affinity for self-antigens presented in the thymus are selected into the Treg lineage at the immature CD4SP stage of thymic development (76). As a result of this selection, induction of Foxp3 is initiated leading to the differentiation of tTreg (77). Additionally, both CD80/CD86 and CD28 are required for the differentiation of tTreg cells (78, 79).

Further, cytokines like IL-2 play an essential role in tTreg development and Foxp3 expression (80, 81). Indeed, mice deficient in IL-2 or CD25 can only maintain half of the

Treg pool compared to WT counterpart (80). The incomplete loss of Treg in the absence of IL-2 suggested some redundancy in factors required for their development. Indeed, additional loss of IL-15 and IL-7 resulted in the complete absence of tTreg (82). Thus, evidence from previous research provided great insight into the complex signals that contribute to tTreg differentiation including cytokines and surface expression of co- stimulatory molecules.

2.2. Peripherally derived Treg (pTreg)

In addition to their development in the thymus, Foxp3 expression can be induced in naïve T cells post-thymic development driving their conversion into pTreg. This can occur through two major pathways, one involves TCR stimulation along with TGF-ß and the other involves TCR stimulation and retinoic acid. Differentiation of Treg from

12

CD4+CD25− naive T cell required co-stimulation with T cell receptors (TCRs) and

transforming growth factor β (TGF-ß). These pTreg are functionally similar to nTreg in

suppressing T cell proliferation and Th1 and Th2 cytokine production (83). Further

investigation for the molecular mechanisms involved in TGF-β mediated induction of

Foxp3 in naive CD4+CD25− indicated that IL-2 but not CD28 regulated the induction of

Foxp3 under TGF-β stimulatory conditions. The second pathway of pTreg conversion

occurs primarily in the gut and specifically Peyer’s patches. Here, the induction of pTreg

is mediated by CD103+ dendritic cells which act in the presence of TGF-β and retinoic acid to enhance Foxp3 induction (84). Thus, the peripheral Treg compartment is comprised of nTreg and pTreg both of which likely contribute to immune homeostasis.

As both tTreg and pTreg express CD4, CD25 and Foxp3, a significant amount of

research has investigated potential markers distinguishing tTreg from pTreg. A few

studies have identified the transcription factor Helios and the cell surface marker

neuropilin-1 as being markers that are specifically expressed on tTreg cells (85, 86).

Further complicating the matter in human CD4+ T cells is the fact that normal T cell activation can drive transient increases in Foxp3 expression. Therefore, other markers have been suggested to faithfully identify human Treg cells. For example, a lack of CD127 expression can be used to identify human Treg cells, especially when

combined with CD25. Thus, Low surface expression of CD127 allows for reliable

separation of human Treg (CD25+ CD127−) from effector T cells (CD25+CD127+) (87).

Together, the aforementioned studies describe our current understanding of Treg cells,

their development and their homeostasis. However, a substantial amount of is

beginning to unravel their function in aged mice and humans.

13

2.3. Treg functionality and specialization

Treg suppressive ability is a key factor for maintaining immune homeostasis. Extensive research has uncovered that FoxP3+ Treg utilize both secreted and cell surface molecules to elaborate their immune suppressive effects. Treg suppression can be mediated via expression of several regulatory molecules: CTLA-4, CD39, CD73, LAG-3,

TIGIT and GITR (71). Further, Treg also express several immune-suppressive

cytokines, such as: IL-10, TGF-ß and IL-35 (88, 89). Treg can also act on target cells

inducing their cytolysis via production of Granzyme B (90).

Notably, functionality of Treg not only depends on production and expression of

regulatory mediators but also on their ability to modulate their phenotype in response to immunological stimuli. Interestingly, the concept of functional specialization of Treg cells was recently introduced into the field (91). The concept states that specialization can be

induced in Treg cells through the activation of transcription factors as well as differential

expression of chemokine receptors under the effect of certain biological stimuli and in

conjunction with Foxp3 expression. Thus, leading to the emergence of new distinct

tissue-specific Treg cell which can play a suppressive or an effector role relevant to its

phenotype (92).

For example, the expression of Tbet in Treg under Th1 polarization conditions leads to the induction of CXCR3, which promotes Treg migration to the site of infection (93).

Similarly, the expression of BCL6 and CXCR5 in a subset of Treg supports the existence of T follicular regulatory cells which may function to suppress Tfh and GC B cells during the germinal center reaction (94). Recently, two distinct subsets of Treg with

14

unique homeostatic phenotype were identified by Campbell et al. CCR7(hi) CD44(lo)

CD62L(hi) Treg maintained by the production of IL-2 in secondary lymphoid tissues and

CD44(hi) CD62L(lo) CCR7(lo) Treg localized to non-lymphoid and are maintained via

the co-stimulatory receptor ICOS (inducible co-stimulator) (95). Thus, it is evident that

Treg can acquire functional plasticity in response to environmental stimuli which enable

them to control different types of inflammatory responses.

The role of Treg has been extensively investigated in many inflammatory conditions.

Despite the beneficial regulatory effect of Treg in limiting chronic inflammation and

deleterious immunopathology, Treg can also play a detrimental role promoting pathogen

persistence and limiting effector T cells responses (96). Thus, Treg seems to play a

dual role balancing both protective and aberrant hyperactive immune responses.

2.4. Treg in aging

Aging is associated with increased risk of chronic infections, cancer and loss of

vaccination responses. Although T cell intrinsic defects play a major role in the decline of immune responses with age, accrual of immune-suppressive Treg are suspected to be major contributors as well. Findings from human and mouse studies provide solid

evidence for age related changes in Treg homeostasis (97-99). It is generally accepted that accumulation of Treg cells with age is due to their enhanced survival and specifically the downregulation in pro-apoptotic molecule Bim (100). Additionally, recent work has shown that ICOS contributes to maintenance of Treg with age, likely by

inhibiting Bim-mediated death (101). Functionally, in vitro and in vivo studies have reported similar or increased suppressive capacity of aged Treg compared to their

15 young counterparts which might be model dependent (97, 98, 102). However, many questions about how Treg can regulate immune responses in aged animals need to be answered. Taken together, a significant role for Treg cells in regulating immune responses with age is increasingly appreciated. We will discuss the role of Treg in aging in the upcoming sections entitled Treg homeostasis with age and Treg function with age within chapter 2.

3. Cytomegalovirus (CMV)

Cytomegalovirus (CMV) is a ubiquitous beta-herpesvirus infecting 60 to 90% of adult individuals (103). Margaret Smith was the first to describe murine cytomegalovirus

(MCMV) and human cytomegalovirus (HCMV) in 1954 and 1956, respectively (104,

105). The virus was first called salivary gland virus (SGV) (104). When the virus enters the host target cell, viral genome enters the nucleus, followed by a process of replication of viral DNA and viral gene expression. The process ends in the assembly of a new virion that is ready to spread into a new host cell via lytic replication. CMV can infect many cell types including epithelial, endothelial, smooth muscle, and connective tissue cells, as well as specialized parenchymal cells in multiple tissues.

The virus is known to evade eradication by modulating the antiviral immune system using its own CMV-encoded proteins. For example, MCMV encodes a viral C-C chemokine MCK2 which facilitates viral dissemination. Indeed, MCK2 resulted in the recruitment of myeloid cells to the site of infection during early stages of viral entry.

Patrolling monocytes then become infected with the virus and, following their circulation, naturally disseminate the virus to multiple sites, including salivary glands, enabling

16

widespread distribution of the virus to multiple tissues (106-108). In addition, recruitment of CCR2 expressing monocytes in response to viral MCK2 has been shown to inhibit the differentiation and cytolytic activity of viral specific CD8 T cells via iNOS-mediated

NO production (109). On the other hand, HCMV expresses proteins like CD55 and

CD59 which target the complement system leading to inhibition of complement fixation, cell lysis and enhanced replication in fibroblasts (110). Together, diverse mechanisms are utilized by CMV to support viral replication, spreading, and targeting the immune system at multiple levels including recruitment of immune effector cells, complement activation and effector T cell responses.

3.1. Acute CMV

In general, viral infection typically activates both innate and adaptive arms of the

immune system, whose function is to ultimately contain, control or eradicate the viral

infection. However, in all cases and despite induction of strong immune responses, the virus is never cleared from the host and establishes a life-long latent infection. A major complication for viral persistence is the reactivation from latency which often ends in re- initiation of productive virus replication resulting in life-threatening diseases in immunosuppressed individuals.

The innate immune system is induced rapidly after CMV infection. The detection of virus is carried out by activation of Toll-like receptors (TLRs) which recognize viral products.

After ligation, several innate immune cells are activated including DCs, macrophages and NK cells. For example, TLR3 can recognize double-stranded RNA (dsRNA) and

TLR9 recognizes double-stranded DNA unmethylated at CpG motifs (111). Once

17 activated, a cascade of signaling pathways result in the production of alpha/beta interferon (IFN-α/β) by DCs and macrophages leading to the activation of natural killer

(NK) cells. Notably, using depletion and adoptive transfer approaches it has been shown that NK cells protect against MCMV (112, 113). More importantly, the strain susceptibility of MCMV in BALB/c mice compared to C57BL/6 has been attributed to the lack of Ly49H activation receptor on NK cells which binds the viral m157 protein resulting in potent NK cell activation and lysis of virally infected cells (114).

Together, TLR signaling is critical for NK cell activation during early MCMV infection. In addition, engagement of Ly49H on NK cells by m157 is indispensable for the control of the initial magnitude of viral replication and dissemination.

Several studies investigated the role of antibodies during CMV infection. Their role remains controversial. For example, antibodies seem not to play an important role in the control of acute MCMV replication. However, MCMV-specific antibodies are able to inhibit viral dissemination. Indeed, primary CMV infection results in the production of neutralizing antibodies against gH and gB, two viral proteins that are critical for viral attachment to host immune cells (115, 116). These neutralizing antibodies controlled viral dissemination into endothelial/epithelial cells (117). Additionally, the transfer of antibodies from HCMV sero-positive individual into newborn infants at risk of infection provided protection against HCMV infection (118). Nonetheless, future studies are needed to fully evaluate the role of antibodies in control of acute CMV infection.

T cells are critical for the control of CMV infection. Early work from Reddehase et al. indicated that CD8 T cells can prevent early replication of the virus in the lungs and the

18 adoptive transfer CD8 T cell from MCMV infected into immune-compromised mice was able to limit viral replication of an established lung infection (119). Additionally, epitope- specific CD8+ T cells are able to control acute MCMV infection in the spleen (120).

In humans, transfer of CMV-specific CD8 T cell clones from donors of bone marrow transplants were sufficient to prevent infection in recipients of allogeneic marrow transplantation after transplantation (121, 122). Although CD8 T cells can control the virus in many organs during MCMV infection they have limited control in the salivary gland due to the CMV-induced downregulation of MHC class I molecules in CMV infected cells in this organ (123). This CMV-driven loss of class I MHC expression makes the salivary gland somewhat of an immune privileged site, however, IFN-y production from CD4 T cells can partially substitute for CD8+ T cells and limit viral load in the salivary gland (124).

In contrast, to the role of effector CD4+ IFN-y-producing cells in controlling viral replication, CD4+ regulatory T cells (Foxp3+) and Foxp3- IL-10+ cells seem to have a negative impact. Indeed, depletion of Treg prior to acute infection resulted in enhanced

T-cell activation and reduced viral titers in the salivary glands (125). Additionally, specific deletion of IL-10 in CD4+ and not Foxp3+ T cells resulted in elevated IFN-y production by T cells and reduction in viral load in the salivary glands (125). Thus, CD4

T cells are pivotal for efficient viral control, while both Treg and IL-10 contribute to the ineffective control of infection resulting in CMV persistence in the salivary gland.

19

3.2. Latent CMV

CMV latency is defined by the presence of viral genomes in the absence of detectable

replication with the ability of the viral genome to reactivate (126). Latency can be

established in multiple organs and different types of cells including CD34+

hematopoietic progenitor cells, myeloid cells, alveolar macrophages, epithelial cells,

endothelial cells in the kidney, liver, heart, and spleen (127-130). Establishment of latency is a major goal for the virus to ensure persistence and transmission. To achieve such latency, CMV has developed several strategies. For example, HCMV encodes an

IL-10 homologue. Viral IL-10 shares minimum similarity to human IL-10 (27%) (131).

Although v-IL-10 binds to host IL-10R with limited affinity compared to h-IL-10, this binding enables the virus to evade host immune system recognition by modulating expression of MHC II on antigen presenting cells and by downregulating expression of

TNFα by monocytes (132-134). It is important to note that MCMV does not encode its

own IL-10 homolog but instead uses cellular IL-10 to modulate host immunity (135).

Another viral protein encoded by HCMV is gpUS2, which interferes with CD8 and CD4

T cells by targeting MHC class I and II for proteasome degradation (136). Once the

virus has evaded the recognition by the immune system and established latency it starts

to reactivate periodically. These periodic reactivation events are tightly controlled by

viral-specific CD8+ T cells for most tissues, but likely by viral-specific CD4+ T cells in

the salivary gland. Clearly, future studies will be needed to elucidate the cellular and

molecular mechanisms that regulate the maintenance of the viral genome during

latency and those that trigger reactivation.

20

3.2.1. Cellular and molecular mechanisms controlling CMV reactivation from

latency

It is widely accepted that chromatin remodeling of the viral major immediate-early

promoter (MIE) under different stimuli (e.g. cytokines) is a major inducer of reactivation

from latency (137). Notably, in vitro allogeneic stimulation of latently infected peripheral mononuclear cells resulted in full reactivation of HCMV. The cellular source in blood for viral reactivation was identified as latently infected monocytes expressing dendritic cell

markers (138). Additionally, Hummel et al. showed that allogeneic transplantation

resulted in the expression of the MCMV ie1 gene which was accompanied by increased

expression of transcripts encoding inflammatory cytokines, including TNF-α, IL-2, and

IFN-y. These data suggested that the induction of these cytokines drives the activation of the major immediate-early (MIE) gene and initiating reactivation from latency.

Interestingly, the authors were able to show that injecting TNF-α alone into latently infected mice was able to induce ie1 gene expression but did not lead to full viral reactivation (139). Thus, factors in addition to TNF-α likely control viral reactivation in vivo. Notably, full reactivation of MCMV was reported in the lungs following allogeneic transplantation combined with immunosuppression (e.g. irradiation) (140).

Thus, these studies provided evidence that reactivation from latency is potentially controlled via a multistep process which is initiated by induction of ie1 gene expression followed by full lytic replication in the presence of immunosuppressive environment.

While these studies argue that latency is controlled at transcriptional levels, there is also periodic, low-level reactivation from latency that is controlled primarily by CD8 T cells,

21 but also by CD4 T cells and NK cells. Indeed, the combined depletion of T cells and NK cells resulted in the reactivation of MCMV in multiple tissues (141). Thus, it is likely that latency and reactivation from latency is a tightly regulated process regulated by adaptive immune cells and cytokine production which combine to control transcription of immediate early viral genes. Nonetheless, mechanisms regulating CMV reactivation from latency remain poorly understood

3.2.2. CMV in aging

CMV infection is well tolerated by an immune competent host. However, CMV can result in great morbidity and mortality in organ and bone marrow transplant patients and HIV patients (142) (103). In addition, CMV is envisioned as one of the driving forces of the age-related immune dysfunction and inflammaging (143, 144). Also, CMV has been associated with multiple age-related pathologies such as type 2 diabetes, cancer and cardiovascular disease (145).

CMV infection persists latently in the host and reactivates periodically (146). Repeated reactivation of the virus drives the expansion of CMV-specific CD8+ T cell which can constitute up to 40% of CD8+ T cells in the peripheral blood of HCMV- seropositive individuals (147, 148). Although such expansion is expected to protect against reactivation and perhaps elimination of the virus, these cells have also been thought to be inducers of excessive inflammation. For example, stimulation of human PBMCs with

HCMV antigens resulted in increased production of IFN-y which was eight times greater in very old than in young subjects, suggesting that CMV infection might play detrimental effect with age by contributing to the age-related pro-inflammatory environment.

22

Despite the presence of functional HCMV-specific T cells with age, higher frequency

and greater viral load of CMV was reported in elderly (149). Related to the higher viral

load is that both the frequency and magnitude of CMV reactivation are increased with

age (142). These studies suggested that aging might favor the induction of a

suppressive environment that can modulate T cell responses and facilitate viral

persistence and reactivation. Nonetheless, major gaps remain in our understanding of

the cellular and molecular regulatory mechanisms controlling immune responses and

viral reactivation from latency with age.

3.2.3. Regulatory mechanisms controlling CMV

(i) Treg in CMV

CMV causes a persistent, lifelong latent infection despite a robust immune response. It

is unclear whether Teg cells have an impact on the insufficient host immune response

against the virus and whether they contribute to viral persistence. Clinical studies in

humans have allowed for important insights into the role of Treg in modulating immune

responses to CMV infection. For example, in vitro studies by Aandahl et al.

demonstrated that the depletion of Treg CD25+ cells from peripheral blood of CMV

sero-positive humans resulted in enhanced IFN-y and IFN-α production by syngeneic

cytomegalovirus (CMV)-specific CD8 T cells (150). These data suggested that Treg

might be limiting CD8 T cell responses. In agreement, murine studies indicated that

Treg interfere with an effective anti-mouse immune response and the depletion of Treg resulted in the control of viral persistence in the salivary gland (125). Thus Treg seems to negatively regulate immune responses to CMV favoring viral persistence. However,

23

the role of Treg in viral reactivation from latency is unclear. We will discuss the role of

Treg in latent MCMV in chapter 3.

(ii) IL-10 in CMV

IL-10 is a potent anti-inflammatory cytokine that regulates host immunity to acute as

well as chronic infections. IL-10 can also be exploited by pathogens to modulate the

immune response in favor of persistence in the host. Prior data shows that IL-10 controls CMV viral replication and regulates the establishment of latency (135), this has both benefits and negative consequences. For example, IL-10 has been shown to negatively regulate cellular immunity particularly in the salivary glands in favor of viral persistence (135). Indeed, interfering with IL-10R signaling restricted viral replication in the salivary gland (135). Likewise, the absence of IL-10 promoted the crosstalk between

NK cells, myeloid dendritic cells (mDCs) and in virus-specific CD4+ T cell responses resulting in reduced virus persistence in the salivary glands (151). To further investigate

the cell-specific role of IL-10 in MCMV, a recent study has shown that the specific

deletion of IL-10 in CD4 T cells, but not Foxp3+, cells led to enhanced antiviral T cell

responses and restricted MCMV persistence in salivary glands (125).

In contrast, other studies suggested a beneficial effect for IL-10 during acute MCMV.

For example, IL-10 controlled the weight loss and inflammatory cytokine production in

acute MCMV infection (152). The findings suggest that the role of IL-10 in MCMV is

complicated and might have differential effects on viral load based on IL-10 cellular

sources and the site of infection. We will discuss the role of IL-10 in latent MCMV in chapter 3.

24

4. Interleukin-10 (IL-10)

IL-10 is a regulatory cytokine, with potent anti-inflammatory effects. The regulatory

effect of IL-10 could play a beneficial role (e.g. resolution of inflammation and controlling

excessive immunopathology) or a detrimental role (e.g. persistence of pathogens and

inhibition of antitumor immunity) on host immunity. The discovery of IL-10 was first

made in 1989 by Mosmann and co-workers. The cytokine was described first to be produced by Type 2 helper cells (Th2) with a potent inhibitory effect on interleukin (IL-2)

and interferon (IFN-y) production and proliferation of Type1 helper cells (Th1) (153,

154).

IL-10 was initially referred to as “cytokine synthesis inhibitory factor” (CSIF). The gene

encoding IL-10 is located on chromosome 1 in humans and mice and covers a total of

5.1 kb pairs or 4.7 kb pairs, respectively. The IL-10 gene comprises five exons which

encode 178 amino acids. Both human and mouse IL-10 share 75% homology in the

amino acid sequences. As stated before, human IL-10 is known to share homology with some open reading frames in human viruses like Epstein–Barr virus, Herpes type 2

virus, Parapoxvirus, and Cytomegalovirus (155). To date, multiple cytokines have been shown to share structural similarity to IL-10 and collectively are known as IL-10 family

members (e.g. IL-10, IL-19, IL-20, IL-22, IL-24, and IL-26) (156, 157).

Since its discovery, extensive work with age has been focused on the cellular sources of IL-10 and their biological relevance. To date, multiple cells have the capability of producing IL-10, including adaptive immune cells (e.g T and B cells) and innate immune cells ( e.g dendritic cells (DC), γδ T cells, NK cells, mast cells, neutrophils, eosinophils),

25 keratinocytes (158), epithelia and some tumor cells (159). Together, IL-10 has a broad range of cellular sources and its production in subjected to various regulatory mechanisms that vary between tissues and under different inflammatory conditions.

4.1. Transcriptional regulation of IL-10 production in CD4 T cells

IL-10 expression is governed by multiple layers of regulation, including both transcriptional and post-transcriptional mechanisms. These mechanisms are influenced by a myriad of environmental stimuli (e.g. cytokines) as well as by tissues or cell types expressing IL-10. Importantly, the sequence identity and location of transcription factor binding sites in the IL-10 locus are well conserved in both humans and mice.

Understanding the transcriptional regulation of IL-10 is critical given the major roles of

IL-10 in controlling immune homeostasis and the potential therapeutic application of IL-

10 in multiple inflammatory diseases.

4.1.1. TCR mediated IL-10 expression

Following TCR stimulation multiple signaling pathways are initiated in CD4 T cells resulting in the induction of IL-10 expression through the activation of downstream transcription factors including AP-1 and c-Maf downstream of ERK, nuclear factor of activated T cells (NFAT) and NF-κB. After translocating from the cytoplasm into the nucleus NFAT is known to interact with AP-1 (compromised of JunB and c-Jun) and other transcriptional partners to promote IL-10 expression in CD4 T cells (160). In addition to AP-1, c-Maf activation in an ERK-dependent manner induces IL-10 expression in CD4 T cells. Further investigation indicated that c-Maf is able to bind and

26 trans-activate the IL-10 promoter (161, 162). Additionally, a role for NF-κB in IL-10 expression has been demonstrated in different Th subsets, including Th17, Th2 and

Th1 (163), however this pathway has been less characterized compared to NFAT1 and

AP-1.

4.1.2. Cytokine mediated IL-10 expression

The induction of IL-10 expression is complex, it can be controlled by multiple distinct cytokines and different CD4 T cell subsets. For example, IL-6, IL-21, IL-27, type 1 interferons, IL-12 and TGF-β can induce IL-10 production. Interestingly, IL-10 can induce its own expression (164). Further, the regulation of IL-10 expression following cytokine stimulation involves the activation of several STAT molecules and distinct transcription factors. Indeed, STAT3 is essential for IL-6-mediated IL-10 production

(165). Further, IL-27 as IL-12 cytokine family member can induce IL10 gene expression in a STAT3/STAT1 dependent manner (166-168). Similarly, IL-21 signaling via STAT3 is also required for the expression of IL-10 (169). Mechanistically, STAT3 activation under IL-21 and IL-27 has been shown to activate c-Maf expression. Interestingly, AHR synergizes with c-Maf to promote the production IL-10 in Tr1 cells. AHR forms a protein complex with c-Maf, to jointly trans-activate IL-10 promoter (170). Additionally, the expression of IL-10 in Tr1 cells is further regulated by ICOS expression. Indeed, ICOS- deficient T cells show reduced IL-10 production and c-Maf expression under IL-27 stimulatory conditions (171).

Another pathway that regulates IL-10 expression under the effect of IL-27/STAT3 is

Blimp-1. Indeed, Blimp-1 downstream of IL-27 is able to mediate IL-10 induction in CD4

27

T cells (172). In Treg cells, Blimp-1 and IRF-4 jointly induce the expression of IL-10

through direct binding to IL-10 promoter (173). Notably, BCL6 a known transcriptional repressor of Blimp-1, has been shown to negatively regulate IL-10 expression in CD4 T cells (174). Importantly, the negative regulation of IL-10 expression in CD4 T cells by

BCL6 seems to be independent of Blimp-1(175).

Similar to STAT-3, STAT-4 plays a role in the induction of IL-10 production in CD4 T

cells. Indeed, STAT-4 activation results in the development of IL-10/IFN-y double

producing cells under the effect of IL-12 and high antigen load (176). Further, SMAD

molecules in response to TGF-ß can induce IL-10 expression. Indeed, SMAD4 has been shown to activate IL-10 expression in Th1 cells (177), while SMAD3 been shown to induce IL-10 production in response to TGF-ß in Th2 cells (178).

In summary, several mediators are involved in the regulation of IL-10 expression in CD4

T cells including, TCR signaling and cytokine signaling through STAT/SMAD molecules promoting the activation and nuclear localization of several transcription factors

(Figure1). These different circuits allow the accessibility of IL-10 in response to multiple environmental stimuli, likely acting in different cell types or differentiation states to be able to combat various forms of inflammation. However, more work is necessary to understand the environmental contexts that promote IL-10 with age. In addition, as most environments possess multiple cytokine inputs, it is unclear how these multiple inputs contribute to IL-10 production in vivo. Although distinct CD4 T cell subsets require different stimulatory conditions, almost all CD4 T cell subsets have the capability to express IL-10 under differential regulatory mechanisms.

28

promoter

Figure1. Molecular events regulating IL-10 expression in CD4 T cells. IL-10 expression

in CD4 T cells is regulated by signals induced by TCR stimulation and cytokines acting via STAT/SMAD molecules and other transcription factors.

4.2. IL-10/IL-10R and signaling pathway

Similar to mice human IL-10 is a 35 kD homodimer consists of 178 amino acids, with an

N-terminal hydrophobic leader sequence 18 amino acids long (179). IL-10 serum half- life is between 1.1 and 2.6 hours (180). IL-10 signals through IL-10 receptor

(heterotetramer) which is composed of two unique chains of IL-10R1 (ligand binding), expressed on most hematopoietic cells at low levels, with higher levels on CD4+ memory than on naive T cells (181). It is also up-regulated by various stimuli on non- hematopoietic cells, such as fibroblasts and epithelial cells. IL-10R2 chains are constitutively expressed as two IL-10R2 (signal transduction) chains expressed on immune cells in addition to epithelial cells and keratinocytes. IL-10R2 is utilized as a

29

receptor by other cytokines from the IL-10 family, including IL-22, IL-26 and IL-28 and

IL-29 (181).

The activation of IL-10R through the binding of its ligand leads to the activation of downstream signaling involving Janus tyrosine kinases Jak1 constitutively associated with IL-10R1 and Tyk2 which is constitutively associated with IL-10R2 chain (182).

Ligand–driven phosphorylation of Jak1 promotes the phosphorylation and activation of

STAT3 and STAT1. STAT3 homodimers upon activation by IL-10 translocate into the nucleus and bind to STAT elements on promoters of various genes, including IL-10 itself and the suppressor of cytokine signaling (SOCSs) genes, amongst others. Of note, compared to other STAT3-activating cytokines (ie IL-6 and IL-21), IL-10 rapidly activates a more sustained phosphorylation of STAT3 while IL-6 mediated STAT3 activation is transient.

One explanation for this differential activation of STAT3 under IL-10 versus IL-6 stimulation might be the differential induction of SOCS3. Notably, SOCS3 among other

SOCSs molecules plays a major role in the negative regulation of various cytokines including IL-10, IL-6 and IL-21. SOCS3 is induced upon cytokine receptor activation via

STAT3-dependent mechanism. Such induction results in the initiation of a negative feedback loop resulting in the inhibition of the JAK/STAT signaling pathway. However,

SOCS3 can only interfere with JAK/STAT signaling pathway in the presence of receptors, such as gp130. Indeed, binding of SOCS3 to the Src homology phosphatase-

2 (SHP-2) binding domains of gp130 is required for the initiation of SOCS3 regulatory effect (183). Thus, the fact that IL-10 receptor chains do not contain a consensus motif

30

(SOCS box) for suppressor of cytokine signalling-3 (SOCS3) explains the loss of the negative regulatory effect of SOCS3 on IL-10/STAT3 sustained activation compared to

IL-6/STAT3 (184, 185).

The role of STAT3 in the IL-10 signaling pathway is indispensable. Indeed, previous work indicated that the specific deletion of STAT3 in myeloid lineage (macrophages, neutrophils, and dendritic cells) leads to the development of colitis similar to IL-10-/- mice

with enhanced Th1/IFN-y responses (186). The authors in the study concluded that the

increased production of IFN-y from CD4 T cells in STAT3 deficient mice would feedback

on myeloid cells and enhance the severity of colitis in the absence of IL-10/STAT3

signaling (187). In contrast to STAT3, little is known about the role of STAT1 in the

biological effects of IL-10. A study by Meraz et al. indicated that in contrast to STAT3,

deficiency of STAT1 did not result in inflammatory colitis. More importantly,

macrophages from STAT1 deficient mice were completely responsive to IL-10 (188).

Thus, the role of IL-10-driven STAT1 remains to be determined.

4.3. Biological effects of IL-10

IL-10 has a plethora of both negative and positive effects on immune responses.

Indeed, IL-10 can negatively regulate immune responses via inhibition of production of

tumor necrosis factor-α (TNF-α), IL-1, IL-12, IL-6 and granulocyte–macrophage colony-

stimulating factor (189). Additionally, IL-10 can inhibit nitric oxide (NO) production, and

expression of class II MHC and costimulatory molecules such as CD80/CD86 on

dendritic cells and macrophages (190). IL-10 can also inhibit T cells by regulating IL-2,

TNFα, and IL-5 production (191, 192), as well as enhancing the expression of soluble

31

TNFα receptors and IL-1 receptor antagonist (IL-1RA) (193, 194). Together these functions of IL-10 decrease adaptive immune responses.

In contrast, IL-10 has a stimulatory effect on NK cells, enhancing their proliferation, cytokine production, and cytotoxicity (195). Likewise, in vitro studies indicated that IL-10 can promote B cell survival and Immunoglobulin (Ig) class switch (196). Thus, the biological functions of IL-10 are very broad and complex and likely depend on the context of the immune response.

IL-10 plays an indispensable role in the regulation of intestinal homeostasis (197).

Notably, IL-10 production from Foxp3- type 1 regulatory T (Tr1) cells has been shown to

play a critical role in regulating a variety of inflammatory diseases including

development of colitis (198). Additionally, different subsets of T cells like Th1, Th17,

have been shown to experience some plasticity under the effect of inflammatory

environment and acquire the ability to produce IL-10 (199, 200), suggesting that IL-10

production from effector T cells might play a counter-regulatory role against excessive

inflammation.

Tight regulation of IL-10 is also critical to maintain human health. Overproduction of IL-

10 can lead to immune suppression, while underproduction of IL-10 can lead to

autoimmunity. For example, IL-10 produced by Th1 cells prevented excessive

immunopathology upon infection with cutaneous leishmaniasis and toxoplasma gondii

(199, 201). Also, IL-10 from gamma delta T cells prevented excessive tissue injury in

mice infected with Listeria (202). In contrast, excessive IL-10 production can inhibit the

immune responses during infections with Mycobacterium sp. and lymphocytic

32

choriomeningitis virus (LCMV) promoting chronic infection (203-206). On the other hand, IL-10 deficiency resulted in excessive inflammatory colitis by failing to limit Th1/17

responses (197) These data collectively show that dysregulated expression of IL-10 has

serious consequences.

4.4. Aging and IL-10

Aging is associated with low grade systemic inflammation, which contributes heavily to increased mortality and morbidity with age (3, 207, 208). Given its known anti-

inflammatory role, IL-10 might represent a key for balancing inflammaging. However,

the role of IL-10 with age is poorly understood. For example, the possession of -1082G

genotype, which is linked with IL-10 high production, is significantly increased in

centenarians (209), and similarly more in middle-aged controls than in age-matched patients with myocardial infarction (210). While an increase in serum IL-10 levels has been documented in aged humans (211), the cellular sources of IL-10 were not fully addressed. Thus, to date we lack full understanding of the cellular sources of IL-10 with age.

In addition to its role in balancing inflammation, IL-10 limits responses to infections and vaccines (212, 213). Interestingly, a balance between IFN-γ and IL-10 plays a role in vaccination responses with age. Indeed, the IFN-γ:IL-10 ratio correlates with protection against influenza infection. However, the IFNγ:IL-10 ratio in response to influenza virus challenge declined with age resulting in increased risk to influenza infection in vaccinated older adults (214, 215). Thus, strategies aimed at maximizing anti- inflammatory responses to vaccination, enhancing IFN-γ production and antagonizing

33

IL-10 responses are expected to support protection following vaccination with age.

Together, more research is needed to determine the cellular sources and effects of IL-

10 on immune responses with age. We will discuss these potential sources and functions of IL-10 in aging in chapter 4.

5. T follicular helper (Tfh) cells

Robust germinal centre (GC) formation directed by T follicular helper (Tfh) and B cell interaction is absolutely required for the generation of T-dependent humoral immunity

(216). Tfh cells are specialized CD4+ T cell subset that differentiates from naïve cells in response to antigen challenge (217). This differentiation of Tfh cells is indispensable for the activation of B cells, antibody class switching and affinity maturation which results in the production of high affinity circulating antibodies as well as formation of memory B cells (218). The generation of Tfh cells is an integrated process directed by transcriptional program that is including T cell receptor (TCR) signaling, T-B cell interaction and the cytokine environment. Indeed, the transcriptional repressor BCL-6 whose expression is promoted by IL-6 and IL-21 driven STAT-3 signaling is essential for the generation of Tfh cells (219).

Additionally, the cell surface marker CXCR5 is critical for Tfh localization to the GC and allows for B/T cell interaction (220). CXCL13 production by stromal cells binds to

CXCR5 on B and Tfh cells and promotes their migration to the B cell zone, where Tfh cells provide help in the form of CD40L and IL-21 production to GC B cells resulting in efficient antibody production (221). On the other hand, B cells provide help to Tfh cells through the expression of ICOS-L which is binds to ICOS and promotes Tfh

34

differentiation, BCL-6 expression and maintenance of Tfh cells (222). In addition, lymphocyte activation molecule-associated protein (SAP)/ signaling lymphocyte activation molecule (SLAM) signaling further stabilizes the crosstalk between Tfh and B cells. This interdependent relationship between Tfh and B cells is required for optimal B cell activation and GC formation (223). Given the critical role played by Tfh cells in shaping immune responses towards infections and to support the efficiency of vaccination, any changes that modify the function of Tfh cells or their differentiation could have a major impact on humoral responses. Notably, multiple age-related defects have been reported in Tfh cells which partly contributed to high risk of infections and loss in vaccination responses in elderly individuals. Together, Tfh generation and function are carfeully regulated via interlinked signals including, transcription factors, cytokines as well as expression of suface markers.

5.1. Aging and Tfh cells

Like other cell types, other studies have documented that aged mouse and human Tfh cells are phenotypically and likely functionally different compared to young Tfh cells.

Indeed, detrimental changes to aged Tfh cells have been reported in both human and

mouse studies. Some of these changes are extrinisic caused by the aging environment

and resulting in defective migration, localization and induction of Tfh cells, while other

are intrinsic changes result in impaired differentiation and function and hence poor B

cell help, reducing their ability to produce protective circulating antibodies. For example,

the aged environment failed to support the recruitment of naïve CD4+ T cells to

35

secondary lymphoid organs and hence the differentiation of Tfh cells resulting in poor

vaccination responses in old mice (224).

Additionally, a previous study showed that aged antigen-specific Tfh cells are defective

in their maturation status. The authors argued that aged Tfh cells are defective in their

homing to the GC because of their lower expression of CXCR5 compared to their young

counter parts (225). Further, enhanced production of IL-10 and lower expression of

ICOS on aged antigen -specific CD4+ T cells resulted in impaired B cells reponses

(225). Althogh this regulatory phenotype of CD4+ T cells have been attriabuted to their

Foxp3 expression, the existence of Foxp3- IL-10 producing cells with age is yet to be clear. Notably, a previous study has shown that aged mice demonstrated delayed clearance of influenza virus compared to young mice. The authors attributed this defect to the dyregulated differentiation of Th1 towards Tfh cells in the lungs of aged infected mice which resulted in increased lung viral load and inability to control primary lung infection as well as enhanced suscepitality to secondary infections (68). However, mechanisms by which Tfh cells altered viral clearance were not addressed in this study.

In humans, circulating CXCR5 + CD4 + T cells are increased in elderly individuals and this correlated with higher levels of IL-21 produced by aged PBMCs compared with young subjects (226). Mechanistically, prolonged activation of STAT-4 in response to IL-

12 in aged CD4+ T cells resulted in increased IL-21 with age (227). Together, several immunologic changes have been described in aged Tfh cells, including defective priming, differentiation and cytokine production. Importantly, knowledge of the age- related defects in Tfh cells and the contribution of the inflammatory environment is

36 critical to further advance our understanding of how aging impacts both infections and vaccine responses.

6. Interleukin-6 (IL-6)

6.1. IL-6 signaling pathway

Human IL-6 was first cloned in 1986 and given the name B cell stimulator factor BSF-2 due to its positive effects on promoting plasma cell growth (228). Multiple cell types can produce IL-6, nearly all stromal cells, vascular endothelial cells, and adipocytes at physiological conditions. Current research provided substantial insight into IL-

6 signaling pathways and its versatile biological activities.

Signaling by IL-6 is complex and involves signaling through cells that express IL-6Rα

(classical pathway) and cells that do not express IL-6Rα (trans-signaling). IL-6 signals through IL-6R which is composed of a unique receptor subunit (IL-6Rα) that binds IL-6.

This chain is mainly expressed on, hepatocytes and immune cells like T cells, neutrophils, and monocytes/macrophages (229). The receptor is also composed of two subunits of (gp130) for signal transduction (230). Notably, gp130 is also shared by several other cytokines like IL-11, IL-27, oncostatin-M and expressed ubiquitously on many cells.

6.1.1. Classical pathway

The binding of IL-6 to its membrane-bound receptor (mIL-6Rα) chain results in the formation of a complex with the homodimer gp130 subunits which triggers a

37

downstream signal cascade. Subsequently, kinases of the Jak family (Jak1, Jak2 and

Tyk2) are activated which leads to the activation and phosphorylation of STAT

molecules. As mentioned previously, IL-6 is known to signal primarily through STAT1,

STAT3 and to a lesser extent STAT2 (231). The phosphorylation of STAT molecules

leads to their dimerization and translocation to the nucleus to activate transcription of

genes containing STAT response elements. Notably, STAT3 activation induces the

suppressor of cytokine signaling 1 SOCS1 (232) and SOCS3 (233). SOCS1 binds to tyrosine-phosphorylated JAK, whereas SOCS3 binds to tyrosine-phosphorylated gp130 resulting in the termination of IL-6 cytokine signaling via a negative feedback loop.

6.1.2. Trans-signaling pathway

IL-6 can signal to cells that do not express membrane-bound receptor (mIL-6Rα). This occurs by IL-6 binding to soluble IL-6R (sIL-6Rα), followed by the IL-6/sIL-6R complex binding to gp130 subunits on the surface of cells, including those that do not formally express IL-6Rα on their surface. The binding of IL-6 to its soluble receptor (sIL-6Rα) can activate the dimerization of gp130 subunits similar to the classical pathway. sIL-

6Rα is generated under two mechanisms including, mRNA alternative splicing, and shedding of the membrane-bound IL-6R mediated by metalloproteases ADAM17 and

ADAM10 (234). Interestingly, a soluble form of gp130 is also produced and can act as

an antagonist for IL-6 trans-signaling pathway (235). Together, IL-6 is unique in

signaling via a membrane bound and a soluble receptor and it remains unclear how these distinct forms of signaling impact the biological effects of IL-6.

38

6.2. Biological effects of IL-6

IL-6 is involved in a broad spectrum of biological activities. Indeed, the production of IL-

6 early in the immune response regulates the production of acute phase proteins like

serum amyloid, C-reactive protein and fibrinogen (236, 237). It is generally accepted

that IL-6 activation through the trans-signaling pathway accounts for most of IL-6

biological activity early in the immune response. Additionally, IL-6 regulates

T cell subsets differentiation including Th17, Th2, Th1, Tfh, and Tr1 cells. For example,

IL-6 plays an indispensable role in Th17 differentiation together with transforming growth factor (TGF-β), IL-1β and IL-23 (238) (239). In Th2 cells, IL-6 enhances the expression of IL-4 and IL-13 (240-242), predominantly through an NFATc2 and c-Maf regulated pathway. In contrast to its positive regulatory effect on Th17 and Th2 cell differentiation, in Th1 cells, IL-6 interferes with IFN-y production by CD4 T cells mainly through upregulation of suppressors of cytokine signaling (SOCS1or SOCS3) (243).

One critical role of IL-6 is to induce the differentiation of a unique subset of cells termed

T follicular helper cells (Tfh). Tfh cells are critical for facilitating germinal center

reactions by providing help for B cells. Notably, IL-6 signaling through STAT3 is critical

for the upregulation of BCL6, the signature transcription factor of Tfh cells (244).

Importantly, in a Bcl6-dependent manner, Tfh cells express CXCR5 which enables them

to localize to GCs where they are in close proximity to B cells during the GC reaction

(245) (246). Although most models suggest that IL-6 is required for Tfh differentiation,

there have been a few reports of an IL-6-independent pathway for Tfh differentiation

(246). Current evidence suggests that this IL-6-independent pathway is predominantly

39

mediated by IL-21. Nonetheless, in most models, IL-6 is critical for the initial

development of Tfh cells.

In addition to its role in promoting Th17 and Tfh cells, IL-6 has also been shown to promote IL-10 production from CD4 T cells (247). Indeed, early studies investigating

Th17 cell differentiation have shown that both TGF-β and IL-6 drive the activation of

STAT3 leading to their co-production of IL-10 (162). Additionally, IL-6 was required for the induction of CD4 type 1 regulatory T cells (Tr1) and their production of IL-10 (247).

Further, IL-21/c-Maf expression was indispensable for the induction of IL-10 in Tr1 cells under IL-6 stimulatory conditions (247). Thus, based on the biological context, IL-6 plays a complex role in shaping the immune response.

6.3. Aging and IL-6

As a signature of the “inflammaging” process, several groups, including ours, have

reported increased systemic levels of IL-6 in aged mice and humans (101, 248-250).

Such increases in IL-6 are implicated in the pathogenesis of multiple age-related diseases including atherosclerosis, osteoporosis, sarcopenia, and cardiovascular disease, resulting in high mortality in the elderly (251-254). Importantly, increased

production of IL-6 with age is a major risk for age-associated frailty resulting in

functional decline and serious adverse health problems. A previous study has shown

that frail elderly had higher levels of IL-6 than non-frail, age-matched individuals (255).

Additionally, using IL-10 deficient mice as a model of frailty, old IL-10-/- mice with a frailty

phenotype had elevated IL-6 levels compared to the control WT mice (256). This latter

study also suggested a counter-regulatory role for the anti-inflammatory cytokine IL-10

40

in regulating IL-6 inflammation with age. Thus, findings from both human and mouse

studies provided evidence for the strong association of IL-6 to frailty and its adverse

outcome in elderly.

Interestingly, available data suggest that IL-6 and IL-10 may play important roles in

balancing inflammaging. The fact that centenarian males had an IL-10 polymorphism

associated with high IL-10 production in one study (209) and IL-6 serum levels were

increased among the same age group in another study is very intriguing (248). Although

measurements in both studies were independent, the findings suggested the possibility

that enhanced IL-10 levels might be a compensatory mechanism driven by increased

IL-6 to guard against excessive inflammation with age. Thus, investigating the role of IL-

6 in the induction of IL-10 with age may shed light on the development of more specific

therapeutic approaches targeting age related diseases, especially those where IL-6

plays a dominant role. Our data in chapter 4 will shed light on the relationship between

IL-6 and IL-10 in aged mice.

7. Interleukin-21 (IL-21)

7.1. IL-21 signaling pathway

Both IL-21 and its receptor were discovered in 2000 (257). IL-21 is produced primarily by activated CD4 T cells, natural killer T cells, and follicular T helper cells. The

heterodimeric receptor for IL-21 is composed of (IL-21R αβ) and the common cytokine

receptor (γc) chain, which is also a shared chain for IL-2, IL-4, IL-7, IL-9, and IL-15. IL-

21R can be expressed on many cells including, dendritic cells, macrophages, epithelial

41 cells, and keratinocytes. In addition to B, T, and NK cells (258). Signaling through IL-

21R is well known to function primarily by activating STAT transcription factors as well as ERK and PI3-kinase-Akt. IL-21 is known to activate STAT3 and STAT1 and to lesser extent STAT5 (259).

7.2. Biological effects of IL-21

As expected from the broad expression of its receptor, IL-21 is a pleiotropic cytokine with multiple actions on B cells, NK cells and CD4 T cells. On B cells, IL-21 seems to play opposing roles. For example, IL-21 plays an inhibitory effect on human and mouse

B cell proliferation and survival (260). However, it can enhance the differentiation of B cells into plasma cells in a STAT3 dependent manner (261). Additionally, mice with genetic loss in IL-21 receptor expression had reduced levels of serum IgG1 and high levels of IgE (262), similar to what has been reported in IL-21-deficient humans (263), suggesting an indispensable role for IL-21 in promoting immunoglobulin production.

Together, effects of IL-21 on B cells may be modulated by the differentiation status of B cells. Further, IL-21 plays a positive regulatory effect on NK cells. Indeed, NK cells experienced reduced cytolytic activity in humans deficient in IL-21R (264). Additionally,

IL-21 promoted survival and upregulated the expression of several surface receptors, including NKG2A/C/E on human NK cells (265), suggesting that IL-21 is enhancing survival and functionality of human NK cells.

In CD4 T cells, IL-21 can contribute to Th17 differentiation along with transforming growth factor (TGF-β) and IL-6 (266). Similarly, IL-21 together with IL-6 has been shown to be fundamental in Tfh development (245). Mechanistically, IL-21 is required for high

42 level expression of BCL6, which is important for Tfh differentiation and their sustained expression of CXCR5. Indeed, IL-21-KO mice showed reduced expression of BCL6

(267) and CXCR5 surface expression on CD4+ T cells was reduced in the absence of

IL-21 (268). In addition to its effect on BCL6 in Tfh cells, IL-21 can regulate other transcription factors involved in Tfh differentiation. For example, c-Maf, another transcription factor involved in the differentiation of Tfh cell is regulated by the expression of IL-21. Indeed, mice lacking c-Maf in the T cell compartment had reduced numbers of Tfh cells with limited secretion of high-affinity antibodies (269). Thus, IL-21 plays a significant role in promoting Tfh and Th17 differentiation.

In contrast to its role in the differentiation of inflammatory Th17 cells, IL21 can exert immunosuppressive effects via induction of IL-10-producing cells. Indeed, IL-21 is known to induce the expression of IL-10 in CD4 T cells (169). Several cytokines were shown to govern the production of IL-21 from T cells and subsequently the induction of

IL-10 in an autocrine manner. For example, IL-27 was shown to stimulate the production of IL-21 from type 1 regulatory T cells. The autocrine effect of IL-21 was then required to enhance and maintain the production of IL-10 from Tr1 cells (270). Similar effect of IL-

21 on IL-10 production was reported in IL-10-secreting type 1 regulatory T (Tr1) cells stimulated under IL-6 (247). Thus, the effects IL-21 on CD4 T cells differentiation can be complex and vary depending upon potential environmental stimuli.

7.3. Aging and IL-21

Aging is associated with severe alterations in immune function which contribute to increased susceptibility to infections and reduced response to vaccination. B and Tfh

43 cells are major players in regulating immune responses to vaccination with age. Given the role played by IL-21 in dictating both B and Tfh cell responses, some studies investigated the production of IL-21 with age. The data remains controversial. For example, a previous study demonstrated that aged PBMCs produced significantly higher levels of IL-21 compared with young individuals (226). In agreement, another study reported that CD4+ T cells from aged individuals secreted significantly higher levels of IL-21 on priming with dendritic cells (DCs) (227). Further examination revealed that the response of aged CD4+ T cells was due to alteration in IL-12/STAT4 pathway with age. In contrast, aged memory CD4 + T cells expressed lower levels of IL-21 after stimulation with anti-CD3/ anti-CD28 (271). The discrepancy in the findings might relate to approaches utilized and different experimental conditions For example, CD4 T cells were cultured with dendritic cells for 6 days, while the stimulation of CD4 T cells with anti-CD3/anti-CD28 lasted for 48hrs only. Nonetheless, whether IL-21 levels are altered with age or not remains unclear.

Summary

Aging is associated with profound immune dysregulation that contributes to the increased morbidity and mortality in elderly. Viral infections including CMV are implicated in accelerating age-related immune dysfunction. It is well established that

Treg and IL-10 play critical roles in regulating immune responses to acute CMV.

However the interplay between Treg and IL-10 producing cells during latent CMV infection is not clear. To date, we lack full understanding of factors contributing to latent

MCMV. More importantly, there is still a gap in knowledge about the role of Treg and IL-

44

10 on controlling the reactivation of latent MCMV. Our data suggest the existence of a

crosstalk between Treg and Foxp3- IL-10 producing T cells that differentially regulates the replication/reactivation of MCMV. Further, we show that the biologic outcome of this

dynamic between FoxP3+ CD4+ Treg cells and IL-10-producing FoxP3- CD4+ T cells depends upon the infected tissue. The data supporting this concept are the subject of chapter 3.

Inflammaging is characterized by increased production of inflammatory cytokines (e.g.

IL-6). On the other hand, production of anti-inflammatory cytokines such as IL-10 is envisioned as a strategy to counterbalance inflammaging. However, the role of IL-10 in aging is not clear. Notably, IL-6 and IL-21 normally promote Tfh development. Further, both cytokines are strong inducers of IL-10 production from CD4 T cells. However, it is unclear how Tfh development and IL-10 production proceed when the expression of IL-

6 and/or IL-21 becomes dysregulated. Our data provides strong evidence supporting the accrual of Tfh10 cells with age. Further, our data provide strong biological relevance for their existence through counterbalancing inflammaging on one hand and on the other hampering vaccination responses with age. The data supporting this concept are the subject of chapter 4.

45

References

1. UN. un.org/en/development/desa/population/publications/pdf/ageing/WPA2017

2017.

2. Fulop T, Dupuis G, Witkowski JM, Larbi A. The Role of Immunosenescence in the Development of Age-Related Diseases. Rev Invest Clin. 2016;68(2):84-91.

3. Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, et al.

Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mechanisms of ageing and development.

2007;128(1):92-105.

4. Fuentes E, Fuentes M, Alarcon M, Palomo I. Immune System Dysfunction in the

Elderly. An Acad Bras Cienc. 2017;89(1):285-99.

5. Lu Y, Tan CT, Nyunt MS, Mok EW, Camous X, Kared H, et al. Inflammatory and

immune markers associated with physical frailty syndrome: findings from Singapore

longitudinal aging studies. Oncotarget. 2016;7(20):28783-95.

6. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of

aging. Cell. 2013;153(6):1194-217.

7. Zhang X, Meng X, Chen Y, Leng SX, Zhang H. The Biology of Aging and Cancer:

Frailty, Inflammation, and Immunity. Cancer J. 2017;23(4):201-5.

8. Lord JM. The effect of ageing of the immune system on vaccination responses.

Hum Vaccin Immunother. 2013;9(6):1364-7.

9. Niccoli T, Partridge L. Ageing as a risk factor for disease. Curr Biol.

2012;22(17):R741-52.

46

10. Xia S, Zhang X, Zheng S, Khanabdali R, Kalionis B, Wu J, et al. An Update on

Inflamm-Aging: Mechanisms, Prevention, and Treatment. J Immunol Res.

2016;2016:8426874.

11. Ogata K, Yokose N, Tamura H, An E, Nakamura K, Dan K, et al. Natural killer

cells in the late decades of human life. Clinical immunology and immunopathology.

1997;84(3):269-75.

12. Nogusa S, Murasko DM, Gardner EM. Differential effects of stimulatory factors

on natural killer cell activities of young and aged mice. The journals of gerontology

Series A, Biological sciences and medical sciences. 2012;67(9):947-54.

13. Nogusa S, Ritz BW, Kassim SH, Jennings SR, Gardner EM. Characterization of age-related changes in natural killer cells during primary influenza infection in mice.

Mechanisms of ageing and development. 2008;129(4):223-30.

14. Facchini A, Mariani E, Mariani AR, Papa S, Vitale M, Manzoli FA. Increased number of circulating Leu 11+ (CD 16) large granular lymphocytes and decreased NK activity during human ageing. Clin Exp Immunol. 1987;68(2):340-7.

15. Shehata HM, Hoebe K, Chougnet CA. The aged nonhematopoietic environment impairs natural killer cell maturation and function. Aging cell. 2015;14(2):191-9.

16. MacGregor RR, Shalit M. Neutrophil function in healthy elderly subjects. J

Gerontol. 1990;45(2):M55-60.

17. Butcher SK, Chahal H, Nayak L, Sinclair A, Henriquez NV, Sapey E, et al.

Senescence in innate immune responses: reduced neutrophil phagocytic capacity and

CD16 expression in elderly humans. J Leukoc Biol. 2001;70(6):881-6.

47

18. Tortorella C, Piazzolla G, Spaccavento F, Pece S, Jirillo E, Antonaci S.

Spontaneous and Fas-induced apoptotic cell death in aged neutrophils. J Clin Immunol.

1998;18(5):321-9.

19. Albright JW, Albright JF. Ageing alters the competence of the immune system to control parasitic infection. Immunol Lett. 1994;40(3):279-85.

20. Ding A, Hwang S, Schwab R. Effect of aging on murine macrophages.

Diminished response to IFN-gamma for enhanced oxidative metabolism. J Immunol.

1994;153(5):2146-52.

21. Yoon P, Keylock KT, Hartman ME, Freund GG, Woods JA. Macrophage hypo- responsiveness to interferon-gamma in aged mice is associated with impaired signaling through Jak-STAT. Mechanisms of ageing and development. 2004;125(2):137-43.

22. Stout RD, Jiang C, Matta B, Tietzel I, Watkins SK, Suttles J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J Immunol. 2005;175(1):342-9.

23. Lumeng CN, Liu J, Geletka L, Delaney C, Delproposto J, Desai A, et al. Aging is associated with an increase in T cells and inflammatory macrophages in visceral adipose tissue. J Immunol. 2011;187(12):6208-16.

24. Briceno O, Lissina A, Wanke K, Afonso G, von Braun A, Ragon K, et al. Reduced naive CD8(+) T-cell priming efficacy in elderly adults. Aging cell. 2016;15(1):14-21.

25. Agrawal A, Agrawal S, Gupta S. Role of Dendritic Cells in Inflammation and Loss of Tolerance in the Elderly. Frontiers in immunology. 2017;8:896.

48

26. Agrawal A, Agrawal S, Cao JN, Su H, Osann K, Gupta S. Altered innate immune

functioning of dendritic cells in elderly humans: a role of phosphoinositide 3-kinase-

signaling pathway. J Immunol. 2007;178(11):6912-22.

27. Paula C, Motta A, Schmitz C, Nunes CP, Souza AP, Bonorino C. Alterations in dendritic cell function in aged mice: potential implications for immunotherapy design.

Biogerontology. 2009;10(1):13-25.

28. Li G, Smithey MJ, Rudd BD, Nikolich-Zugich J. Age-associated alterations in

CD8alpha+ dendritic cells impair CD8 T-cell expansion in response to an intracellular bacterium. Aging cell. 2012;11(6):968-77.

29. Moretto MM, Lawlor EM, Khan IA. Aging mice exhibit a functional defect in mucosal dendritic cell response against an intracellular pathogen. J Immunol.

2008;181(11):7977-84.

30. Chougnet CA, Thacker RI, Shehata HM, Hennies CM, Lehn MA, Lages CS, et al.

Loss of Phagocytic and Antigen Cross-Presenting Capacity in Aging Dendritic Cells Is

Associated with Mitochondrial Dysfunction. J Immunol. 2015;195(6):2624-32.

31. Rivera A, Siracusa MC, Yap GS, Gause WC. Innate cell communication kick- starts pathogen-specific immunity. Nat Immunol. 2016;17(4):356-63.

32. Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring

Harb Perspect Biol. 2009;1(6):a001651.

33. Kufer TA, Nigro G, Sansonetti PJ. Multifaceted Functions of NOD-Like Receptor

Proteins in Myeloid Cells at the Intersection of Innate and Adaptive Immunity. Microbiol

Spectr. 2016;4(4).

34. Barik S. What Really Rigs Up RIG-I? J Innate Immun. 2016;8(5):429-36.

49

35. Renshaw M, Rockwell J, Engleman C, Gewirtz A, Katz J, Sambhara S. Cutting

edge: impaired Toll-like receptor expression and function in aging. J Immunol.

2002;169(9):4697-701.

36. van Duin D, Mohanty S, Thomas V, Ginter S, Montgomery RR, Fikrig E, et al.

Age-associated defect in human TLR-1/2 function. J Immunol. 2007;178(2):970-5.

37. Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, et al. Age- associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol. 2010;184(5):2518-27.

38. Maue AC, Eaton SM, Lanthier PA, Sweet KB, Blumerman SL, Haynes L.

Proinflammatory adjuvants enhance the cognate helper activity of aged CD4 T cells. J

Immunol. 2009;182(10):6129-35.

39. Dempsey LA. Aging B cell repertoires. Nature Immunology. 2014;15:142.

40. Scholz JL, Diaz A, Riley RL, Cancro MP, Frasca D. A comparative review of aging and B cell function in mice and humans. Current opinion in immunology.

2013;25(4):504-10.

41. Dunn-Walters DK, Ademokun AA. B cell repertoire and ageing. Current opinion in immunology. 2010;22(4):514-20.

42. Johnson SA, Rozzo SJ, Cambier JC. Aging-dependent exclusion of antigen- inexperienced cells from the peripheral B cell repertoire. J Immunol. 2002;168(10):5014-

23.

43. LeMaoult J, Manavalan JS, Dyall R, Szabo P, Nikolic-Zugic J, Weksler ME.

Cellular basis of B cell clonal populations in old mice. J Immunol. 1999;162(11):6384-

91.

50

44. Labrie JE, 3rd, Sah AP, Allman DM, Cancro MP, Gerstein RM. Bone marrow microenvironmental changes underlie reduced RAG-mediated recombination and B cell generation in aged mice. J Exp Med. 2004;200(4):411-23.

45. Riley RL, Blomberg BB, Frasca D. B cells, E2A, and aging. Immunological reviews. 2005;205:30-47.

46. Rossi MI, Yokota T, Medina KL, Garrett KP, Comp PC, Schipul AH, Jr., et al. B lymphopoiesis is active throughout human life, but there are developmental age-related changes. Blood. 2003;101(2):576-84.

47. Frasca D, Blomberg BB. Aging affects human B cell responses. J Clin Immunol.

2011;31(3):430-5.

48. Shi Y, Yamazaki T, Okubo Y, Uehara Y, Sugane K, Agematsu K. Regulation of aged humoral immune defense against pneumococcal bacteria by IgM memory B cell. J

Immunol. 2005;175(5):3262-7.

49. Frasca D, Landin AM, Lechner SC, Ryan JG, Schwartz R, Riley RL, et al. Aging down-regulates the transcription factor E2A, activation-induced cytidine deaminase, and

Ig class switch in human B cells. J Immunol. 2008;180(8):5283-90.

50. Taub DD, Longo DL. Insights into thymic aging and regeneration. Immunological reviews. 2005;205:72-93.

51. Aw D, Taylor-Brown F, Cooper K, Palmer DB. Phenotypical and morphological changes in the thymic microenvironment from ageing mice. Biogerontology.

2009;10(3):311-22.

52. Goronzy JJ, Fang F, Cavanagh MM, Qi Q, Weyand CM. Naive T cell maintenance and function in human aging. J Immunol. 2015;194(9):4073-80.

51

53. Haynes L, Swain SL. Why aging T cells fail: implications for vaccination.

Immunity. 2006;24(6):663-6.

54. Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nat Immunol. 2004;5(2):133-9.

55. Hale JS, Boursalian TE, Turk GL, Fink PJ. Thymic output in aged mice. Proc Natl

Acad Sci U S A. 2006;103(22):8447-52.

56. Tsukamoto H, Huston GE, Dibble J, Duso DK, Swain SL. Bim dictates naive CD4

T cell lifespan and the development of age-associated functional defects. J Immunol.

2010;185(8):4535-44.

57. Schwab R, Szabo P, Manavalan JS, Weksler ME, Posnett DN, Pannetier C, et al.

Expanded CD4+ and CD8+ T cell clones in elderly humans. J Immunol.

1997;158(9):4493-9.

58. Nikolich-Zugich J, Slifka MK, Messaoudi I. The many important facets of T-cell

repertoire diversity. Nature reviews Immunology. 2004;4(2):123-32.

59. Wertheimer AM, Bennett MS, Park B, Uhrlaub JL, Martinez C, Pulko V, et al.

Aging and cytomegalovirus infection differentially and jointly affect distinct circulating T

cell subsets in humans. J Immunol. 2014;192(5):2143-55.

60. Tough DF, Borrow P, Sprent J. Induction of bystander T cell proliferation by

viruses and type I interferon in vivo. Science. 1996;272(5270):1947-50.

61. Goronzy JJ, Li G, Yu M, Weyand CM. Signaling pathways in aged T cells - a

reflection of T cell differentiation, cell senescence and host environment. Semin

Immunol. 2012;24(5):365-72.

52

62. Miller RA. Effect of aging on T lymphocyte activation. Vaccine.

2000;18(16):1654-60.

63. Fulop T, Jr., Douziech N, Larbi A, Dupuis G. The role of lipid rafts in T

lymphocyte signal transduction with aging. Ann N Y Acad Sci. 2002;973:302-4.

64. Miller RA. Calcium signals in T lymphocytes from old mice. Life sciences.

1996;59(5-6):469-75.

65. Frasca D, Diaz A, Romero M, Landin AM, Phillips M, Lechner SC, et al. Intrinsic defects in B cell response to seasonal influenza vaccination in elderly humans. Vaccine.

2010;28(51):8077-84.

66. Goronzy JJ, Fulbright JW, Crowson CS, Poland GA, O'Fallon WM, Weyand CM.

Value of immunological markers in predicting responsiveness to influenza vaccination in elderly individuals. J Virol. 2001;75(24):12182-7.

67. Nichol KL, Nordin JD, Nelson DB, Mullooly JP, Hak E. Effectiveness of influenza vaccine in the community-dwelling elderly. N Engl J Med. 2007;357(14):1373-81.

68. Lefebvre JS, Lorenzo EC, Masters AR, Hopkins JW, Eaton SM, Smiley ST, et al.

Vaccine efficacy and T helper cell differentiation change with aging. Oncotarget.

2016;7(23):33581-94.

69. Goodwin K, Viboud C, Simonsen L. Antibody response to influenza vaccination in

the elderly: a quantitative review. Vaccine. 2006;24(8):1159-69.

70. Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng NY, et al.

Limited efficacy of inactivated influenza vaccine in elderly individuals is associated with

decreased production of vaccine-specific antibodies. The Journal of clinical

investigation. 2011;121(8):3109-19.

53

71. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of

differentiation and function. Annual review of immunology. 2012;30:531-64.

72. Powell BR, Buist NR, Stenzel P. An X-linked syndrome of diarrhea,

polyendocrinopathy, and fatal infection in infancy. The Journal of pediatrics.

1982;100(5):731-7.

73. Lyon MF, Peters J, Glenister PH, Ball S, Wright E. The scurfy mouse mutant has

previously unrecognized hematological abnormalities and resembles Wiskott-Aldrich syndrome. Proc Natl Acad Sci U S A. 1990;87(7):2433-7.

74. Godfrey VL, Wilkinson JE, Rinchik EM, Russell LB. Fatal lymphoreticular disease in the scurfy (sf) mouse requires T cells that mature in a sf thymic environment: potential model for thymic education. Proc Natl Acad Sci U S A. 1991;88(13):5528-32.

75. Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol. 2007;8(2):191-7.

76. Apostolou I, Sarukhan A, Klein L, von Boehmer H. Origin of regulatory T cells with known specificity for antigen. Nat Immunol. 2002;3(8):756-63.

77. Lee HM, Bautista JL, Scott-Browne J, Mohan JF, Hsieh CS. A broad range of self-reactivity drives thymic regulatory T cell selection to limit responses to self.

Immunity. 2012;37(3):475-86.

78. Tai X, Cowan M, Feigenbaum L, Singer A. CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2. Nat Immunol. 2005;6(2):152-62.

79. Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, et al.

B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+

54

immunoregulatory T cells that control autoimmune diabetes. Immunity. 2000;12(4):431-

40.

80. Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY. A function for interleukin

2 in Foxp3-expressing regulatory T cells. Nat Immunol. 2005;6(11):1142-51.

81. Murawski MR, Litherland SA, Clare-Salzler MJ, Davoodi-Semiromi A.

Upregulation of Foxp3 expression in mouse and human Treg is IL-2/STAT5 dependent:

implications for the NOD STAT5B mutation in diabetes pathogenesis. Ann N Y Acad

Sci. 2006;1079:198-204.

82. Vang KB, Yang J, Mahmud SA, Burchill MA, Vegoe AL, Farrar MA. IL-2, -7, and -

15, but not thymic stromal lymphopoeitin, redundantly govern CD4+Foxp3+ regulatory T cell development. J Immunol. 2008;181(5):3285-90.

83. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003;198(12):1875-86.

84. Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med.

2007;204(8):1757-64.

85. Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, et al.

Expression of Helios, an Ikaros transcription factor family member, differentiates thymic- derived from peripherally induced Foxp3+ T regulatory cells. J Immunol.

2010;184(7):3433-41.

55

86. Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, Bailey-

Bucktrout S, et al. Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J Exp Med. 2012;209(10):1713-22, S1-19.

87. Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, Landay A, et al. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med. 2006;203(7):1693-700.

88. Chaturvedi V, Collison LW, Guy CS, Workman CJ, Vignali DA. Cutting edge:

Human regulatory T cells require IL-35 to mediate suppression and infectious tolerance.

J Immunol. 2011;186(12):6661-6.

89. Sakaguchi S, Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T. Regulatory T

cells: how do they suppress immune responses? International immunology.

2009;21(10):1105-11.

90. Loebbermann J, Thornton H, Durant L, Sparwasser T, Webster KE, Sprent J, et

al. Regulatory T cells expressing granzyme B play a critical role in controlling lung

inflammation during acute viral infection. Mucosal Immunol. 2012;5(2):161-72.

91. Campbell DJ, Koch MA. Phenotypical and functional specialization of FOXP3+

regulatory T cells. Nature reviews Immunology. 2011;11(2):119-30.

92. Wei S, Kryczek I, Zou W. Regulatory T-cell compartmentalization and trafficking.

Blood. 2006;108(2):426-31.

93. Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ.

The transcription factor T-bet controls regulatory T cell homeostasis and function during

type 1 inflammation. Nat Immunol. 2009;10(6):595-602.

56

94. Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, et al. Follicular

regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions.

Nature medicine. 2011;17(8):983-8.

95. Smigiel KS, Richards E, Srivastava S, Thomas KR, Dudda JC, Klonowski KD, et al. CCR7 provides localized access to IL-2 and defines homeostatically distinct

regulatory T cell subsets. J Exp Med. 2014;211(1):121-36.

96. Belkaid Y. Regulatory T cells and infection: a dangerous necessity. Nature

reviews Immunology. 2007;7(11):875-88.

97. Lages CS, Suffia I, Velilla PA, Huang B, Warshaw G, Hildeman DA, et al.

Functional Regulatory T Cells Accumulate in Aged Hosts and Promote Chronic

Infectious Disease Reactivation. The Journal of Immunology. 2008;181(3):1835-48.

98. Sharma S, Dominguez AL, Lustgarten J. High accumulation of T regulatory cells

prevents the activation of immune responses in aged animals. J Immunol.

2006;177(12):8348-55.

99. Zhao L, Sun L, Wang H, Ma H, Liu G, Zhao Y. Changes of CD4+CD25+Foxp3+

regulatory T cells in aged Balb/c mice. J Leukoc Biol. 2007;81(6):1386-94.

100. Chougnet CA, Tripathi P, Lages CS, Raynor J, Sholl A, Fink P, et al. A major role

for Bim in regulatory T cell homeostasis. J Immunol. 2011;186(1):156-63.

101. Raynor J, Karns R, Almanan M, Li KP, Divanovic S, Chougnet CA, et al. IL-6 and

ICOS Antagonize Bim and Promote Regulatory T Cell Accrual with Age. J Immunol.

2015;195(3):944-52.

102. Nishioka T, Shimizu J, Iida R, Yamazaki S, Sakaguchi S. CD4+CD25+Foxp3+ T

cells and CD4+CD25-Foxp3+ T cells in aged mice. J Immunol. 2006;176(11):6586-93.

57

103. Azevedo LS, Pierrotti LC, Abdala E, Costa SF, Strabelli TM, Campos SV, et al.

Cytomegalovirus infection in transplant recipients. Clinics (Sao Paulo). 2015;70(7):515-

23.

104. Smith MG. Propagation of salivary gland virus of the mouse in tissue cultures.

Proc Soc Exp Biol Med. 1954;86(3):435-40.

105. Smith MG. Propagation in tissue cultures of a cytopathogenic virus from human

salivary gland virus (SGV) disease. Proc Soc Exp Biol Med. 1956;92(2):424-30.

106. Noda S, Aguirre SA, Bitmansour A, Brown JM, Sparer TE, Huang J, et al.

Cytomegalovirus MCK-2 controls mobilization and recruitment of myeloid progenitor cells to facilitate dissemination. Blood. 2006;107(1):30-8.

107. Hokeness KL, Kuziel WA, Biron CA, Salazar-Mather TP. Monocyte chemoattractant protein-1 and CCR2 interactions are required for IFN-alpha/beta- induced inflammatory responses and antiviral defense in liver. J Immunol.

2005;174(3):1549-56.

108. Fleming P, Davis-Poynter N, Degli-Esposti M, Densley E, Papadimitriou J,

Shellam G, et al. The murine cytomegalovirus chemokine homolog, m131/129, is a determinant of viral pathogenicity. J Virol. 1999;73(8):6800-9.

109. Daley-Bauer LP, Wynn GM, Mocarski ES. Cytomegalovirus impairs antiviral

CD8+ T cell immunity by recruiting inflammatory monocytes. Immunity. 2012;37(1):122-

33.

110. Spear GT, Lurain NS, Parker CJ, Ghassemi M, Payne GH, Saifuddin M. Host cell-derived complement control proteins CD55 and CD59 are incorporated into the

58 virions of two unrelated enveloped viruses. Human T cell leukemia/lymphoma virus type

I (HTLV-I) and human cytomegalovirus (HCMV). J Immunol. 1995;155(9):4376-81.

111. Tabeta K, Georgel P, Janssen E, Du X, Hoebe K, Crozat K, et al. Toll-like receptors 9 and 3 as essential components of innate immune defense against mouse cytomegalovirus infection. Proc Natl Acad Sci U S A. 2004;101(10):3516-21.

112. Bukowski JF, Woda BA, Habu S, Okumura K, Welsh RM. Natural killer cell depletion enhances virus synthesis and virus-induced hepatitis in vivo. J Immunol.

1983;131(3):1531-8.

113. Bukowski JF, Warner JF, Dennert G, Welsh RM. Adoptive transfer studies demonstrating the antiviral effect of natural killer cells in vivo. J Exp Med.

1985;161(1):40-52.

114. Brown MG, Dokun AO, Heusel JW, Smith HR, Beckman DL, Blattenberger EA, et al. Vital involvement of a natural killer cell activation receptor in resistance to viral infection. Science. 2001;292(5518):934-7.

115. Macagno A, Bernasconi NL, Vanzetta F, Dander E, Sarasini A, Revello MG, et al.

Isolation of human monoclonal antibodies that potently neutralize human cytomegalovirus infection by targeting different epitopes on the gH/gL/UL128-131A complex. J Virol. 2010;84(2):1005-13.

116. Scrivano L, Sinzger C, Nitschko H, Koszinowski UH, Adler B. HCMV spread and cell tropism are determined by distinct virus populations. PLoS Pathog.

2011;7(1):e1001256.

117. Gerna G, Sarasini A, Patrone M, Percivalle E, Fiorina L, Campanini G, et al.

Human cytomegalovirus serum neutralizing antibodies block virus infection of

59

endothelial/epithelial cells, but not fibroblasts, early during primary infection. J Gen Virol.

2008;89(Pt 4):853-65.

118. Preventing transfusion-acquired cytomegalovirus infection in infants. Lancet.

1989;2(8655):160-1.

119. Reddehase MJ, Weiland F, Munch K, Jonjic S, Luske A, Koszinowski UH.

Interstitial murine cytomegalovirus pneumonia after irradiation: characterization of cells that limit viral replication during established infection of the lungs. J Virol.

1985;55(2):264-73.

120. Pahl-Seibert MF, Juelch M, Podlech J, Thomas D, Deegen P, Reddehase MJ, et al. Highly protective in vivo function of cytomegalovirus IE1 epitope-specific memory

CD8 T cells purified by T-cell receptor-based cell sorting. J Virol. 2005;79(9):5400-13.

121. Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med.

1995;333(16):1038-44.

122. Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD.

Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science. 1992;257(5067):238-41.

123. Lu X, Pinto AK, Kelly AM, Cho KS, Hill AB. Murine cytomegalovirus interference with antigen presentation contributes to the inability of CD8 T cells to control virus in the salivary gland. J Virol. 2006;80(8):4200-2.

60

124. Jonjic S, Mutter W, Weiland F, Reddehase MJ, Koszinowski UH. Site-restricted

persistent cytomegalovirus infection after selective long-term depletion of CD4+ T

lymphocytes. J Exp Med. 1989;169(4):1199-212.

125. Jost NH, Abel S, Hutzler M, Sparwasser T, Zimmermann A, Roers A, et al.

Regulatory T cells and T-cell-derived IL-10 interfere with effective anti-cytomegalovirus immune response. Immunol Cell Biol. 2014;92(10):860-71.

126. Liu XF, Wang X, Yan S, Zhang Z, Abecassis M, Hummel M. Epigenetic control of cytomegalovirus latency and reactivation. Viruses. 2013;5(5):1325-45.

127. Koffron AJ, Hummel M, Patterson BK, Yan S, Kaufman DB, Fryer JP, et al.

Cellular localization of latent murine cytomegalovirus. J Virol. 1998;72(1):95-103.

128. Mercer JA, Wiley CA, Spector DH. Pathogenesis of murine cytomegalovirus

infection: identification of infected cells in the spleen during acute and latent infections. J

Virol. 1988;62(3):987-97.

129. Reeves M, Sinclair J. Aspects of human cytomegalovirus latency and

reactivation. Curr Top Microbiol Immunol. 2008;325:297-313.

130. Reeves MB, MacAry PA, Lehner PJ, Sissons JG, Sinclair JH. Latency, chromatin

remodeling, and reactivation of human cytomegalovirus in the dendritic cells of healthy

carriers. Proc Natl Acad Sci U S A. 2005;102(11):4140-5.

131. Kotenko SV, Saccani S, Izotova LS, Mirochnitchenko OV, Pestka S. Human cytomegalovirus harbors its own unique IL-10 homolog (cmvIL-10). Proc Natl Acad Sci

U S A. 2000;97(4):1695-700.

61

132. Spencer JV. The cytomegalovirus homolog of interleukin-10 requires

phosphatidylinositol 3-kinase activity for inhibition of cytokine synthesis in monocytes. J

Virol. 2007;81(4):2083-6.

133. Spencer JV, Lockridge KM, Barry PA, Lin G, Tsang M, Penfold ME, et al. Potent

immunosuppressive activities of cytomegalovirus-encoded interleukin-10. J Virol.

2002;76(3):1285-92.

134. de Waal Malefyt R, Haanen J, Spits H, Roncarolo MG, te Velde A, Figdor C, et al. Interleukin 10 (IL-10) and viral IL-10 strongly reduce antigen-specific human T cell proliferation by diminishing the antigen-presenting capacity of monocytes via downregulation of class II major histocompatibility complex expression. J Exp Med.

1991;174(4):915-24.

135. Humphreys IR, de Trez C, Kinkade A, Benedict CA, Croft M, Ware CF.

Cytomegalovirus exploits IL-10-mediated immune regulation in the salivary glands. J

Exp Med. 2007;204(5):1217-25.

136. Jones TR, Sun L. Human cytomegalovirus US2 destabilizes major histocompatibility complex class I heavy chains. J Virol. 1997;71(4):2970-9.

137. Dorsch-Hasler K, Keil GM, Weber F, Jasin M, Schaffner W, Koszinowski UH. A long and complex enhancer activates transcription of the gene coding for the highly abundant immediate early mRNA in murine cytomegalovirus. Proc Natl Acad Sci U S A.

1985;82(24):8325-9.

138. Soderberg-Naucler C, Fish KN, Nelson JA. Reactivation of latent human cytomegalovirus by allogeneic stimulation of blood cells from healthy donors. Cell.

1997;91(1):119-26.

62

139. Hummel M, Zhang Z, Yan S, DePlaen I, Golia P, Varghese T, et al. Allogeneic

transplantation induces expression of cytomegalovirus immediate-early genes in vivo: a

model for reactivation from latency. J Virol. 2001;75(10):4814-22.

140. Kurz SK, Reddehase MJ. Patchwork pattern of transcriptional reactivation in the

lungs indicates sequential checkpoints in the transition from murine cytomegalovirus

latency to recurrence. J Virol. 1999;73(10):8612-22.

141. Polic B, Hengel H, Krmpotic A, Trgovcich J, Pavic I, Luccaronin P, et al.

Hierarchical and redundant lymphocyte subset control precludes cytomegalovirus replication during latent infection. J Exp Med. 1998;188(6):1047-54.

142. Stowe RP, Kozlova EV, Yetman DL, Walling DM, Goodwin JS, Glaser R. Chronic herpesvirus reactivation occurs in aging. Experimental gerontology. 2007;42(6):563-70.

143. Brunner S, Herndler-Brandstetter D, Weinberger B, Grubeck-Loebenstein B.

Persistent viral infections and immune aging. Ageing Res Rev. 2011;10(3):362-9.

144. Fulop T, Larbi A, Pawelec G. Human T cell aging and the impact of persistent viral infections. Frontiers in immunology. 2013;4:271.

145. Aiello AE, Chiu YL, Frasca D. How does cytomegalovirus factor into diseases of aging and vaccine responses, and by what mechanisms? Geroscience. 2017;39(3):261-

71.

146. Sinclair J, Sissons P. Latency and reactivation of human cytomegalovirus. J Gen

Virol. 2006;87(Pt 7):1763-79.

147. Khan N, Hislop A, Gudgeon N, Cobbold M, Khanna R, Nayak L, et al.

Herpesvirus-specific CD8 T cell immunity in old age: cytomegalovirus impairs the response to a coresident EBV infection. J Immunol. 2004;173(12):7481-9.

63

148. Khan N, Shariff N, Cobbold M, Bruton R, Ainsworth JA, Sinclair AJ, et al.

Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality

in healthy elderly individuals. J Immunol. 2002;169(4):1984-92.

149. Thomasini RL, Pereira DS, Pereira FSM, Mateo EC, Mota TN, Guimaraes GG, et

al. Aged-associated cytomegalovirus and Epstein-Barr virus reactivation and

cytomegalovirus relationship with the frailty syndrome in older women. PLoS One.

2017;12(7):e0180841.

150. Aandahl EM, Michaelsson J, Moretto WJ, Hecht FM, Nixon DF. Human CD4+

CD25+ regulatory T cells control T-cell responses to human immunodeficiency virus and cytomegalovirus antigens. J Virol. 2004;78(5):2454-9.

151. Mandaric S, Walton SM, Rulicke T, Richter K, Girard-Madoux MJ, Clausen BE, et

al. IL-10 suppression of NK/DC crosstalk leads to poor priming of MCMV-specific CD4 T

cells and prolonged MCMV persistence. PLoS Pathog. 2012;8(8):e1002846.

152. Oakley OR, Garvy BA, Humphreys S, Qureshi MH, Pomeroy C. Increased weight

loss with reduced viral replication in interleukin-10 knock-out mice infected with murine

cytomegalovirus. Clin Exp Immunol. 2008;151(1):155-64.

153. Moore KW, O'Garra A, de Waal Malefyt R, Vieira P, Mosmann TR. Interleukin-10.

Annual review of immunology. 1993;11:165-90.

154. Fiorentino DF, Bond MW, Mosmann TR. Two types of mouse T helper cell. IV.

Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J Exp Med.

1989;170(6):2081-95.

64

155. Kim JM, Brannan CI, Copeland NG, Jenkins NA, Khan TA, Moore KW. Structure

of the mouse IL-10 gene and chromosomal localization of the mouse and human genes.

J Immunol. 1992;148(11):3618-23.

156. Volk H, Asadullah K, Gallagher G, Sabat R, Grutz G. IL-10 and its homologs:

important immune mediators and emerging immunotherapeutic targets. Trends in

immunology. 2001;22(8):414-7.

157. Sabat R. IL-10 family of cytokines. Cytokine & growth factor reviews.

2010;21(5):315-24.

158. Sabat R, Grutz G, Warszawska K, Kirsch S, Witte E, Wolk K, et al. Biology of

interleukin-10. Cytokine & growth factor reviews. 2010;21(5):331-44.

159. Gastl GA, Abrams JS, Nanus DM, Oosterkamp R, Silver J, Liu F, et al.

Interleukin-10 production by human carcinoma cell lines and its relationship to interleukin-6 expression. Int J Cancer. 1993;55(1):96-101.

160. Brownlie RJ, Zamoyska R. T cell receptor signalling networks: branched, diversified and bounded. Nature reviews Immunology. 2013;13(4):257-69.

161. Cao S, Liu J, Song L, Ma X. The protooncogene c-Maf is an essential transcription factor for IL-10 gene expression in macrophages. J Immunol.

2005;174(6):3484-92.

162. Xu J, Yang Y, Qiu G, Lal G, Wu Z, Levy DE, et al. c-Maf regulates IL-10

expression during Th17 polarization. J Immunol. 2009;182(10):6226-36.

163. Gerondakis S, Grumont R, Gugasyan R, Wong L, Isomura I, Ho W, et al.

Unravelling the complexities of the NF-kappaB signalling pathway using mouse

knockout and transgenic models. Oncogene. 2006;25(51):6781-99.

65

164. Staples KJ, Smallie T, Williams LM, Foey A, Burke B, Foxwell BM, et al. IL-10 induces IL-10 in primary human monocyte-derived macrophages via the transcription factor Stat3. J Immunol. 2007;178(8):4779-85.

165. Stumhofer JS, Silver JS, Laurence A, Porrett PM, Harris TH, Turka LA, et al.

Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10. Nat

Immunol. 2007;8(12):1363-71.

166. Wang H, Meng R, Li Z, Yang B, Liu Y, Huang F, et al. IL-27 induces the differentiation of Tr1-like cells from human naive CD4+ T cells via the phosphorylation of

STAT1 and STAT3. Immunol Lett. 2011;136(1):21-8.

167. Pot C, Jin H, Awasthi A, Liu SM, Lai CY, Madan R, et al. Cutting edge: IL-27 induces the transcription factor c-Maf, cytokine IL-21, and the costimulatory receptor

ICOS that coordinately act together to promote differentiation of IL-10-producing Tr1 cells. Journal of immunology. 2009;183(2):797-801.

168. Awasthi A, Carrier Y, Peron JP, Bettelli E, Kamanaka M, Flavell RA, et al. A dominant function for interleukin 27 in generating interleukin 10-producing anti- inflammatory T cells. Nat Immunol. 2007;8(12):1380-9.

169. Spolski R, Kim HP, Zhu W, Levy DE, Leonard WJ. IL-21 mediates suppressive effects via its induction of IL-10. J Immunol. 2009;182(5):2859-67.

170. Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D, et al. The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol. 2010;11(9):854-61.

171. Pot C, Apetoh L, Awasthi A, Kuchroo VK. Induction of regulatory Tr1 cells and inhibition of T(H)17 cells by IL-27. Semin Immunol. 2011;23(6):438-45.

66

172. Iwasaki Y, Fujio K, Okamura T, Yanai A, Sumitomo S, Shoda H, et al. Egr-2 transcription factor is required for Blimp-1-mediated IL-10 production in IL-27-stimulated

CD4+ T cells. Eur J Immunol. 2013;43(4):1063-73.

173. Cretney E, Xin A, Shi W, Minnich M, Masson F, Miasari M, et al. The transcription factors Blimp-1 and IRF4 jointly control the differentiation and function of effector regulatory T cells. Nat Immunol. 2011;12(4):304-11.

174. Hollister K, Kusam S, Wu H, Clegg N, Mondal A, Sawant DV, et al. Insights into the role of Bcl6 in follicular Th cells using a new conditional mutant mouse model.

Journal of immunology. 2013;191(7):3705-11.

175. Xie MM, Koh BH, Hollister K, Wu H, Sun J, Kaplan MH, et al. Bcl6 promotes follicular helper T-cell differentiation and PD-1 expression in a Blimp1-independent manner in mice. Eur J Immunol. 2017;47(7):1136-41.

176. Saraiva M, Christensen JR, Veldhoen M, Murphy TL, Murphy KM, O'Garra A.

Interleukin-10 production by Th1 cells requires interleukin-12-induced STAT4 transcription factor and ERK MAP kinase activation by high antigen dose. Immunity.

2009;31(2):209-19.

177. Kitani A, Fuss I, Nakamura K, Kumaki F, Usui T, Strober W. Transforming growth factor (TGF)-beta1-producing regulatory T cells induce Smad-mediated interleukin 10 secretion that facilitates coordinated immunoregulatory activity and amelioration of TGF- beta1-mediated fibrosis. J Exp Med. 2003;198(8):1179-88.

178. Blokzijl A, ten Dijke P, Ibanez CF. Physical and functional interaction between

GATA-3 and Smad3 allows TGF-beta regulation of GATA target genes. Curr Biol.

2002;12(1):35-45.

67

179. Vieira P, de Waal-Malefyt R, Dang MN, Johnson KE, Kastelein R, Fiorentino DF,

et al. Isolation and expression of human cytokine synthesis inhibitory factor cDNA

clones: homology to Epstein-Barr virus open reading frame BCRFI. Proc Natl Acad Sci

U S A. 1991;88(4):1172-6.

180. Li MC, He SH. IL-10 and its related cytokines for treatment of inflammatory bowel

disease. World journal of gastroenterology : WJG. 2004;10(5):620-5.

181. Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A. Interleukin-10 and the interleukin-10 receptor. Annual review of immunology. 2001;19:683-765.

182. Liu Y, Wei SH, Ho AS, de Waal Malefyt R, Moore KW. Expression cloning and

characterization of a human IL-10 receptor. J Immunol. 1994;152(4):1821-9.

183. Fujimoto M, Naka T. Regulation of cytokine signaling by SOCS family molecules.

Trends in immunology. 2003;24(12):659-66.

184. Niemand C, Nimmesgern A, Haan S, Fischer P, Schaper F, Rossaint R, et al.

Activation of STAT3 by IL-6 and IL-10 in primary human macrophages is differentially modulated by suppressor of cytokine signaling 3. J Immunol. 2003;170(6):3263-72.

185. Murray PJ. The JAK-STAT signaling pathway: input and output integration. J

Immunol. 2007;178(5):2623-9.

186. Takeda K, Clausen BE, Kaisho T, Tsujimura T, Terada N, Forster I, et al.

Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity. 1999;10(1):39-49.

187. Kobayashi M, Kweon MN, Kuwata H, Schreiber RD, Kiyono H, Takeda K, et al.

Toll-like receptor-dependent production of IL-12p40 causes chronic enterocolitis in

68

myeloid cell-specific Stat3-deficient mice. The Journal of clinical investigation.

2003;111(9):1297-308.

188. Meraz MA, White JM, Sheehan KC, Bach EA, Rodig SJ, Dighe AS, et al.

Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity

in the JAK-STAT signaling pathway. Cell. 1996;84(3):431-42.

189. Williams LM, Ricchetti G, Sarma U, Smallie T, Foxwell BM. Interleukin-10

suppression of myeloid cell activation--a continuing puzzle. Immunology.

2004;113(3):281-92.

190. Ding L, Linsley PS, Huang LY, Germain RN, Shevach EM. IL-10 inhibits

macrophage costimulatory activity by selectively inhibiting the up-regulation of B7

expression. J Immunol. 1993;151(3):1224-34.

191. de Waal Malefyt R, Yssel H, de Vries JE. Direct effects of IL-10 on subsets of

human CD4+ T cell clones and resting T cells. Specific inhibition of IL-2 production and

proliferation. J Immunol. 1993;150(11):4754-65.

192. Schandene L, Alonso-Vega C, Willems F, Gerard C, Delvaux A, Velu T, et al.

B7/CD28-dependent IL-5 production by human resting T cells is inhibited by IL-10. J

Immunol. 1994;152(9):4368-74.

193. Joyce DA, Gibbons DP, Green P, Steer JH, Feldmann M, Brennan FM. Two

inhibitors of pro-inflammatory cytokine release, interleukin-10 and interleukin-4, have contrasting effects on release of soluble p75 tumor necrosis factor receptor by cultured monocytes. Eur J Immunol. 1994;24(11):2699-705.

69

194. Jenkins JK, Malyak M, Arend WP. The effects of interleukin-10 on interleukin-1 receptor antagonist and interleukin-1 beta production in human monocytes and neutrophils. Lymphokine Cytokine Res. 1994;13(1):47-54.

195. Cai G, Kastelein RA, Hunter CA. IL-10 enhances NK cell proliferation, cytotoxicity

and production of IFN-gamma when combined with IL-18. Eur J Immunol.

1999;29(9):2658-65.

196. Malisan F, Briere F, Bridon JM, Harindranath N, Mills FC, Max EE, et al.

Interleukin-10 induces immunoglobulin G isotype switch recombination in human CD40-

activated naive B lymphocytes. J Exp Med. 1996;183(3):937-47.

197. Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993;75(2):263-74.

198. Groux H, O'Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, et al. A

CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis.

Nature. 1997;389(6652):737-42.

199. Jankovic D, Kullberg MC, Feng CG, Goldszmid RS, Collazo CM, Wilson M, et al.

Conventional T-bet(+)Foxp3(-) Th1 cells are the major source of host-protective

regulatory IL-10 during intracellular protozoan infection. J Exp Med. 2007;204(2):273-

83.

200. Gagliani N, Amezcua Vesely MC, Iseppon A, Brockmann L, Xu H, Palm NW, et

al. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation.

Nature. 2015;523(7559):221-5.

70

201. Anderson CF, Oukka M, Kuchroo VJ, Sacks D. CD4(+)CD25(-)Foxp3(-) Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. J Exp Med. 2007;204(2):285-97.

202. Rhodes KA, Andrew EM, Newton DJ, Tramonti D, Carding SR. A subset of IL-10- producing gammadelta T cells protect the liver from Listeria-elicited, CD8(+) T cell- mediated injury. Eur J Immunol. 2008;38(8):2274-83.

203. Ejrnaes M, Filippi CM, Martinic MM, Ling EM, Togher LM, Crotty S, et al.

Resolution of a chronic viral infection after interleukin-10 receptor blockade. J Exp Med.

2006;203(11):2461-72.

204. Roque S, Nobrega C, Appelberg R, Correia-Neves M. IL-10 underlies distinct susceptibility of BALB/c and C57BL/6 mice to Mycobacterium avium infection and influences efficacy of antibiotic therapy. J Immunol. 2007;178(12):8028-35.

205. Belkaid Y, Hoffmann KF, Mendez S, Kamhawi S, Udey MC, Wynn TA, et al. The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure. J Exp Med.

2001;194(10):1497-506.

206. Omer FM, de Souza JB, Riley EM. Differential induction of TGF-beta regulates proinflammatory cytokine production and determines the outcome of lethal and nonlethal Plasmodium yoelii infections. J Immunol. 2003;171(10):5430-6.

207. Giunta B, Fernandez F, Nikolic WV, Obregon D, Rrapo E, Town T, et al.

Inflammaging as a prodrome to Alzheimer's disease. J Neuroinflammation. 2008;5:51.

208. Fulop T, Larbi A, Witkowski JM, McElhaney J, Loeb M, Mitnitski A, et al. Aging, frailty and age-related diseases. Biogerontology. 2010;11(5):547-63.

71

209. Lio D, Scola L, Crivello A, Colonna-Romano G, Candore G, Bonafe M, et al.

Gender-specific association between -1082 IL-10 promoter polymorphism and longevity.

Genes Immun. 2002;3(1):30-3.

210. Lio D, Candore G, Crivello A, Scola L, Colonna-Romano G, Cavallone L, et al.

Opposite effects of interleukin 10 common gene polymorphisms in cardiovascular diseases and in successful ageing: genetic background of male centenarians is protective against coronary heart disease. J Med Genet. 2004;41(10):790-4.

211. Lustig A, Liu HB, Metter EJ, An Y, Swaby MA, Elango P, et al. Telomere

Shortening, Inflammatory Cytokines, and Anti-Cytomegalovirus Antibody Follow Distinct

Age-Associated Trajectories in Humans. Frontiers in immunology. 2017;8:1027.

212. Dobber R, Tielemans M, Nagelkerken L. The in vivo effects of neutralizing antibodies against IFN-gamma, IL-4, or IL-10 on the humoral immune response in young and aged mice. Cellular immunology. 1995;160(2):185-92.

213. McKinstry KK, Strutt TM, Buck A, Curtis JD, Dibble JP, Huston G, et al. IL-10 deficiency unleashes an influenza-specific Th17 response and enhances survival against high-dose challenge. J Immunol. 2009;182(12):7353-63.

214. McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, et al. T cell responses are better correlates of vaccine protection in the elderly. J Immunol.

2006;176(10):6333-9.

215. Skowronski DM, Hottes TS, McElhaney JE, Janjua NZ, Sabaiduc S, Chan T, et al. Immuno-epidemiologic correlates of pandemic H1N1 surveillance observations: higher antibody and lower cell-mediated immune responses with advanced age. The

Journal of infectious diseases. 2011;203(2):158-67.

72

216. Gatto D, Brink R. The germinal center reaction. The Journal of allergy and clinical

immunology. 2010;126(5):898-907; quiz 8-9.

217. Crotty S. Follicular helper CD4 T cells (TFH). Annual review of immunology.

2011;29:621-63.

218. Crotty S. T follicular helper cell differentiation, function, and roles in disease.

Immunity. 2014;41(4):529-42.

219. Yu D, Rao S, Tsai LM, Lee SK, He Y, Sutcliffe EL, et al. The transcriptional

repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity.

2009;31(3):457-68.

220. Haynes NM, Allen CD, Lesley R, Ansel KM, Killeen N, Cyster JG. Role of CXCR5

and CCR7 in follicular Th cell positioning and appearance of a programmed cell death

gene-1high germinal center-associated subpopulation. J Immunol. 2007;179(8):5099-

108.

221. Ding BB, Bi E, Chen H, Yu JJ, Ye BH. IL-21 and CD40L synergistically promote plasma cell differentiation through upregulation of Blimp-1 in human B cells. J Immunol.

2013;190(4):1827-36.

222. Gigoux M, Shang J, Pak Y, Xu M, Choe J, Mak TW, et al. Inducible costimulator promotes helper T-cell differentiation through phosphoinositide 3-kinase. Proc Natl Acad

Sci U S A. 2009;106(48):20371-6.

223. Cannons JL, Tangye SG, Schwartzberg PL. SLAM family receptors and SAP adaptors in immunity. Annual review of immunology. 2011;29:665-705.

73

224. Lefebvre JS, Maue AC, Eaton SM, Lanthier PA, Tighe M, Haynes L. The aged

microenvironment contributes to the age-related functional defects of CD4 T cells in

mice. Aging cell. 2012;11(5):732-40.

225. Lefebvre JS, Masters AR, Hopkins JW, Haynes L. Age-related impairment of

humoral response to influenza is associated with changes in antigen specific T follicular

helper cell responses. Sci Rep. 2016;6:25051.

226. Zhou M, Zou R, Gan H, Liang Z, Li F, Lin T, et al. The effect of aging on the

frequency, phenotype and cytokine production of human blood CD4 + CXCR5 + T

follicular helper cells: comparison of aged and young subjects. Immun Ageing.

2014;11:12.

227. Agrawal A, Su H, Chen J, Osann K, Agrawal S, Gupta S. Increased IL-21

secretion by aged CD4+T cells is associated with prolonged STAT-4 activation and

CMV seropositivity. Aging (Albany NY). 2012;4(9):648-59.

228. Hirano T, Yasukawa K, Harada H, Taga T, Watanabe Y, Matsuda T, et al.

Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin. Nature. 1986;324(6092):73-6.

229. Yamasaki K, Taga T, Hirata Y, Yawata H, Kawanishi Y, Seed B, et al. Cloning and expression of the human interleukin-6 (BSF-2/IFN beta 2) receptor. Science.

1988;241(4867):825-8.

230. Hibi M, Murakami M, Saito M, Hirano T, Taga T, Kishimoto T. Molecular cloning and expression of an IL-6 signal transducer, gp130. Cell. 1990;63(6):1149-57.

74

231. Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L. Interleukin-6-

type cytokine signalling through the gp130/Jak/STAT pathway. Biochem J. 1998;334 (

Pt 2):297-314.

232. Naka T, Narazaki M, Hirata M, Matsumoto T, Minamoto S, Aono A, et al.

Structure and function of a new STAT-induced STAT inhibitor. Nature.

1997;387(6636):924-9.

233. Schmitz J, Weissenbach M, Haan S, Heinrich PC, Schaper F. SOCS3 exerts its inhibitory function on interleukin-6 signal transduction through the SHP2 recruitment site of gp130. J Biol Chem. 2000;275(17):12848-56.

234. Mullberg J, Oberthur W, Lottspeich F, Mehl E, Dittrich E, Graeve L, et al. The soluble human IL-6 receptor. Mutational characterization of the proteolytic cleavage site.

J Immunol. 1994;152(10):4958-68.

235. Jostock T, Mullberg J, Ozbek S, Atreya R, Blinn G, Voltz N, et al. Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur J

Biochem. 2001;268(1):160-7.

236. Gillmore JD, Lovat LB, Persey MR, Pepys MB, Hawkins PN. Amyloid load and

clinical outcome in AA amyloidosis in relation to circulating concentration of serum

amyloid A protein. Lancet. 2001;358(9275):24-9.

237. Heinrich PC, Castell JV, Andus T. Interleukin-6 and the acute phase response.

Biochem J. 1990;265(3):621-36.

238. Ghoreschi K, Laurence A, Yang XP, Tato CM, McGeachy MJ, Konkel JE, et al.

Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature.

2010;467(7318):967-71.

75

239. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory

T cells. Nature. 2006;441(7090):235-8.

240. Neveu WA, Allard JB, Dienz O, Wargo MJ, Ciliberto G, Whittaker LA, et al. IL-6 is required for airway mucus production induced by inhaled fungal allergens. J Immunol.

2009;183(3):1732-8.

241. Yang Y, Ochando J, Yopp A, Bromberg JS, Ding Y. IL-6 plays a unique role in initiating c-Maf expression during early stage of CD4 T cell activation. J Immunol.

2005;174(5):2720-9.

242. Diehl S, Chow CW, Weiss L, Palmetshofer A, Twardzik T, Rounds L, et al.

Induction of NFATc2 expression by interleukin 6 promotes T helper type 2 differentiation. J Exp Med. 2002;196(1):39-49.

243. Diehl S, Anguita J, Hoffmeyer A, Zapton T, Ihle JN, Fikrig E, et al. Inhibition of

Th1 differentiation by IL-6 is mediated by SOCS1. Immunity. 2000;13(6):805-15.

244. Choi YS, Eto D, Yang JA, Lao C, Crotty S. Cutting edge: STAT1 is required for

IL-6-mediated Bcl6 induction for early follicular helper cell differentiation. J Immunol.

2013;190(7):3049-53.

245. Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or

17 cell lineages. Immunity. 2008;29(1):138-49.

246. Eto D, Lao C, DiToro D, Barnett B, Escobar TC, Kageyama R, et al. IL-21 and IL-

6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS One. 2011;6(3):e17739.

76

247. Jin JO, Han X, Yu Q. Interleukin-6 induces the generation of IL-10-producing Tr1

cells and suppresses autoimmune tissue inflammation. J Autoimmun. 2013;40:28-44.

248. Giuliani N, Sansoni P, Girasole G, Vescovini R, Passeri G, Passeri M, et al.

Serum interleukin-6, soluble interleukin-6 receptor and soluble gp130 exhibit different patterns of age- and menopause-related changes. Experimental gerontology.

2001;36(3):547-57.

249. Ferrucci L, Corsi A, Lauretani F, Bandinelli S, Bartali B, Taub DD, et al. The origins of age-related proinflammatory state. Blood. 2005;105(6):2294-9.

250. Wei J, Xu H, Davies JL, Hemmings GP. Increase of plasma IL-6 concentration with age in healthy subjects. Life sciences. 1992;51(25):1953-6.

251. Michaud M, Balardy L, Moulis G, Gaudin C, Peyrot C, Vellas B, et al.

Proinflammatory cytokines, aging, and age-related diseases. J Am Med Dir Assoc.

2013;14(12):877-82.

252. Cohen HJ, Pieper CF, Harris T, Rao KM, Currie MS. The association of plasma

IL-6 levels with functional disability in community-dwelling elderly. The journals of gerontology Series A, Biological sciences and medical sciences. 1997;52(4):M201-8.

253. Ershler WB, Keller ET. Age-associated increased interleukin-6 gene expression, late-life diseases, and frailty. Annu Rev Med. 2000;51:245-70.

254. Maggio M, Guralnik JM, Longo DL, Ferrucci L. Interleukin-6 in aging and chronic disease: a magnificent pathway. The journals of gerontology Series A, Biological sciences and medical sciences. 2006;61(6):575-84.

77

255. Leng S, Chaves P, Koenig K, Walston J. Serum interleukin-6 and hemoglobin as

physiological correlates in the geriatric syndrome of frailty: a pilot study. J Am Geriatr

Soc. 2002;50(7):1268-71.

256. Walston J, Fedarko N, Yang H, Leng S, Beamer B, Espinoza S, et al. The

physical and biological characterization of a frail mouse model. The journals of

gerontology Series A, Biological sciences and medical sciences. 2008;63(4):391-8.

257. Parrish-Novak J, Dillon SR, Nelson A, Hammond A, Sprecher C, Gross JA, et al.

Interleukin 21 and its receptor are involved in NK cell expansion and regulation of

lymphocyte function. Nature. 2000;408(6808):57-63.

258. Ozaki K, Kikly K, Michalovich D, Young PR, Leonard WJ. Cloning of a type I

cytokine receptor most related to the IL-2 receptor beta chain. Proc Natl Acad Sci U S

A. 2000;97(21):11439-44.

259. Zeng R, Spolski R, Casas E, Zhu W, Levy DE, Leonard WJ. The molecular basis

of IL-21-mediated proliferation. Blood. 2007;109(10):4135-42.

260. Jin H, Carrio R, Yu A, Malek TR. Distinct activation signals determine whether IL-

21 induces B cell costimulation, growth arrest, or Bim-dependent apoptosis. J Immunol.

2004;173(1):657-65.

261. Diehl SA, Schmidlin H, Nagasawa M, van Haren SD, Kwakkenbos MJ, Yasuda

E, et al. STAT3-mediated up-regulation of BLIMP1 Is coordinated with BCL6 down- regulation to control human plasma cell differentiation. J Immunol. 2008;180(7):4805-

15.

262. Ozaki K, Spolski R, Feng CG, Qi CF, Cheng J, Sher A, et al. A critical role for IL-

21 in regulating immunoglobulin production. Science. 2002;298(5598):1630-4.

78

263. Salzer E, Kansu A, Sic H, Majek P, Ikinciogullari A, Dogu FE, et al. Early-onset

inflammatory bowel disease and common variable immunodeficiency-like disease caused by IL-21 deficiency. The Journal of allergy and clinical immunology.

2014;133(6):1651-9 e12.

264. Kotlarz D, Zietara N, Uzel G, Weidemann T, Braun CJ, Diestelhorst J, et al. Loss- of-function mutations in the IL-21 receptor gene cause a primary immunodeficiency syndrome. J Exp Med. 2013;210(3):433-43.

265. Skak K, Frederiksen KS, Lundsgaard D. Interleukin-21 activates human natural killer cells and modulates their surface receptor expression. Immunology.

2008;123(4):575-83.

266. Zhou L, Ivanov, II, Spolski R, Min R, Shenderov K, Egawa T, et al. IL-6 programs

T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007;8(9):967-74.

267. Nurieva RI, Chung Y, Martinez GJ, Yang XO, Tanaka S, Matskevitch TD, et al.

Bcl6 mediates the development of T follicular helper cells. Science.

2009;325(5943):1001-5.

268. Vogelzang A, McGuire HM, Yu D, Sprent J, Mackay CR, King C. A fundamental role for interleukin-21 in the generation of T follicular helper cells. Immunity.

2008;29(1):127-37.

269. Andris F, Denanglaire S, Anciaux M, Hercor M, Hussein H, Leo O. The

Transcription Factor c-Maf Promotes the Differentiation of Follicular Helper T Cells.

Frontiers in immunology. 2017;8:480.

79

270. Vasanthakumar A, Kallies A. IL-27 paves different roads to Tr1. Eur J Immunol.

2013;43(4):882-5.

271. Yu M, Li G, Lee WW, Yuan M, Cui D, Weyand CM, et al. Signal inhibition by the dual-specific phosphatase 4 impairs T cell-dependent B-cell responses with age. Proc

Natl Acad Sci U S A. 2012;109(15):E879-88.

80

Chapter 2

T-reg Homeostasis and Functions in Ageing

Treg and Aging

Maha Almanan, Claire Chougnet, and David A. Hildeman

Abstract Aging has a profound impact on immune responses. Both innate and adaptive compartments of the immune system undergo fundamental changes with age. It is well recognized that intrinsic defects, imprinted on T cells, contribute signifi- cantly to age-related diseases. However, recent data show a profound accumula- tion of regulatory T cells in aged mice and humans. The accumulation of these regulatory T cells, and their production of immune suppressive cytokines, likely contributes to immune dysregulation in aging. On the other hand, a great deal of work has shown substantially enhanced inflammation with age, with IL-6 being one of the hallmark inflammatory cytokines associated with this age-driven inflammation. Importantly, recent data coming from our labs and others suggest a novel paradigm, e.g., that the “inflammaging” environment also contributes to the accumulation of regulatory T cells with age. Indeed, while it is well known that the lack of IL-10 enhances inflammation, our recent data show that the lack of a key inflammatory cytokine, IL-6, actually reduces the accumulation of regula- tory T cells. Thus, we think that during aging there is an interrelated, mixed inflammatory/anti-inflammatory environment that is attempting to maintain homeostasis. Recent findings regarding the homeostasis of regulatory T cells and their role in controlling immune responses with age will be highlighted in this chapter.

# Springer International Publishing AG 2018

81

Claire Chougnet and David A. Hildeman contributed equally to this work.

M. Almanan • C. Chougnet (*) • D.A. Hildeman (*) Department of Pediatrics and Division of Immunobiology, MLC 7038, University of Cincinnati College of Medicine and Children’s Hospital Medical Center, Cincinnati, OH, USA Division of Infectious Diseases, Children’s Hospital Medical Center, Cincinnati, OH, USA e-mail: [email protected]; [email protected]; [email protected]

# Springer International Publishing AG 2018 T. Fulop et al. (eds.), Handbook of Immunosenescence, https://doi.org/10.1007/978-3-319-64597-1_82-1

82

T-reg Homeostasis and Functions in Ageing

Keywords CD25 • CD8+ T cells • Cytotoxic T-lymphocyte-associated antigen4 (CTLA-4) • Foxp3+ Treg cells • Myeloid-derived suppressor cells (MDSC) • Peripherally derived terg (pTreg) • Thymic-derived treg (tTreg) • Tr1 cells • Transforming growth factor-β (TGF-β) • Treg homeostasis

Contents Introduction ...... 2 Foxp3+ Treg Characterization ...... 3 Foxp3+ Treg Function ...... 5 Foxp3+ Treg Homeostasis with Age ...... 5 Environmental-Mediated Regulation of Treg Homeostasis with Age ...... 6 Inflammation Regulates Tissue-Specific Treg Accrual with Age ...... 8 Treg Function with Age ...... 8 CD8+ Treg Homeostasis and Function with Age ...... 11 Tr1 Homeostasis and Function with Age ...... 12 Functional Cross Talk between Regulators with Age ...... 14 Conclusion ...... 14 References ...... 16

Introduction

The world’s population over age 60 is predicted to increase to 9.7 billion by 2050 (UN). With age, the immune system is subjected to deleterious changes that shape its phenotype and functionality (Lopez-Otin et al. 2013; Goronzy et al. 2007; Arnold et al. 2011; Lefebvre and Haynes 2012; Linton and Dorshkind 2004; Eaton et al. 2004; Miller 1996, 2000; Miller et al. 1997; Weng 2006; Nikolich-Zugich 2014; Garcia and Miller 2002). As a result of this decreased functionality, higher incidence of infections, cancer, and poor vaccination contribute significantly to increased morbidity and mortality reported among the elderly (Lord 2013; Foster et al. 2011; Cevenini et al. 2013; Niccoli and Partridge 2012). A substantial amount of work has already characterized age-driven deleterious changes in adaptive immune cells (B, T) (Linton and Dorshkind 2004). Among these changes are (i) thymic involution and lack of naïve T cell production (Aspinall and Andrew 2000; Hale et al. 2006), (ii) intrinsic defects in T and B cell receptor-signaling (Miller 2000; Scholz et al. 2013; Tamir et al. 2000), (iii) terminal differentiation and acquisition of a replicative senescence phenotype (Effros et al. 2005), and finally, (iv) the expansion of sup- pressive cells in aged hosts (Nishioka et al. 2006; Sharma et al. 2006; Lages et al. 2008; Agius et al. 2009). Over the past 20 years, there has been a rebirth of “suppressor cell” immunology. Initially described in the 1970s, the phenomenon of “suppressive” T cells made a splash and then eventually went by the wayside as the cells were difficult to clone and characterize (Gershon and Kondo 1970; Sakaguchi et al. 2007). Further, a putative MHC cell surface marker that some suppressor cells reportedly expressed, I–J, was later found not to be encoded by an actual gene (Kronenberg et al. 1983).

83

T-reg Homeostasis and Functions in Ageing

It was not until Shimon Sakaguchi described a small population of CD25-expressing T cells must be present in mice in the neonatal period to prevent fatal autoimmunity, that “suppressive” T cells were reborn as “regulatory” T cells (Sakaguchi et al. 1995). Now equipped with modern genetic tools, these cells have been well defined and are characterized by the expression of Foxp3, a master transcription factor that is essential and sufficient for their intrathymic development and acquisition of function (Fontenot et al. 2003; Hori et al. 2003; Wildin et al. 2001; Brunkow et al. 2001; Yagi et al. 2004; Sakaguchi et al. 2010). Extensive work has gone into understanding how they develop, how they are maintained, and the mechanisms they utilize to suppress immune responses (Stephens et al. 2001; Jonuleit et al. 2001; Dieckmann et al. 2001; Baecher-Allan et al. 2001; Miyara and Sakaguchi 2007; Sakaguchi et al. 2009; Schmidt et al. 2012). Moreover, in addition to these Foxp3+ regulatory T cells, many other flavors of “regulatory” T cells have been shown to exist. Notably, CD8+ regulatory T cells (Lu and Cantor 2008), IL-10-producing Foxp3 negative regulatory T cell type 1 (Tr1) (Roncarolo et al. 2006), transforming growth factor-β (TGF-β)- secreting T-helper cell type 3 (Th3) (Wan and Flavell 2007), γδ T cells (Caccamo et al. 2013), and iNKT cells (Ikehara et al. 2000) were shown to temper adaptive and innate immune responses. Thus, the adaptive compartment not only promotes protective immunity but also employs cellular mechanisms to temper those responses. While most of the regulatory mechanisms utilized by these “regulatory” T cell subsets are still controversial, their major role is to maintain tolerance to self- antigens to prevent autoimmune diseases in some situations (Sakaguchi et al. 1995; Sakaguchi 2000) and to guard against exaggerated inflammatory responses to parasites and viruses in other situations (Belkaid 2007). New research also suggests some specialization in their role. There is growing evidence supporting the existence of nonlymphoid tissue specific Foxp3+ Treg cells, with distinct functionality and phenotype that regulate specific tissue resident immune cells (Burzyn et al. 2013; Cipolletta 2014). Importantly, regulatory T cells seem to play differential roles in modulating innate immune responses, for example, IL-10+ non-Treg cells appear more efficient at controlling inflammasome activation than regular Foxp3+ cells (Yao et al. 2015). Importantly, most of the characterization of regulatory subsets with age has focused on Foxp3 + CD4+ Treg and CD8+ Treg in addition to some data that suggest quantitative changes in IL-10 producing CD4+ T cells with age. However, we note that this does not imply that age-driven changes in other regulatory subsets does not occur, it just has not been reported yet to the best of our knowledge.

Foxp3+ Treg Characterization

Initially, most of the characterization of Treg in humans and mice lacked consen- sus on the reliable markers for their identification. For example, cytotoxic T- lymphocyte-associated antigen4 (CTLA-4), glucocorticoid-induced tumor necrosis factor receptor (GITR), Helios, programmed cell death-1 (PD-1), and ICOS (induc- ible costimulator) are all expressed on Treg, but also on activated T cells (Baecher- Allan et al. 2001; Dhuban and Piccirillo 2014). Further, although Foxp3 is a marker

84

T-reg Homeostasis and Functions in Ageing for Treg in mice and humans, one caveat is that, in humans, it is induced with T cell activation (Tran et al. 2007; Pillai et al. 2007; Walker et al. 2003). Also, as Foxp3 is an intracellular molecule and therefore intractable to functional analyses after intra- cellular staining, the lack of expression of CD127 (IL-7Rα) and the high expression of CD25 (IL-2Rα) have been used as an isolation strategy for human Treg allowing greater than 95% purity in most cases (Shevach 2004; Liu et al. 2006; Seddiki et al. 2006). However, this strategy leaves out the CD25lo CD127lo Foxp3+ cells that may be enriched in memory Treg (Raynor et al. 2013). In mice, the development of several Foxp3 reporter strains has greatly aided in their isolation and purification for adoptive transfer studies and biochemical analysis (Wan and Flavell 2005; Fontenot et al. 2005a). However, it is important to point out that, one of these reporter mice that generates Foxp3-GFP fusion protein appears to have altered Treg function, so studies using these mice have to be interpreted with caution (Bettini et al. 2012). Nonetheless, the ability to have distinct markers has greatly facilitated our under- standing of the development, homeostasis, and function of these cells. Classification of Treg cells has been an area of active research for years. Based on their development and site of generation, Treg were initially categorized into two main subpopulations of thymic-derived (tTreg) and peripherally derived (pTreg) Treg (Feuerer et al. 2009; Huehn et al. 2009). Both populations express CD4, CD25, and Foxp3. While tTreg constitutively express Foxp3, Foxp3 expression in pTreg is acquired in vivo in CD4 + Foxp3- Tcells in response to a variety of stimuli. However, both tTreg and pTreg have been shown to exert comparable suppressive abilities and in some experimental models they also synergized to obtain optimal regulation (Haribhai et al. 2009). Early insight into the mechanisms involved in the induction of pTreg came from in vitro studies. Both TGF-β1 and IL-2 were required for in vitro induction of Foxp3 expression in naïve CD4 + Foxp3- T cells, leading to a population of in vitro-induced Treg (iTreg) (Davidson et al. 2007; Chen et al. 2003). Following these initial studies, different murine models have confirmed the in vivo generation of pTreg in different tolerogenic settings. For example, pTreg develop following intra- venous administration of antigen in the absence of optimal costimulatory signals (Thorstenson and Khoruts 2001), or oral administration of antigen (Mucida et al. 2005). In the gut, CD103(+) mesenteric lymph node DCs induce pTreg in a TGF-β and retinoic acid (RA)-dependent manner (Coombes et al. 2007). More recently, additional phenotypic and functional analysis identified Treg as heterogeneous cells with distinct chemokine receptor expression that governs their migration to different tissues resulting in regulation of different inflammatory conditions. The transcriptional machinery regulating these diverse Treg phenotypes is believed to involve multiple ancillary transcription factors that act in conjunction with Foxp3 (Liston and Gray 2014; Campbell and Koch 2011). Importantly, work by Campbell et al. provided significant insight into the molecular mechanisms involved in Treg homeostatic localization. The study identified fundamental sub- divisions in Foxp3+ Treg cells, notably, lymphoid homing central Treg (CD44loCD62LhiCCR7+CD25hi) maintained by IL-2 and circulating tissue-homing effector Treg (CD44hiCD62LloCCR7-CD25lo) maintained by continuous signaling through the costimulatory receptor ICOS (Smigiel et al. 2014). Interestingly, similar phenotypic analysis in humans also identified two distinct Treg subpopulations:

85

T-reg Homeostasis and Functions in Ageing

CD45RA+Foxp3lo resting Treg cells (rTreg cells) and CD45RA-Foxp3hi activated Treg cells (aTreg cells) (Miyara et al. 2009). Taken together, work from humans and mice supports the existence of significant heterogeneity within Foxp3+ Treg.

Foxp3+ Treg Function

Treg are known to utilize multiple mechanisms for suppression, some of those mechanisms require contact and others do not require contact with conventional T cells or other cells such as dendritic cells (Sakaguchi et al. 2009). For example, as mentioned above, Treg express surface molecules like cytotoxic T-lymphocyte associated protein-4 (CTLA-4) that can modulate DC maturation or function by inhibiting the interaction of CD80/86 on DCs with CD28 on conventional T cells (Oderup et al. 2006). Treg also express other inhibitory molecules, such as lymphocyte-activation gene 3 (LAG3) (Huang et al. 2004), Programmed cell death protein 1 (PD-1), T cell immunoglobulin mucin 3 (TIM-3) (Gupta et al. 2012), which can all directly interfere with T cell activation. Additionally, Treg can also exert cytotoxic ability via production of granzyme A in humans and granzyme B in mice (Velaga et al. 2015; Cao et al. 2007). Importantly, while many of the factors that Treg utilize to exert their suppressive function have been identified, it is likely that some remain undiscovered, particularly under inflammatory contexts in different tissues. Additionally, Treg can indirectly inhibit effector cells via the induction of regu- latory biochemical pathways via the expression of ectoenzymes CD39 and CD73. High expression of both ectoenzymes on Treg controls the conversion of ADP/ATP to AMP and AMP to adenosine. Notably, extracellular adenosine is known to inhibit T cell proliferation, activation, and cytokine production (Deaglio et al. 2007; Bono et al. 2015). Moreover, Treg can induce the upregulation of IDO in DCs which facilitates the differentiation of naïve CD4(+) T cells toward a Foxp3+ (inducible Treg) (Fallarino et al. 2003) and can disrupt T cell metabolic functions by IL-2 deprivation and induction of apoptosis (Pandiyan et al. 2007). Treg can also produce immunoregulatory cytokines such as TGF-β, IL-10, and IL-35, which can all provide contact independent suppressive effects (Nakamura et al. 2001; Asseman et al. 1999; Collison et al. 2007). Thus, Treg deploy different suppressive weapons from their arsenal, depending upon the context and environment.

Foxp3+ Treg Homeostasis with Age

In aged mice, Treg are increased in both frequency and number in the lymphoid tissues (spleen, lymph nodes), but not in the blood (Sharma et al. 2006; Lages et al. 2008; Raynor et al. 2013; Zhao et al. 2007; Chougnet et al. 2011; Williams-Bey et al. 2011; Chiu et al. 2007; Han et al. 2009). Importantly, age-driven Treg accrual was observed in multiple strains of mice, making it a generalizable phenomenon. Further, the accumulation of Treg is progressive with middle-aged mice displaying interme- diate levels of Treg relative to young or aged mice (Raynor et al. 2013; Chougnet et al. 2011). In humans, an increased frequency of Treg within circulating mononuclear

86

T-reg Homeostasis and Functions in Ageing cells was observed in most studies, but these increases were modest (Lages et al. 2008; Rosenkranz et al. 2007; Gottenberg et al. 2005; Gregg et al. 2005). However, one study showed a significant expansion in the proportion of Foxp3+ Treg cells in the skin of old subjects compared with young ones (Agius et al. 2009), suggesting that Treg proportion is also increased with age in humans, but these cells are largely sequestered in the tissues. While Treg accumulate with age, the mechanisms underlying their accrual have just begun to be investigated. Given the massive thymic involution with age, it is unlikely that thymic output contributes significantly to Treg accrual (Chougnet et al. 2011; Aw and Palmer 2011). Another possibility is that aging enhances the conver- sion of conventional T cells into Treg. However, several pieces of data argue against this possibility. First, compared to young mice, conventional T cells from aged mice have less of an ability to undergo peripheral conversion (Chougnet et al. 2011). Second, the vast majority of Treg from aged mice express both Helios and Neuropilin, two markers of nTreg cells (Raynor et al. 2015). Third, using RNAseq analysis, we showed that Treg from aged mice are most closely related to young Treg, rather than to aged or young naïve or memory T cells (Raynor et al. 2015). As Treg accrual did not appear to be due to enhanced production or conversion, the other possibilities were proliferation or survival. We showed that, if anything, Treg from aged mice proliferated less, not more, than their young counterparts (Chougnet et al. 2011; Raynor et al. 2015). Instead, we found that both in vitro and in vivo, aged Treg survived longer than young Treg (Raynor et al. 2013, 2015; Chougnet et al. 2011). Such enhanced survival was due to their progressive decrease in expression of the key proapoptotic molecule Bim (Raynor et al. 2013; Chougnet et al. 2011). Further, mice deficient in Bim (even Treg-specific loss of Bim) accumulated periph- eral Treg rapidly, the peripheral Treg levels in a 4–6-month-old Bim-/- mouse resembled a 1.5-year-old wildtype mouse (Raynor et al. 2013; Chougnet et al. 2011). Thus, aging drives a loss of Bim expression in Treg, which likely contributes to their preferential survival and accumulation.

Environmental-Mediated Regulation of Treg Homeostasis with Age

In young mice, CD25 signaling plays an important role in promoting the develop- ment and peripheral survival of Treg (Bayer et al. 2007; D’Cruz and Klein 2005; Fontenot et al. 2005b; Burchill et al. 2007a, b; Mahmud et al. 2013). However, while Treg are reduced in young CD25-deficient mice, they are not completely absent. One potential reason is that Treg can use other common gamma chain cytokines (e.g., IL-7 and IL-15), when CD25 is missing (Burchill et al. 2007b; Soper et al. 2007; Vang et al. 2008), although, in the normal setting IL-2 seems to be the primary driver of Treg development and peripheral survival. The aging environment is associated with an altered cytokine milieu (O’Mahony et al. 1998; Bruunsgaard et al. 2003; Ershler and Keller 2000) and we recently showed that circulating IL-2 levels are significantly decreased with age (Raynor et al. 2013). Consistent with this, we found

87

T-reg Homeostasis and Functions in Ageing that roughly half of the Treg that accumulate with age express low levels of CD25 (Raynor et al. 2013). These data suggested that, in contrast to Treg in young mice, Treg in aged mice are likely less reliant on IL-2, and may rely more on other cytokines. Indeed, we found that Treg from aged mice have higher levels of CD122 (IL-2Rß, the receptor for IL-15) (Raynor et al. 2013). Neutralization of IL- 15, but not IL-2, led to a significant reduction of Treg in aged mice and the additional loss of Bim in IL-15-/- mice rescued the loss of Treg in middle-aged IL-15-/- mice (Raynor et al. 2013). Further, in-depth characterization of old Treg showed significant selective accumulation of Bimlo CD25lo (Raynor et al. 2013). Of note, although aged Treg express increased levels of CD127, IL-7 neutralization studies had no effect on Treg accrual (Chougnet et al. 2011). Thus, it appears that IL-15 favors age-driven Treg accrual perhaps as a compen- satory mechanism for the loss of IL-2 with age. However, since Treg are not completely absent in aged IL-15 deficient mice, it was possible that other cytokines or molecules can also promote Treg accrual with age. Interestingly, similar to earlier characterization of Treg by Campbell et al. (Smigiel et al. 2014), we showed that the vast majority of accumulated Treg in aged mice were of an “effector” phenotype (CD44hi CD62LloICOShiBlimp-1hi) and these were partially maintained by ICOS (Raynor et al. 2015). In contrast, there were less “central” Treg (CD44lo CD62Lhi), which fits the overall model as central Treg cells are largely maintained by IL-2 (Smigiel et al. 2014). Thus, IL-15 and ICOS contribute to Treg accumulation with age. As IL-6 is significantly increased with age (Mysliwska et al. 1998), and IL-6 transgenic mice have elevated numbers of tTreg (Fujimoto et al. 2011), we hypoth- esized that IL-6 increased IL-6 with age might have a modulatory effect on Treg homeostasis with age. Interestingly, with age there was a significant loss of total Treg and more precisely effector Treg in IL-6 deficient mice, likely through the regulation of ICOS expression which in turn likely contributes to the expression of Bim (Raynor et al. 2015). The role of IL-6-ICOS-Bim axis on Treg accrual with age was a novel finding considering the inflammatory role of IL-6. It suggests that the increase in effector Treg population with age acts as a counterbalance to augmented inflammatory IL-6 production. How this IL-6-ICOS-Bim axis signals to control Treg homeostasis with age remains unclear. ICOS signals through the activation of the PI3K/Akt pathway which is known to modulate FOXO3a transcription factor acti- vation, which has been shown to alter Bim expression (Gigoux et al. 2009; Tang et al. 1999; Brunet et al. 1999; Salih and Brunet 2008; Stahl et al. 2002). However, our preliminary data suggest that PI3K/Akt pathway is not solely responsible for regulating Bim expression (unpublished data). Another mechanism that could con- tribute to altered Bim expression includes epigenetic modification to the Bcl2L11 gene (the gene encoding Bim) (San Jose-Eneriz et al. 2009; Paschos et al. 2009). This mechanism is intriguing because it would be a stable and heritable way to ensure lowered Bim expression in a population of cells that periodically divides. Future studies are needed to better elucidate the mechanisms involved in the control of Treg/effector Treg survival with age.

88

T-reg Homeostasis and Functions in Ageing

Inflammation Regulates Tissue-Specific Treg Accrual with Age

In addition to the role played by IL-6, ICOS, and IL-15 in promoting the accumu- lation of Treg with age in the lymphoid tissues, other factors may regulate (both positively and negatively) Treg accumulation in the tissues. Interestingly, a recent study showed that induced inflammation during muscle injury can lead to a Treg deficit with age via an IL-33-ST2 dependent mechanism (Kuswanto et al. 2016). Treg have high expression of ST2 (IL-33R) which controls their specific migration to injured muscle as loss of ST2 on Treg in young mice compromised their accrual in injured muscles but not in the spleen. In aged mice, Treg had a muscle specific gene signature that was unique compared to other lymphoid tissues and hence regulated their migration to/retention in injured muscles. In-depth investigation of the role of IL-33-ST2 axis on accrual of muscle Treg with age showed that reduced IL-33 production upon muscle injury led to diminished accumulation of Treg in injured muscle and resulted in poor repair of skeletal injury of old mice compared to young mice. Importantly, IL-33 supplementation resulted in enhanced Treg accumulation in injured muscle and tissue regeneration suggesting that muscle repair during aging is impaired due to reduced Treg muscle retention. In contrast, prior work showed that Treg accumulate in the skin of aged humans which may be triggered by local inflammatory stimuli or other factors (Agius et al. 2009). Further, the accumulation of Treg correlated with decreased TNF-α secretion by cutaneous macrophages in aged individuals. The implication is that Treg accumulation in human skin with age might contribute to increased risk of cutaneous malignancy and infections. None- theless, the accumulation or lack of accumulation of Treg in nonlymphoid tissues suggests their tissue-specific roles in age-driven immune dysregulation. Combined, these intriguing data support further investigation of mechanisms regulating Treg tissue homeostasis and their role in driving immune-suppression with age.

Treg Function with Age

The increased numbers of Treg with age implies that they have a role in age-driven immune suppression in mice and humans. Recent work has begun to dissect the functionality of aged Treg at the population and individual cell level, and although there are a few exceptions, overall the data suggest increased Treg function with age at both levels. Further, the data suggest a role for Treg in controlling susceptibility to autoimmunity, infections, and tumors in aged mice and humans. For example, recent work showed that Treg from patients with Alzheimer’s as well as Parkinson’s disease had enhanced suppressive activity on a per cell basis, compared to age matched controls (Rosenkranz et al. 2007). In another study, an association was found between an increased frequency of Foxp3-expressing CD4+CD25+CD127lo Treg cells in elderly patients with metastatic primary nonsmall cell lung cancer, suggesting a potential suppressive effect of Treg population on antitumor immunity (Pan et al. 2012). In mice, Treg depletion using α-CD25 antibodies restored the antitumor immune response and led to a significant reduction in tumor burden,

89

T-reg Homeostasis and Functions in Ageing suggesting that aged Treg reduced antitumor immunity (Sharma et al. 2006). Sim- ilarly, we showed that same depletion strategy in aged mice reduced the lesion size in a Leishmania major infection model (Lages et al. 2008). Additionally, aged Treg inhibited antigen-specific IFN- γ, but not IL-10 production from effector T cells (Lages et al. 2008). Notably, this latter study was the first to show the similar if not enhanced suppressive activity of old Treg relative to young mice by using the Foxp3-GFP knock-in mice. This is important because, with age, many Treg are CD25lo (Raynor et al. 2013) and therefore, responses measured based on CD25+ Treg cells are likely to be an underestimate of Treg function. In another infection model, a significant expansion of Treg was reported in the spleen of old compared to young mice (Williams-Bey et al. 2011). Although the study implicated Treg expan- sion in diminished CD8 T cell responses to influenza infection with age, unfortu- nately the authors did not test the role of Treg in modulating the anti-influenza responses. Given the findings that supported enhanced functionality of Treg with age, a few studies have investigated the molecular mechanism underlying this enhanced func- tionality. For example, CD40 and CD86 expression on myeloid DCs was enhanced upon depletion of CD25+ T cells in aged mice, suggesting Treg reduce DC function with age (Chiu et al. 2007). Another study showed high suppressive function of Treg from old mice that correlated with an increase in Foxp3 expression driven by the hypomethylation of the upstream Foxp3 enhancer with age (Garg et al. 2014). The authors demonstrated that aged Treg limited the availability of extracellular cysteine which led to imbalanced redox potential that was unfavorable for T cell activation and proliferation. Additionally, Treg modulated the CD86 expression on APCs in an IL-10-dependent manner thus further limiting effector T cell proliferation. Other studies show an equivalent suppression of T cell proliferation in response to Phytohaemagglutinin (PHA) by Treg from young and old individuals (Gregg et al. 2005) as well as a similar inhibitory effect on effector cytokine production (e.g., IFN-γ, TNF-α, IL-6 and IL-2) (Hwang et al. 2009). Similarly, studies in mice have shown that CD4 + CD25+ sorted Treg cells had a similar suppressive effect on the proliferation (Zhao et al. 2007) and IFN- γ production by effector T cells (Sun et al. 2012) compared to their young counterparts. Nonetheless, even if certain models show similar Treg function in aged and young animals, the elevated numbers of Treg would suggest an enhanced overall function. In contrast, in a few other studies, Treg function seemed to decline with age. In some studies, examination of Treg functionality in humans reported a decline in the ability of aged Treg (relative to young) to suppress the proliferation of CD4 + CD25- T cells stimulated by a combination of anti-CD3 and anti-CD28 mAb, independent of the gender of the aged individual (Tsaknaridis et al. 2003). Old Treg cells in mice also had diminished suppression of IL-2 and IFN- γ production in vitro as well as a delayed hypersensitivity reaction (DTH) in vivo (Zhao et al. 2007). Additionally, Treg from old mice were defective in suppressing IL-17 production from T cells compared to young Treg in a model of colitis, probably due to a reduction in STAT3

90

T-reg Homeostasis and Functions in Ageing activation (Sun et al. 2012). This suggests that the inability of aged Treg to restrain Th17 cells might contribute to the higher incidence of IL-17-driven, age-related autoimmunity. Collectively, the data strongly suggest increased Treg function with age, however, we do acknowledge, in some instances, decreased Treg function is observed. We think there are a few explanations for these differences. For example, in vitro suppression assays have been used to assess Treg function, there are some major caveats regarding their use and interpretation. First, they may not represent the in vivo mechanisms by which Treg control effector T cell responses. For example, soluble factors like IL-10 or TGF-ß are dispensable for Treg in vitro but not in vivo suppression (Sakaguchi et al. 2009; Asseman et al. 1999; Thornton and Shevach 1998; Suri-Payer and Cantor 2001; Piccirillo et al. 2002). Second, accumulation of CD8 + CD25 + Foxp3+ Treg has also been reported with age, suggesting that the results obtained through the use of α-CD25 antibodies in some of the previous studies might not be CD4 + CD25+ Treg-specific (Sharma et al. 2006). Third, the use of young or aged responder cells might contribute to the observed differential results. Thus, the responsiveness of CD4 + CD25- target cells might differ based on the type and strength of stimuli used, independent of the regulatory properties of the Treg or their number in the assay (Kozlowska et al. 2007). Fourth, aged Treg might differ in function with respect to the type of in vivo models used (e.g., infectious disease, autoimmune, or tumor). Each model adds another layer of complexity because the local inflammatory environment would influence not only cellular effector responses, but also the phenotype, function, and homing of Treg (Wei et al. 2006). For example, the expression of CD62L and CD103 both change with age and can affect migration of Treg cells into secondary lymphoid organs and tissues (Lages et al. 2008; Zhao et al. 2007; Braun et al. 2015; Anz et al. 2011; Suffia et al. 2005). Additionally, CD4 + Foxp3 + CD25lo Treg from aged naïve mice were functional but less suppressive than CD4 + Foxp3 + CD25hi Treg in vitro. However, the two Treg subsets were equally suppressive in aged tumor-bearing mice, suggesting that the functionality of a specific subset of Treg can be altered by the local inflammatory environment (Hurez et al. 2012). More importantly, in humans, functional studies require sampling CD25+ Treg cells from the blood, and as mentioned, CD25 as a surface marker is not sufficient to isolate all Treg, particularly in the aging population as many of these cells are CD25lo and the function of CD4 + CD25+ T cells do not necessarily correlate with CD4 + Foxp3+ Treg cells. Also, Treg from blood are poorly suppressive as they express lower levels of CTLA- 4. Similar tissue specific expression of CTLA-4 was also reported in mice (Lages et al. 2008). Thus, in addition to the potential lack of relevance of the functional assay to their in vivo function, sampling Treg activity in human blood might not be representative of Treg that accumulate in the tissues. Thus, more work is required to determine the heterogeneity within the Treg compartment and how this changes with age so that similar populations can be compared between young and aged mice. Nevertheless, these prior studies have begun to pave the road toward better under- standing of Treg functionality with age.

91

T-reg Homeostasis and Functions in Ageing

CD8+ Treg Homeostasis and Function with Age

CD8+ T cells with immunosuppressive functions were first identified by Gershon and Kondo in the 1970s (Gershon and Kondo 1970, 1971). Regulatory CD8+ T cells are a functionally and phenotypically distinct subset from the classical cytotoxic CD8+ T cells, and play an essential role in fine-tuning immune responses to prevent autoimmunity and inflammation (Konya et al. 2009). A fair amount of work aimed at investigating the development, characterization, and functions of CD8+ Treg using different models. Interestingly, similar to CD4 + Foxp3+ Treg, CD8+ Treg that express Foxp3 have been shown to be produced naturally in the thymus or induced in the periphery through antigen-dependent or antigen-independent mechanisms (Kapp and Bucy 2008; Tang et al. 2005; Churlaud et al. 2015). They are character- ized by the expression of several markers such as CD8aa, CD25, CD38, CD45RA, CD45RO, CD56, Foxp3, CXCR3, LAG-3, CD103, and CD122, as well as by the lack of expression of CD28 and CD127 (Tang et al. 2005; Pomie et al. 2008; Dinesh et al. 2010). Clearly, CD8+ Treg are a heterogeneous population composed of a variety of subsets, each with a distinct phenotype. CD8+ Treg can directly inhibit immune responses via cell contact-dependent or -independent mechanisms. For example, surface expression of CTLA-4 and/or production of soluble regulatory cytokines such as IL-10, TGF-ß can all induce a tolerogenic state in APCs and modulate effector T cell responses (Vukmanovic-Stejic et al. 2001). Notably, much of what is now known about regulatory T cells in aging has been learned from studies investigating CD4 + Foxp3+ Treg, with limited information available on CD8+ Treg cells. With age, qualitative and quantitative changes in CD8+ Treg have been reported in both humans and murine models. While some studies have shown that both the percentage and number of CD8+ Treg were significantly increased in the blood of older individuals and in aged mice (Sharma et al. 2006; Simone et al. 2008), others have shown reduced frequencies of CD8+ CCR7+ Treg induced under low-dose of anti-CD3 and IL-15 (Suzuki et al. 2012) or similar frequencies of circulating CD8 + Foxp3 + CCR7+ Treg in older individuals compared to their young coun- terparts (Wen et al. 2016). However, there remains considerable uncertainty in increased CD8+ Foxp + Treg with age and it needs to be mentioned that with age CD8+ Foxp3+ Treg cells remain of extremely low frequency (<2% of all splenic CD8+ T cells, unpublished data). Likewise, there has also been disagreement as to the functionality of these cells with age; some human studies reported equivalent regulatory function with age, where both old and young CD8+ Treg showed similar abilities in suppressing proliferation and cytokine production from responding cells (Simone et al. 2008). However, others have shown that CD8 + CCR7+ Treg derived from aged individuals had reduced expression of Foxp3+ suggesting reduced sup- pressive abilities (Suzuki et al. 2012) (Nakagawa et al. 2010; Singh et al. 2007). The reduced functionality of CD8+ Treg, however, may not be due wholly to reduced Foxp3 expression; other molecular defects have also been identified in relation to age-related failure of CD8 + Treg functionality. For example, CD8+ Treg from older individuals with giant cell arteritis (GCA) a disease characterized

92

T-reg Homeostasis and Functions in Ageing by inflammatory vasculopathy, suffered from inefficient induction of NADPH oxi- dase and shedding of NOX2-containing exosomes. NOX2-containing exosomes are known to regulate the activity of effector CD4+ T cells by abrogating the phosphor- ylation of the upstream signaling molecules ZAP70 and linker of activated T cells (LAT) (Wen et al. 2016). The study suggested that aged patients with autoimmune diseases might have less functional CD8+ Treg, thus highlighting the possible therapeutic applications in manipulating CD8+ Treg with age. Notably, variations in reporting CD8+ Treg percentages or functions with age were clearly related to the lack of consensus on specific markers for CD8+ Treg identification which vary among reports. To date, there is considerable uncertainty about the molecular mechanisms regulating the homeostasis and function of CD8+ Treg with age. Surprisingly, IL-6 positively regulates the Foxp3 + CD8+ Treg development and function, suggesting that the effects of IL-6 in the development of regulatory T cells are not likely to be selective for CD4+ Foxp3+ T cells over CD8+ Foxp3+ T cells. In F759 mice, which carry a Y759F mutation in the gp130 signal transducer of the IL-6 receptor complex, the Foxp3+ CD8+ Treg number was significantly increased with age via enhanced IL-6 signaling (Nakagawa et al. 2010). However, this study only looked at Foxp3+ CD8+ Treg up to 6 months of age. The role of IL-6 and IL-6- dependent molecular mechanisms in modulating the Foxp3+ CD8+ Treg population with age thus requires further investigation. Collectively, whether the aged inflam- matory environment plays a role in Foxp3 + CD8+ Treg accrual and whether conversion, excessive proliferation, or survival mechanisms play a role in main- taining aged CD8+ Treg similar to their CD4 + Foxp3+ counterparts remains largely unknown.

Tr1 Homeostasis and Function with Age

CD4 + Foxp3- type 1 regulatory T (Tr1) cells play a major role in peripheral (Roncarolo and Battaglia 2007). Tr1 cells were first described by Roncarolo et al. in 1988 in the blood of a severe combined immunodeficiency (SCID) patient, who developed long-term tolerance after HLA-mismatched fetal liver hematopoietic stem cell transplant (HSCT) (Roncarolo et al. 1988). Tr1 cells are characterized by their ability to produce high levels of IL-10 and to mediate immune suppression in humans and mice despite their lack of Foxp3 expression (Roncarolo et al. 2006). Several cytokines are involved in the generation of Tr1 cells. For example, IL-10, IL-6, IL-21, IL-27, and type-1 interferons (Vasanthakumar and Kallies 2013; Ziegler- Heitbrock et al. 2003; Stumhofer et al. 2007; Levings et al. 2001; Jin et al. 2013; Pot et al. 2009). Additionally, Tr1 cells can also secrete variable amounts of TGF-ß, IFN-γ, and IL-5 (Battaglia et al. 2006; Groux et al. 1997). Although Tr1 cells lack expression of a specific lineage tracing marker, the cell-surface markers CD49b, LAG-3, and CD226 were recently identified as reliable markers for the characteriza- tion of Tr1 cells in humans and mice (Gagliani et al. 2013). Several different mechanisms have been identified in Tr1 immune-suppression (Gregori et al. 2012). Tr1 cells can modulate T cells and APC function via IL-10 and

93

T-reg Homeostasis and Functions in Ageing

TGF-β secretion (Fiorentino et al. 1991; de Waal Malefyt et al. 1991; Cavani et al. 2000). Additionally, the production of granzyme B (GZB), perforin, and the inter- action of CD226 on Tr1 cells with CD155 on APCs can lead to the death of myeloid APCs (Magnani et al. 2011). More importantly, CTLA-4 signaling represents a major mechanism of Tr1 suppression (Meiler et al. 2008; Akdis et al. 2004). Tr1 cells have been shown to play significant roles in the control of autoimmune diseases and organ transplantation. Thus, there are ongoing clinical trials using human Tr1 cells in various immune-related disorders like hematologic malignancies and Crohn’s disease (Roncarolo and Battaglia 2007; Zeng et al. 2015). Despite the well-established role of Tr1 cells in immune tolerance, little information is known about their homeostasis and function in aging. Increased IL-10 production associated with aging has been reported in both aged mice and humans (Corsini et al. 2006) (Hobbs et al. 1994). However, few studies have determined the cellular source of this increased IL-10. In mice, the capacity of CD4+ T cells to produce IL-10 increased significantly with age, both at the level of gene expression as well as cytokine release upon TCR stimulation. Further analysis showed that CD4 + CD44+ were the predominant source of IL-10 in old mice (Hobbs et al. 1994). Additionally, one study provided evidence for an increase in CD3lo CD5hi (high avidity) aged T cells that expressed higher levels of IL-10 at both the mRNA and protein level (Tatari-Calderone et al. 2012). However, it was unclear whether the increase in IL-10 producing cells reflects a specific increase in CD4+ Foxp3- IL-10+ (Tr1) cells with age. The functional consequences of increased IL-10 in aging is also largely unknown, with suggestions that it could be either protective or harmful, depending on the circumstances. For example, in vitro stimulation of whole blood from aged individuals vaccinated for influenza resulted in a significant increase in IL-10 production which correlated with reduced responses to vaccination compared with their young counterparts (Corsini et al. 2006). In contrast, HIV- specific IL-10-producing CD4 + Foxp3- T cells were significantly more frequent in peripheral blood of older AIDS patients treated by antiretroviral therapy (HAART) than in their younger counterparts. Furthermore, in vitro neutralization of IL-10 correlated with enhanced viral replication and higher IL-1β and IL-6 production. The results suggested that in aged HIV-infected individuals, IL-10-producing CD4 + Foxp3- T cells help better control virus replication by diminishing the release of pro-inflammatory cytokines (Andrade et al. 2012). Also interesting was the fact that an IL-10 polymorphism associated with IL-10 high production was frequent in male centenarians and was suggested to be associated with longevity (Lio et al. 2002). Interestingly, a recent study provided evidence for a role of the inflammatory aged environment in the regulation of IL-10-producing CD4+ T cells. This study suggested that the tolerogenic environment permissive of tumors seen in aged mice is not due to T cell cell-intrinsic differences; rather, it is due to the aged environ- ment’s conditioning of these cells, as tumor specific CD4+ T cells from young mice transferred into old mice had defective Th1 differentiation, acquired a Tr1 like phenotype and were unable to inhibit tumor growth (Tsukamoto et al. 2015). The data raises the question about the effect of IL-6 or other aspects of the aged

94

T-reg Homeostasis and Functions in Ageing environment on the induction of Tr1 cells with age. Thus, future studies are required to investigate the homeostasis and function of Tr1 cells with age.

Functional Cross Talk between Regulators with Age

A realistic picture of both Treg homeostasis and functionality with age is based on the presence of crosstalk between different regulatory subsets as well as effector T cells. For example, one study showed that Treg as well as myeloid-derived suppres- sor cells (MDSC) are both increased in with age (Hurez et al. 2012). Depletion of Foxp3 + Treg using diphtheria toxin (DT) further increased the numbers of MDSC in aged melanoma tumor-bearing mice and abrogated the antitumor effects of Treg depletion observed in young mice. These results in contrast with enhanced tumor control in aged mice upon Treg depletion in a BM-185-EGFP tumor model com- pared to young mice (Sharma et al. 2006). Thus, with age Treg might have a differential regulatory effect on effector T cells and other regulatory subsets. Like- wise, we observed a profound increase in CD4 + Foxp3-IL-10-producing regulatory T (Tr1) cells upon Treg depletion in aged mice (manuscript in preparation), and Treg depletion in aged mice latently infected with MCMV resulted in viral reactivation, likely through enhanced accumulation of these CD4 + Foxp3-IL-10-producing regulatory T cells and (Almanan et al. 2017). Our data is consistent with previously work that suggested enhanced capacity of aged CD4 + CD25+ Treg in regulating IL-10 production from responding cells compared to their young counterparts (Hwang et al. 2009). This suggests that, with age, Foxp3+ Treg might play not only their classical role in regulating effector cells but also in regulating other regulatory populations to ensure protective immunity and limit immunopathology. Thus, the ways in which this interconnected network of regulatory and effector T cells evolves with age could present both advantages and disadvantages, depending on its context.

Conclusion

Several important questions remain unanswered: what are the molecular mecha- nisms of aged Treg-mediated suppression? Can specific Treg cells be targeted therapeutically in order to boost antitumor immunity or to control chronic infections to improve the quality of life of the elderly population? The role of IL-6 in Treg homeostasis with age is a novel finding. Although IL-6 plays a central role in the inflammaging process leading to frailty, morbidity, and mortality with age (Maggio et al. 2006), IL-6 also counterbalances excessive inflammation by promoting several Treg subsets accrual with age. These data collectively suggest a reevaluation of the “inflammaging” hypothesis to include its natural anti-inflammatory counterbalance. In all, the aging environment reflects a disturbed equilibrium that is attempting to maintain homeostasis (Fig. 1). It is in this mixed inflammatory/anti-inflammatory environment that immune responses are

95

T-reg Homeostasis and Functions in Ageing

Fig. 1 Age-related mixed pro- and anti-inflammatory environment. Regulatory T cells that are Foxp3+, CD8+ Foxp3+, and Foxp3- IL-10+ (Tr1) accumulate dramatically in aged hosts along with a profound increase in the levels of IL-6 and IL-10. With age IL-6 plays not only its classical inflammatory role but also promotes regulatory T cells accrual and increased production of IL-10, perhaps as a mechanism to counterbalance the dysregulated inflammatory environment and main- tain homeostasis. While Foxp3+ Treg cells have been shown to produce higher levels of IL-10 with age, enhanced production of IL-10 from Tr1 cells with age still unclear. The homeostasis of the aged mixed inflammatory/anti-inflammatory environment is also related to the cross talk between different regulatory and effector T cells. While the accumulation Foxp3+ T cells and perhaps Tr1 cells with age clearly would have a negative impact on naïve/effector T cells or on APCs to inhibit T cell responses, Foxp3+ Treg accrual might also restrain undesirable consequences of Tr1 accumu- lation and accompanied increase in IL-10 production to maintain protective immunity. However, the cross talk between the regulators with age remains unclear instigated and propagated. Thus, a better understanding of mechanisms underlying tissue-specific Treg homeostasis and function will hopefully lead to strategies tailored to manipulate local Treg accumulation and function to selectively enhance (e.g., antitumor immunity, infections) or inhibit (e.g., autoimmunity) effector T cell responses. However, our data also suggest that those manipulations may have unforeseen consequences, due to the intricate network of regulatory cells that act synergistically or antagonistically, depending on the tissue context.

Acknowledgments We thank Kate Carroll for critical reading of the manuscript and the funding from NIH -RO1AG033057 to C.A.C. and D.A.H

96

T-reg Homeostasis and Functions in Ageing

References

Agius E et al (2009) Decreased TNF-alpha synthesis by macrophages restricts cutaneous immunosurveillance by memory CD4+ T cells during aging. J Exp Med 206(9):1929–1940 Akdis M et al (2004) Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells. J Exp Med 199(11): 1567–1575 Almanan et al. (2017) PLOS pathogens (in press). Andrade RM et al (2012) High IL-10 production by aged AIDS patients is related to high frequency of Tr-1 phenotype and low in vitro viral replication. Clin Immunol 145(1):31–43 Anz D et al (2011) CD103 is a hallmark of tumor-infiltrating regulatory T cells. Int J Cancer 129(10):2417–2426 Arnold CR et al (2011) Gain and loss of T cell subsets in old age–age-related reshaping of the T cell repertoire. J Clin Immunol 31(2):137–146 Aspinall R, Andrew D (2000) Thymic involution in aging. J Clin Immunol 20(4):250–256 Asseman C et al (1999) An essential role for interleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation. J Exp Med 190(7):995–1004 Aw D, Palmer DB (2011) The origin and implication of thymic involution. Aging Dis 2(5):437–443 Baecher-Allan C et al (2001) CD4+CD25high regulatory cells in human peripheral blood. J Immunol 167(3):1245–1253 Battaglia M et al (2006) Tr1 cells: from discovery to their clinical application. Semin Immunol 18(2):120–127 Bayer AL, Yu A, Malek TR (2007) Function of the IL-2R for thymic and peripheral CD4+CD25+ Foxp3+ T regulatory cells. J Immunol 178(7):4062–4071 Belkaid Y (2007) Regulatory T cells and infection: a dangerous necessity. Nat Rev Immunol 7(11): 875–888 Bettini ML et al (2012) Loss of epigenetic modification driven by the Foxp3 transcription factor leads to regulatory T cell insufficiency. Immunity 36(5):717–730 Bono MR et al (2015) CD73 and CD39 ectonucleotidases in T cell differentiation: beyond immunosuppression. FEBS Lett 589(22):3454–3460 Braun A et al (2015) Integrin alphaE(CD103) is involved in regulatory T-cell function in allergic contact hypersensitivity. J Invest Dermatol 135(12):2982–2991 Brunet A et al (1999) Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96(6):857–868 Brunkow ME et al (2001) Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 27(1):68–73 Bruunsgaard H et al (2003) Elevated levels of tumor necrosis factor alpha and mortality in centenarians. Am J Med 115(4):278–283 Burchill MA et al (2007a) Interleukin-2 receptor signaling in regulatory T cell development and homeostasis. Immunol Lett 114(1):1–8 Burchill MA et al (2007b) IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J Immunol 178(1):280–290 Burzyn D, Benoist C, Mathis D (2013) Regulatory T cells in nonlymphoid tissues. Nat Immunol 14(10):1007–1013 Caccamo N et al (2013) Mechanisms underlying lineage commitment and plasticity of human gammadelta T cells. Cell Mol Immunol 10(1):30–34 Campbell DJ, Koch MA (2011) Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat Rev Immunol 11(2):119–130 Cao X et al (2007) Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity 27(4):635–646 Cavani A et al (2000) Human CD4+ T lymphocytes with remarkable regulatory functions on dendritic cells and nickel-specific Th1 immune responses. J Invest Dermatol 114(2):295–302

97

T-reg Homeostasis and Functions in Ageing

Cevenini E, Monti D, Franceschi C (2013) Inflamm-ageing. Curr Opin Clin Nutr Metab Care 16(1): 14–20 Chen W et al (2003) Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med 198(12):1875–1886 Chiu BC et al (2007) Increased Foxp3(+) Treg cell activity reduces dendritic cell co-stimulatory molecule expression in aged mice. Mech Ageing Dev 128(11–12):618–627 Chougnet CA et al (2011) A major role for Bim in regulatory T cell homeostasis. J Immunol 186(1):156–163 Churlaud G et al (2015) Human and mouse CD8(+)CD25(+)FOXP3(+) regulatory T cells at steady state and during interleukin-2 therapy. Front Immunol 6:171 Cipolletta D (2014) Adipose tissue-resident regulatory T cells: phenotypic specialization, functions and therapeutic potential. Immunology 142(4):517–525 Collison LW et al (2007) The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 450(7169):566–569 Coombes JL et al (2007) A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med 204(8):1757–1764 Corsini E et al (2006) High interleukin-10 production is associated with low antibody response to influenza vaccination in the elderly. J Leukoc Biol 80(2):376–382 Davidson TS et al (2007) Cutting edge: IL-2 is essential for TGF-beta-mediated induction of Foxp3 + T regulatory cells. J Immunol 178(7):4022–4026 D’Cruz LM, Klein L (2005) Development and function of agonist-induced CD25+Foxp3+ regula- tory T cells in the absence of interleukin 2 signaling. Nat Immunol 6(11):1152–1159 de Waal Malefyt R et al (1991) Interleukin 10 (IL-10) and viral IL-10 strongly reduce antigen- specific human T cell proliferation by diminishing the antigen-presenting capacity of monocytes via downregulation of class II major histocompatibility complex expression. J Exp Med 174(4): 915–924 Deaglio S et al (2007) Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 204(6):1257–1265 Dhuban KB, Piccirillo CA (2014) Markers for human FOXP3+ regulatory T cells: current status and implications for immune monitoring in human disease. Int Trends Immun 2(4):162–165 Dieckmann D et al (2001) Ex vivo isolation and characterization of CD4(+)CD25(+) T cells with regulatory properties from human blood. J Exp Med 193(11):1303–1310 Dinesh RK et al (2010) CD8+ Tregs in lupus, autoimmunity, and beyond. Autoimmun Rev 9(8): 560–568 Eaton SM et al (2004) Age-related defects in CD4 T cell cognate helper function lead to reductions in humoral responses. J Exp Med 200(12):1613–1622 Effros RB et al (2005) The role of CD8+ T-cell replicative senescence in human aging. Immunol Rev 205:147–157 Ershler WB, Keller ET (2000) Age-associated increased interleukin-6 gene expression, late-life diseases, and frailty. Annu Rev Med 51:245–270 Fallarino F et al (2003) Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol 4(12):1206–1212 Feuerer M et al (2009) Foxp3+ regulatory T cells: differentiation, specification, subphenotypes. Nat Immunol 10(7):689–695 Fiorentino DF et al (1991) IL-10 inhibits cytokine production by activated macrophages. J Immunol 147(11):3815–3822 Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4(4):330–336 Fontenot JD et al (2005a) Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity 22(3):329–341 Fontenot JD et al (2005b) A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol 6(11):1142–1151

98

T-reg Homeostasis and Functions in Ageing

Foster AD, Sivarapatna A, Gress RE (2011) The aging immune system and its relationship with cancer. Aging Health 7(5):707–718 Fujimoto M et al (2011) The influence of excessive IL-6 production in vivo on the development and function of Foxp3+ regulatory T cells. J Immunol 186(1):32–40 Gagliani N et al (2013) Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med 19(6):739–746 Garcia GG, Miller RA (2002) Age-dependent defects in TCR-triggered cytoskeletal rearrangement in CD4+ T cells. J Immunol 169(9):5021–5027 Garg SK et al (2014) Aging is associated with increased regulatory T-cell function. Aging Cell 13(3):441–448 Gershon RK, Kondo K (1970) Cell interactions in the induction of tolerance: the role of thymic lymphocytes. Immunology 18(5):723–737 Gershon RK, Kondo K (1971) Infectious immunological tolerance. Immunology 21(6):903–914 Gigoux M et al (2009) Inducible costimulator promotes helper T-cell differentiation through phosphoinositide 3-kinase. Proc Natl Acad Sci USA 106(48):20371–20376 Goronzy JJ, Lee WW, Weyand CM (2007) Aging and T-cell diversity. Exp Gerontol 42(5):400–406 Gottenberg JE et al (2005) CD4 CD25high regulatory T cells are not impaired in patients with primary Sjogren’s syndrome. J Autoimmun 24(3):235–242 Gregg R et al (2005) The number of human peripheral blood CD4+ CD25high regulatory T cells increases with age. Clin Exp Immunol 140(3):540–546 Gregori S, Goudy KS, Roncarolo MG (2012) The cellular and molecular mechanisms of immuno- suppression by human type 1 regulatory T cells. Front Immunol 3:30 Groux H et al (1997) A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 389(6652):737–742 Gupta S et al (2012) Allograft rejection is restrained by short-lived TIM-3+PD-1+Foxp3+ Tregs. J Clin Invest 122(7):2395–2404 Hale JS et al (2006) Thymic output in aged mice. Proc Natl Acad Sci USA 103(22):8447–8452 Han GM et al (2009) Age-associated parallel increase of Foxp3(+)CD4(+) regulatory and CD44(+) CD4(+) memory T cells in SJL/J mice. Cell Immunol 258(2):188–196 Haribhai D et al (2009) A central role for induced regulatory T cells in tolerance induction in experimental colitis. J Immunol 182(6):3461–3468 Hobbs MV, Weigle WO, Ernst DN (1994) Interleukin-10 production by splenic CD4+ cells and cell subsets from young and old mice. Cell Immunol 154(1):264–272 Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcrip- tion factor Foxp3. Science 299(5609):1057–1061 Huang CT et al (2004) Role of LAG-3 in regulatory T cells. Immunity 21(4):503–513 Huehn J, Polansky JK, Hamann A (2009) Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage? Nat Rev Immunol 9(2):83–89 Hurez V et al (2012) Mitigating age-related immune dysfunction heightens the efficacy of tumor immunotherapy in aged mice. Cancer Res 72(8):2089–2099 Hwang KA, Kim HR, Kang I (2009) Aging and human CD4(+) regulatory T cells. Mech Ageing Dev 130(8):509–517 Ikehara Y et al (2000) CD4(+) Valpha14 natural killer T cells are essential for acceptance of rat islet xenografts in mice. J Clin Invest 105(12):1761–1767 Jin JO, Han X, Yu Q (2013) Interleukin-6 induces the generation of IL-10-producing Tr1 cells and suppresses autoimmune tissue inflammation. J Autoimmun 40:28–44 Jonuleit H et al (2001) Identification and functional characterization of human CD4(+)CD25(+) T cells with regulatory properties isolated from peripheral blood. J Exp Med 193(11):1285–1294 Kapp JA, Bucy RP (2008) CD8+ suppressor T cells resurrected. Hum Immunol 69(11):715–720 Konya C, Goronzy JJ, Weyand CM (2009) Treating autoimmune disease by targeting CD8(+) T suppressor cells. Expert Opin Biol Ther 9(8):951–965 Kozlowska E et al (2007) Age-related changes in the occurrence and characteristics of thymic CD4(+) CD25(+) T cells in mice. Immunology 122(3):445–453

99

T-reg Homeostasis and Functions in Ageing

Kronenberg M et al (1983) RNA transcripts for I-J polypeptides are apparently not encoded between the I-A and I-E subregions of the murine major histocompatibility complex. Proc Natl Acad Sci USA 80(18):5704–5708 Kuswanto W et al (2016) Poor repair of skeletal muscle in aging mice reflects a defect in local, interleukin-33-dependent accumulation of regulatory T cells. Immunity 44(2):355–367 Lages CS et al (2008) Functional regulatory T cells accumulate in aged hosts and promote chronic infectious disease reactivation. J Immunol 181(3):1835–1848 Lefebvre JS, Haynes L (2012) Aging of the CD4 T cell compartment. Open Longev Sci 6:83–91 Levings MK et al (2001) IFN- and IL-10 induce the differentiation of human type 1 T regulatory cells. J Immunol 166(9):5530–5539 Linton PJ, Dorshkind K (2004) Age-related changes in lymphocyte development and function. Nat Immunol 5(2):133–139 Lio D et al (2002) Gender-specific association between -1082 IL-10 promoter polymorphism and longevity. Genes Immun 3(1):30–33 Liston A, Gray DH (2014) Homeostatic control of regulatory T cell diversity. Nat Rev Immunol 14(3):154–165 Liu W et al (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203(7):1701–1711 Lopez-Otin C et al (2013) The hallmarks of aging. Cell 153(6):1194–1217 Lord JM (2013) The effect of ageing of the immune system on vaccination responses. Hum Vaccin Immunother 9(6):1364–1367 Lu L, Cantor H (2008) Generation and regulation of CD8(+) regulatory T cells. Cell Mol Immunol 5(6):401–406 Maggio M et al (2006) Interleukin-6 in aging and chronic disease: a magnificent pathway. J Gerontol A Biol Sci Med Sci 61(6):575–584 Magnani CF et al (2011) Killing of myeloid APCs via HLA class I, CD2 and CD226 defines a novel mechanism of suppression by human Tr1 cells. Eur J Immunol 41(6):1652–1662 Mahmud SA, Manlove LS, Farrar MA (2013) Interleukin-2 and STAT5 in regulatory T cell development and function. JAKSTAT 2(1):e23154 Meiler F et al (2008) In vivo switch to IL-10-secreting T regulatory cells in high dose allergen exposure. J Exp Med 205(12):2887–2898 Miller RA (1996) The aging immune system: primer and prospectus. Science 273(5271):70–74 Miller RA (2000) Effect of aging on T lymphocyte activation. Vaccine 18(16):1654–1660 Miller RA et al (1997) Early activation defects in T lymphocytes from aged mice. Immunol Rev 160:79–90 Miyara M, Sakaguchi S (2007) Natural regulatory T cells: mechanisms of suppression. Trends Mol Med 13(3):108–116 Miyara M et al (2009) Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 30(6):899–911 Mucida D et al (2005) Oral tolerance in the absence of naturally occurring Tregs. J Clin Invest 115(7):1923–1933 Mysliwska J et al (1998) Increase of interleukin 6 and decrease of interleukin 2 production during the ageing process are influenced by the health status. Mech Ageing Dev 100(3):313–328 Nakagawa T et al (2010) IL-6 positively regulates Foxp3+CD8+ T cells in vivo. Int Immunol 22(2): 129–139 Nakamura K, Kitani A, Strober W (2001) Cell contact-dependent immunosuppression by CD4(+) CD25(+) regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med 194(5):629–644 Niccoli T, Partridge L (2012) Ageing as a risk factor for disease. Curr Biol 22(17):R741–R752 Nikolich-Zugich J (2014) Aging of the T cell compartment in mice and humans: from no naive expectations to foggy memories. J Immunol 193(6):2622–2629 Nishioka T et al (2006) CD4+CD25+Foxp3+ T cells and CD4+CD25-Foxp3+ T cells in aged mice. J Immunol 176(11):6586–6593

100

T-reg Homeostasis and Functions in Ageing

Oderup C et al (2006) Cytotoxic T lymphocyte antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in CD4+ CD25+ regulatory T-cell-mediated sup- pression. Immunology 118(2):240–249 O’Mahony L et al (1998) Quantitative intracellular cytokine measurement: age-related changes in proinflammatory cytokine production. Clin Exp Immunol 113(2):213–219 Pan XD et al (2012) Changes of regulatory T cells and FoxP3 gene expression in the aging process and its relationship with lung tumors in humans and mice. Chin Med J 125(11):2004–2011 Pandiyan P et al (2007) CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation- mediated apoptosis of effector CD4+ T cells. Nat Immunol 8(12):1353–1362 Paschos K et al (2009) Epstein-barr virus latency in B cells leads to epigenetic repression and CpG methylation of the tumour suppressor gene Bim. PLoS Pathog 5(6):e1000492 Piccirillo CA et al (2002) CD4(+)CD25(+) regulatory T cells can mediate suppressor function in the absence of transforming growth factor beta1 production and responsiveness. J Exp Med 196(2): 237–246 Pillai Vet al (2007) Transient regulatory T-cells: a state attained by all activated human T-cells. Clin Immunol 123(1):18–29 Pomie C, Menager-Marcq I, van Meerwijk JP (2008) Murine CD8+ regulatory T lymphocytes: the new era. Hum Immunol 69(11):708–714 Pot C et al (2009) Cutting edge: IL-27 induces the transcription factor c-Maf, cytokine IL-21, and the costimulatory receptor ICOS that coordinately act together to promote differentiation of IL-10-producing Tr1 cells. J Immunol 183(2):797–801 Raynor J et al (2013) IL-15 fosters age-driven regulatory T cell accrual in the face of declining IL-2 levels. Front Immunol 4:161 Raynor J et al (2015) IL-6 and ICOS antagonize bim and promote regulatory T cell accrual with age. J Immunol 195(3):944–952 Roncarolo MG, Battaglia M (2007) Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol 7(8):585–598 Roncarolo MG et al (1988) Autoreactive T cell clones specific for class I and class II HLA antigens isolated from a human chimera. J Exp Med 167(5):1523–1534 Roncarolo MG et al (2006) Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev 212:28–50 Rosenkranz D et al (2007) Higher frequency of regulatory T cells in the elderly and increased suppressive activity in neurodegeneration. J Neuroimmunol 188(1–2):117–127 Sakaguchi S (2000) Regulatory T cells: key controllers of immunologic self-tolerance. Cell 101(5): 455–458 Sakaguchi S et al (1995) Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155(3):1151–1164 Sakaguchi S, Wing K, Miyara M (2007) Regulatory T cells – a brief history and perspective. Eur J Immunol 37(Suppl 1):S116–S123 Sakaguchi S et al (2009) Regulatory T cells: how do they suppress immune responses? Int Immunol 21(10):1105–1111 Sakaguchi S et al (2010) FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol 10(7):490–500 Salih DA, Brunet A (2008) FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol 20(2):126–136 San Jose-Eneriz E et al (2009) Epigenetic down-regulation of BIM expression is associated with reduced optimal responses to imatinib treatment in chronic myeloid leukaemia. Eur J Cancer 45(10):1877–1889 Schmidt A, Oberle N, Krammer PH (2012) Molecular mechanisms of treg-mediated T cell suppression. Front Immunol 3:51 Scholz JL et al (2013) A comparative review of aging and B cell function in mice and humans. Curr Opin Immunol 25(4):504–510

101

T-reg Homeostasis and Functions in Ageing

Seddiki N et al (2006) Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med 203(7):1693–1700 Sharma S, Dominguez AL, Lustgarten J (2006) High accumulation of T regulatory cells prevents the activation of immune responses in aged animals. J Immunol 177(12):8348–8355 Shevach EM (2004) Regulatory/suppressor T cells in health and disease. Arthritis Rheum 50(9): 2721–2724 Simone R, Zicca A, Saverino D (2008) The frequency of regulatory CD3+CD8+CD28- CD25+ T lymphocytes in human peripheral blood increases with age. J Leukoc Biol 84(6):1454–1461 Singh RP et al (2007) CD8+ T cell-mediated suppression of autoimmunity in a murine lupus model of peptide-induced immune tolerance depends on Foxp3 expression. J Immunol 178(12): 7649–7657 Smigiel KS et al (2014) CCR7 provides localized access to IL-2 and defines homeostatically distinct regulatory T cell subsets. J Exp Med 211(1):121–136 Soper DM, Kasprowicz DJ, Ziegler SF (2007) IL-2Rbeta links IL-2R signaling with Foxp3 expression. Eur J Immunol 37(7):1817–1826 Stahl M et al (2002) The forkhead transcription factor FoxO regulates transcription of p27Kip1 and Bim in response to IL-2. J Immunol 168(10):5024–5031 Stephens LA et al (2001) Human CD4(+)CD25(+) thymocytes and peripheral T cells have immune suppressive activity in vitro. Eur J Immunol 31(4):1247–1254 Stumhofer JS et al (2007) Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10. Nat Immunol 8(12):1363–1371 Suffia I et al (2005) A role for CD103 in the retention of CD4+CD25+ Treg and control of Leishmania major infection. J Immunol 174(9):5444–5455 Sun L et al (2012) Aged regulatory T cells protect from autoimmune inflammation despite reduced STAT3 activation and decreased constraint of IL-17 producing T cells. Aging Cell 11(3): 509–519 Suri-Payer E, Cantor H (2001) Differential cytokine requirements for regulation of autoimmune gastritis and colitis by CD4(+)CD25(+) T cells. J Autoimmun 16(2):115–123 Suzuki M et al (2012) CD8+CD45RA+CCR7+FOXP3+ T cells with immunosuppressive proper- ties: a novel subset of inducible human regulatory T cells. J Immunol 189(5):2118–2130 Tamir A et al (2000) Age-dependent alterations in the assembly of signal transduction complexes at the site of T cell/APC interaction. J Immunol 165(3):1243–1251 Tang ED et al (1999) Negative regulation of the forkhead transcription factor FKHR by Akt. J Biol Chem 274(24):16741–16746 Tang XL, Smith TR, Kumar V (2005) Specific control of immunity by regulatory CD8 T cells. Cell Mol Immunol 2(1):11–19 Tatari-Calderone Z et al (2012) Age-related accumulation of T cells with markers of relatively stronger autoreactivity leads to functional erosion of T cells. BMC Immunol 13:8 Thornton AM, Shevach EM (1998) CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med 188(2):287–296 Thorstenson KM, Khoruts A (2001) Generation of anergic and potentially immunoregulatory CD25 +CD4 T cells in vivo after induction of with intravenous or oral antigen. J Immunol 167(1):188–195 Tran DQ, Ramsey H, Shevach EM (2007) Induction of FOXP3 expression in naive human CD4 +FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood 110(8):2983–2990 Tsaknaridis L et al (2003) Functional assay for human CD4+CD25+ Treg cells reveals an age- dependent loss of suppressive activity. J Neurosci Res 74(2):296–308 Tsukamoto H et al (2015) IL-6-mediated environmental conditioning of defective Th1 differenti- ation dampens antitumour immune responses in old age. Nat Commun 6:6702 UN. The World population prospects: 2015 Revision. Population Division of the Department of Economic and Social Affairs of the United Nations

102

T-reg Homeostasis and Functions in Ageing

Vang KB et al (2008) IL-2, -7, and -15, but not thymic stromal lymphopoeitin, redundantly govern CD4+Foxp3+ regulatory T cell development. J Immunol 181(5):3285–3290 Vasanthakumar A, Kallies A (2013) IL-27 paves different roads to Tr1. Eur J Immunol 43(4): 882–885 Velaga S et al (2015) Granzyme A is required for regulatory T-cell mediated prevention of gastrointestinal graft-versus-host disease. PLoS One 10(4):e0124927 Vukmanovic-Stejic M et al (2001) Specificity, restriction and effector mechanisms of immunoreg- ulatory CD8 T cells. Immunology 102(2):115–122 Walker MR et al (2003) Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25- T cells. J Clin Invest 112(9):1437–1443 Wan YY, Flavell RA (2005) Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter. Proc Natl Acad Sci U S A 102(14):5126–5131 Wan YY, Flavell RA (2007) ‘Yin-Yang’ functions of transforming growth factor-beta and T regulatory cells in immune regulation. Immunol Rev 220:199–213 Wei S, Kryczek I, Zou W (2006) Regulatory T-cell compartmentalization and trafficking. Blood 108(2):426–431 Wen Z et al (2016) NADPH oxidase deficiency underlies dysfunction of aged CD8+ Tregs. J Clin Invest 126(5):1953–1967 Weng NP (2006) Aging of the immune system: how much can the adaptive immune system adapt? Immunity 24(5):495–499 Wildin RS et al (2001) X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet 27(1):18–20 Williams-Bey Y, Jiang J, Murasko DM (2011) Expansion of regulatory T cells in aged mice following influenza infection. Mech Ageing Dev 132(4):163–170 Yagi H et al (2004) Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int Immunol 16(11):1643–1656 Yao Y et al (2015) Tr1 cells, but not Foxp3+ regulatory T cells, suppress NLRP3 inflammasome activation via an IL-10-dependent mechanism. J Immunol 195(2):488–497 Zeng H et al (2015) Type 1 regulatory T cells: a new mechanism of peripheral immune tolerance. Cell Mol Immunol 12(5):566–571 Zhao L et al (2007) Changes of CD4+CD25+Foxp3+ regulatory T cells in aged Balb/c mice. J Leukoc Biol 81(6):1386–1394 Ziegler-Heitbrock L et al (2003) IFN- induces the human IL-10 gene by recruiting both IFN regulatory factor 1 and Stat3. J Immunol 171(1):285–290

103

Chapter 3

TISSUE-SPECIFIC CONTROL OF LATENT CMV REACTIVATION BY REGULATORY T CELLS

Maha Almanan1, Jana Raynor1, Allyson Sholl1, Mei Wang2, Claire Chougnet1,

Rhonda D. Cardin2, 3¶*, and David A. Hildeman1¶*

Running title: Treg and latent MCMV infection

1Department of Pediatrics, University of Cincinnati College of Medicine, Division of

Immunobiology, Children’s Hospital Medical Center, Cincinnati, OH, United States of America.

2Department of Pediatrics, University of Cincinnati College of Medicine, Division of Infectious

Diseases, Children’s Hospital Medical Center, Cincinnati, OH, United States of America.

3Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State

University, Baton Rouge, LA, United States of America.

*Corresponding authorsEmail:[email protected](DH),[email protected] (RC)

¶These authors contributed equally to this work

Keywords: Foxp3+ Treg, IL-10, MCMV, virus, tissues

Published in PLOS Pathogens

August 10, 2017

104

Abstract:

Cytomegalovirus (CMV) causes a persistent, lifelong infection. CMV persists in a latent state

and undergoes intermittent subclinical viral reactivation that is quelled by ongoing T cell

responses. While T cells are critical to maintain control of infection, the immunological factors

that promote CMV persistence remain unclear. Here, we investigated the role of regulatory T

cells (Treg) in a mouse model of latent CMV infection using Foxp3-diptheria toxin receptor

(Foxp3-DTR) mice. Eight months after infection, MCMV had established latency in the spleen,

salivary gland, lung, and pancreas, which was accompanied by an increased frequency of Treg.

Administration of diphtheria toxin (DT) after establishment of latency efficiently depleted Treg

and drove a significant increase in the numbers of functional MCMV-specific CD4+ and CD8+

T cells. Strikingly, Treg depletion decreased the number of animals with reactivatable latent

MCMV in the spleen. Unexpectedly, in the same animals, ablation of Treg drove a significant increase in viral reactivation in the salivary gland that was accompanied with augmented local

IL-10 production by Foxp3-CD4+T cells. Further, neutralization of IL-10 after Treg depletion significantly decreased viral load in the salivary gland. Combined, these data show that Treg have divergent control of MCMV infection depending upon the tissue. In the spleen, Treg antagonize CD8+ effector function and promote viral persistence while in the salivary gland

Treg prevent IL-10 production and limit viral reactivation and replication. These data provide new insights into the organ-specific roles of Treg in controlling the reactivation of latent MCMV infection.

105

Author Summary:

Cytomegalovirus (CMV) infection in both mice and humans is normally initially contained by a vigorous adaptive immune response that drives the virus into latency in multiple tissues.

However, the immunologic mechanisms that control latency are not well understood. In this report, we have examined the role of regulatory T cells (Treg) in a mouse model of CMV infection. Interestingly, depletion of regulatory T cells had profound consequences on MCMV latent infection, depending upon the tissue. In the spleen, Treg depletion enhanced CD8+ T cell responses and reduced reactivatable latent infection from the spleen. In striking contrast, in the salivary gland, Treg depletion enhanced the production of IL-10 from CD4+ T cells as well as viral reactivation. Thus, Treg play divergent and tissue specific roles in controlling MCMV reactivation from latency.

106

Introduction:

The immune system has evolved multiple innate and adaptive strategies to control pathogens[1]. Likewise, in order to ensure their persistence, pathogens have developed sophisticated and elaborate mechanisms to avoid the host immune system establishing latent infections that are never cleared from the host [2-7]. Human cytomegalovirus (HCMV) and its murine homolog (MCMV) are well-studied examples of pathogens that have developed multiple means to establish latency [8-10]. MCMV is a reasonable model for HCMV as it shares multiple biological characteristics and significant homology to the genome of HCMV[11]. A large number of HCMV and MCMV genes are involved in modulating innate and adaptive host immune responses [12-15]. During primary infection, these viruses vigorously replicate and disseminate by infecting many cell types, including epithelial, endothelial, smooth muscle, and connective tissue cells, as well as specialized parenchymal cells in multiple tissues [16].

Primary CMV infection is well controlled by a robust early NK cell response followed by

CD4+ and CD8+ T cell responses that ultimately results in control of virus replication, although the virus is not eliminated and persists for the lifetime of the host [17-20]. Interestingly, prior work shows that the control of lytic virus in different tissues requires distinct immune cell populations [21-25]. In the spleen, epitope-specific CD8+ T cells are sufficient to control acute

MCMV infection [26]. Whereas in the salivary gland (SG), CD4+ T cells and, in particular their production of IFN-γ, are crucial for terminating lytic viral replication. Further, IL-10R blockade or the absence of CD4+ cell-derived IL-10 enhanced the accumulation of IFN-γ–producing

CD4+ T cells and inhibited MCMV persistence in the SG [21, 22, 25, 27-29]. This role of CD4+

T cells in the SG is critical, as there appears to be a reservoir of MCMV in non-hematopoietic cells within the SG that downregulate MHC class I and become resistant to CD8+ T cell killing

107

[22]. However, recent work showed that CD8+ T cells can play a role in controlling MCMV in the SG after local re-infection [30]. The likely explanation for this differential role of CD8+ T cells during re-infection is because of the higher expression of class I MHC in acutely infected cells in contrast to cells harboring latent virus. Regulatory T cells (Treg) have also been shown to contribute to immune-mediated control of acute MCMV infection. In vitro, Treg were shown to suppress the function of MCMV-specific CD8+T cells via secretion of TGF-ß [31]. In vivo, during acute MCMV, Treg depletion resulted in enhanced MCMV-specific T cell responses and decreased viral load [32]. Nonetheless, following the termination of lytic infection, the virus persists in a latent state in which viral genomes are present in the absence of replicating virus.

MCMV establishes latency in a number of tissues similar to HCMV, including the spleen, lungs, and bone marrow [33-36]. This latent form of HCMV drives a substantial amount of morbidity when it reactivates in immune suppressed individuals (e.g. aged individuals, persons with HIV infection, transplant patients) [37-39]. During latent infection, HCMV persists in CD34+ hematopoietic stem cells as well as more committed myeloid lineage progenitor cells, monocytes, and macrophages [40-44]. Both myeloid lineage cells [33, 45], as well as non- hematopoietic cells such as endothelial cells in many organs are considered cellular sites of latent infection for MCMV [33, 46, 47].While a fair amount is known regarding immune mechanisms controlling acute MCMV infection, significantly less is known about immune mechanisms contributing to the control of latent MCMV infection. It has been reported that once latency is established, the cooperative function of lymphocytes including NK, CD4+, CD8+ T cells and

CMV-specific antibodies prevent the production of lytic virus from latent pools in the spleen and lungs and SG [48]. In most visceral organs, CD8+ T cells play a crucial role in preventing the emergence of lytic viral replication. For example, CD8 T cells maintain latency by epitope-

108

specific sensing of transcriptional reactivation in the lungs and killing these cells [49]. Similar to

their role in controlling acute infection in the SG, CD4+ T cell production of IFN-γ is critical to

prevent lytic virus production from latently infected cells [22, 50, 51].

During latency, there is substantial epigenetic suppression of viral immediate-early genes

that must be overcome by cellular signals to exit from latency [42, 44, 52]. Two models have been proposed for MCMV reactivation during latency. First, a two-step model proposed by

Hummel et al. [53] in which an inflammatory immune response can drive the activation of the major immediate-early (MIE) gene, initiating reactivation from latency. The second step requires an immune-suppressive environment, such as that which occurs during γ-irradiation or immune- deficiency. This immune-suppressive environment allows the virus to actively replicate, facilitating the production of lytic virus. In one example of this two-step scenario, the production of inflammatory mediators like tumor necrosis factor alpha (TNF-α), interleukin-2

(IL-2), and gamma interferon (IFN-γ) following allogeneic transplantation of kidneys in mice initiated IE gene expression [54]. However, full lytic reactivation of MCMV was only found in immune-deficient recipients [55]. Similarly, in human transplant patients, HCMV reactivation correlated with increased inflammatory responses [56, 57]. A second model focusing on latency and reactivation in the lung suggests that virus is reactivating frequently in immunocompetent hosts as shown by detectable levels of the immediate-early ie1/ie3 transcripts but that checkpoints exist which prevent full production of infectious virus following transcriptional activation [58-60]. Increased TNF-α levels were not enough to fully reactivate virus, although five-fold higher levels of IE transcripts were detected. Presumably memory T cells, specific for the IE proteins, lyse the reactivating cells before virus is produced [9, 58-61]. Multiple immune cells such as CD4+ and CD8+ T cells and other factors such as antibody and IFN-γ have also

109 been shown to be important for maintaining latency [48, 62]. However, the immune cells and molecular factors that control viral latency and whether these cells and factors are the same between different tissues remains unclear.

In recent work, recurrent virus reactivation was accompanied by a concomitant increase in Treg frequency and suppressive functionality [63]. Thus, while Treg have been associated with viral reactivation, a causative role in control of latent CMV/MCMV has not been established. Here, we investigated the role of Treg in controlling latent MCMV infection.

Strikingly, we found that Treg had opposing effects in distinct tissues harboring latent virus. In the spleen, Treg promoted viral persistence and suppressed local MCMV-specific effector T cell responses, while in the SG Treg were required to prevent viral reactivation/replication and IL-10 production by Foxp3- CD4+ T cells. These data show that Treg play divergent and tissue- specific roles in controlling viral reactivation from latency depending upon the type of T cells

(effector or regulatory) they inhibit.

Results:

Activated Treg are increased in the spleen during latent MCMV.

Foxp3+ regulatory T cells (Treg) can suppress effector T cells and promote MCMV replication in the spleen and salivary gland (SG) during acute MCMV infection [32]. However, there are no studies examining the role of Treg during latent MCMV infection. To investigate the role of regulatory T cells (Treg) during latent MCMV infection, we infected cohorts of C57BL/6 and

Foxp3-DTR mice with MCMV and waited for 8 months. Notably, in the C57BL/6 background, low level virus replication continues in the SG and spleen for months longer than is observed in mice on a BALB/c background. Thus, it took 7 to 8 months following infection for the

110 establishment of latency, as measured by the inability to detect replicating virus in multiple tissues, including spleen, lung, liver, pancreas, and SG (Supplementary Table.1). Next, we examined the levels and activation status of Treg in the spleen of latently infected mice.

Interestingly, compared to uninfected mice of the same age, the frequency of Treg (Fig.1A, B), and in particular “effector” Treg (as assessed by CD69 expression) were significantly increased in MCMV infected mice (Fig.1C, D), although the total number of these cells was not different

(Supplementary Fig.1). Thus, latent MCMV infection favors the preferential accrual of activated

Treg in the spleen.

Treg inhibit effector T cell responses and promote latent MCMV infection in the spleen.

Next, we determined the role of Treg in latent MCMV infection using Foxp3-DTR mice, which allows the specific depletion of Treg using diphtheria toxin (DT) [64]. Before DT administration the levels of MCMV-specific T cells were not significantly different between WT and Foxp3-

DTR mice (Supplementary Fig.2A). After DT administration, the frequency and total numbers of Treg were substantially reduced relative to DT-treated controls (Supplementary Fig.2B). As

MCMV-specific T cells control viral replication, we determined the role of Treg in suppressing

MCMV-specific T cell responses during latent infection. Treg depletion resulted in a significant increase in MCMV-specific CD8+ T and CD4+ T cells, relative to control animals (Fig.2A).

MCMV-specific CD8+ T cells in Treg depleted mice expressed markers of differentiation

(KLRG-1) and proliferation (Ki67) (Supplementary Fig.2C, D). Functionally, the frequency of

TNF-α or IFN-γ single producers and the numbers of single and double producers was significantly increased in Treg depleted mice (Fig.2B). Similarly, CD107α expressing MCMV-

111

specific CD8+ T cells were significantly increased after Treg depletion (Fig.2C). Thus, Treg

suppress effector T cell responses during latent MCMV infection.

Given the differences in effector T cell responses following Treg depletion, it was

important to evaluate if loss of Treg would modulate the latent MCMV viral load. Seven days

after initial DT treatment there was no actively replicating virus in the spleen in either controls or

Foxp3-DTR mice as assessed by plaque assay (Supplementary Table.2). Using a spleen explant

assay [65], we examined the impact of Treg on the latent viral pool. The spleen explant assay

provides a functional assay to assess reactivation from latency, and thus, indirectly provide

information on viral load levels since a lower latent viral load is expected to be less efficient at

reactivation [65]. As detected in the explant assay, for the WT control mice, 6 out of 9 mice had

evidence of viral reactivation by 28 days post explant culture, whereas MCMV reactivated only

in 3 out of 9 Foxp3-DTR mice (Fig.3A). Further, titers of reactivating virus in WT mice were

significantly higher in the first two weeks than those detected in the few Treg-depleted mice that

had reactivated MCMV (Fig.3B). To further evaluate whether reduced virus reactivation from

the spleen also correlated with reduced levels of viral DNA following Treg depletion, we

performed qPCR on total splenic DNA to quantify viral genomes. Although there was variation

from one experiment to the next, we saw a consistent decrease in viral genome levels in spleens

from Foxp3-DTR mice relative to C57BL/6 controls across four independent experiments

(Supplementary Fig.3, average percent decrease of 62.72% +/- 9.4; p<0.007). Thus, Treg are critical to maintain the reactivatable latent pool of MCMV in the spleen. However, the increase in overall spleen cellularity (and hence DNA content) following Treg depletion could also contribute to a decreased detection of viral load.

112

Treg are required to prevent MCMV reactivation and MCMV-specific CD4+ T cells in the

SG.

Given the role of Treg in promoting viral persistence in the spleen, we next determined the role

of Treg in control of latent MCMV infection in the SG. Similar to the spleen, latency in the SG

was established by 8 months as evidenced by the lack of replicating virus prior to DT

administration (Fig.4A). However, in stark contrast to the reduction in viral load in the spleen

upon Treg depletion at day 7, Treg depleted mice had detectable replicating virus in the SG.

While 3 out of 11 WT mice had a barely detectable viral reactivation, 9 out of 10 DT-treated

Foxp3-DTR mice had a significant increase in viral reactivation and viral titers in the SG

(Fig.4A, B). Indeed, in several experiments, virus was not detected in the WT SG whereas actively replicating virus was consistently detected in the SG of the Foxp3-DTR mice (Fig.4A,

B). Additionally, reactivation occurred as early as day 4 after Treg depletion with evidence of increased, albeit low, viral titers in reactivating mice observed at that time (Supplementary

Fig.4A, B). Thus, in stark contrast to the spleen, Treg are critical to prevent lytic viral reactivation in the SG.

The SG provides a site for MCMV viral persistence and is a major site for accrual of tissue resident MCMV-specific memory CD8+ and CD4+ T cells [30, 66], although MCMV- specific CD8+ T cells are unable to drive viral clearance in this organ [22]. Given that Treg inhibited viral reactivation in the SG, we next investigated the SG T cell responses following

Treg depletion in latently MCMV-infected mice. First, we established that DT administration efficiently depleted Treg in the SG (Supplementary Fig.5A). In contrast to the spleen, we did not observe any change in the MCMV-specific CD8+ T cells (Fig.4C) or tissue resident MCMV-

specific CD8+ T cells (CD103+CD69+) (Supplementary Fig.5B) in Treg depleted mice.

113

However, depletion of Treg resulted in an increase in the number of MCMV-specific CD4+ T cells in SG (Fig.4D). Thus, Treg suppress CD4+ but not CD8+ T cells in the SG during latent infection.

Treg suppress CD4+ Foxp3- IL-10+ cells in the SG and IL-10 is critical for limiting viral load in the SG after Treg depletion.

Prior work showed that IL-10 is critical for promoting viral replication in the SG [27, 28].

Indeed, we found that depletion of Treg drove a significant increase in SG IL-10 mRNA levels

(Fig.5A). Given the increase in CD4+ T cell responses in the SG, we next examined the potential cellular sources of IL-10. Importantly, CD4+Foxp3-cells were the predominant source for IL-10 production compared to CD8+ T cells and non–T cells (Fig.5B). Combined, these data show that

Treg suppress a population of CD4+ Foxp3- IL-10+ cells, consistent with a potential role of these cells in MCMV replication in the SG. As the effects of Treg depletion on viral reactivation/latency were quite different in the SG compared to the spleen, we examined IL-10 levels in the spleen as reduced IL-10 could potentially explain the different viral loads. Similar to the SG, IL-10-producing FoxPp3- CD4+ T cells were increased in the spleen (Supplementary

Fig. 6A). However, unlike the SG, total IL-10 mRNA was not increased in the spleen, suggesting that the total amount of IL-10 in the spleen is not substantially increased

(Supplementary Fig. 6B).

To further investigate the role of IL-10 in the SG after Treg depletion, we blocked IL-

10R signaling by administration of a blocking IL-10R antibody with and without Treg depletion using Foxp3-DTR mice (Fig.5C). As expected, Treg depletion was again accompanied by an increase in the number of mice harboring reactivating virus as measured by plaque assay

114

(Fig.5D). Strikingly, neutralization of IL-10 during Treg depletion significantly reduced the total number of mice with MCMV reactivation (9/13 to 3/13) (Fig.5D). Thus, our data show that Treg limit IL-10 production in the SG and that IL-10 is essential for viral reactivation/replication in the SG after Treg depletion.

Discussion:

Investigating the role of regulatory T cells in latent MCMV has been hampered by the lack of

appropriate tools. Our ability to assess the role of Treg in latent MCMV infection was greatly

facilitated by two essential tools. First, Foxp3-DTR mice, which express diphtheria toxin

receptor under the Foxp3 promoter allowed for depletion of Treg upon DT administration[64].

Second, the use of a sensitive tissue explant assay and qPCR allowed us to assess the reactivatable latent viral pool in mice with Treg depletion [65, 67]. Herein, we found that

depletion of Treg during latent MCMV infection had profound consequences. In the spleen, Treg

restrained MCMV-specific CD4+ and CD8+ T cells and promoted the latent viral pool. In stark

contrast, in the SG, Treg were essential to limit viral reactivation because they prevented the

emergence of IL-10- secreting Foxp3- CD4+ T cells. Thus, we demonstrate unique tissue-

specific functions of Treg in control of latent MCMV infection.

During latent MCMV infection, we found a significant increase in activated Treg in the

spleen. We speculate that the increase in Treg maybe a direct consequence of the chronic

stimulation due to the periodic low-level viral reactivation during latency. In agreement, in other

chronic infections like Leishmaniasis, hepatitis C virus (HCV), hepatitis B virus (HBV) and

human immunodeficiency virus (HIV), Treg frequency was substantially increased in the spleen

and other tissues [68-70]. Interestingly, human herpes virus 6 (HHV-6) infection, another herpes

115 virus closely related to HCMV, induces virus-specific CD4+ and CD8+ regulatory T cells [71].

Thus, chronic infections appear to promote their own persistence by driving Treg accrual.

Strikingly, Treg depletion resulted in markedly reduced latent virus in the spleen as measured by two assays. Some mice had undetectable viral DNA levels in the spleen, suggesting that viral load had been reduced to levels that were below the limit of detection in our assay. We acknowledge that the increase spleen size and cellularity could contribute to the observed reduction in latent viral load. However, in Treg-depleted mice where viral DNA load was undetectable, the spleen size and cellularity was similar to mice with detectable levels of viral DNA. Nonetheless, future experiments will investigate this in more detail, examining the number of cells harboring latent virus and the levels of virus within such cells. The spleen explant assay allowed us to assess one aspect of latent MCMV infection, the ability to reactivate from latency as a measure of whether reactivatable latent viral loads in the spleen were affected by Treg depletion. In our hands, if virus is replicating in the spleen at the time of isolation, this is detected by seven days post explants [65, 72]. However, if virus is latent in the tissue at the time of isolation, it takes longer for the virus to be detected in the explant assay, usually by day

14, and then rapidly spreads and replicates within the culture. Although this assay is not quantitative per se, we recently demonstrated that a mutant virus was unable to reactivate from the spleen by explant culture and had reduced levels of viral DNA [65], (Cardin, manuscript in preparation). Here, our results show that the Foxp3-DTR mice were less efficient at reactivation in this assay, suggesting lower levels of latent virus in the spleen. Importantly, we found that both the number of Foxp3-DTR mice with reactivating virus and also the levels of virus replication per mouse in the spleen explant cultures were reduced. Decreased reactivation from the spleen was highly reproducible between studies, and indeed, qPCR analysis of viral DNA in

116

the spleen could indicate lower levels of latent virus in mice depleted of Treg, with the caveat of

spleen size as mentioned earlier. Alternatively, we cannot rule out that the increased numbers of

virus-specific T cells in the Treg-depleted spleens are functional after placement into the explant

culture and thus elimination of latently-infected cells could also occur in vitro; however, these T cells likely have a very limited survival in vitro. It is important to note that we were unable to carry out long-term Treg depletion (greater than 14 days), because of the rampant and lethal autoimmunity that ensues with long-term Treg depletion in these mice [64]. Further, the impact of Treg depletion on latent viral genomes likely only impacted those genomes that underwent a reactivation event and became detectable by the immune system due to viral antigen expression.

Given that Treg depletion was accompanied by a substantial increase in cytotoxic MCMV- specific CD8+ T cells in the spleen, it is likely that Treg depletion led to the reactivation of some viral genomes, which was likely quickly quelled in the spleen by an MCMV-specific CD8+ T cell response, possibly even before lytic virus was produced [49]. Additional studies are needed to address this further.

One important question raised by our study is the mechanism(s) by which Treg suppress

CD8+ T cell responses to MCMV. Prior in vitro work suggested that Treg utilize TGF-β to suppress MCMV- specific T cell responses [31], although CTLA-4 and IL-10 may also have a regulatory role [73, 74]. In a chronic Friend virus (FV) infection model, FV-specific CD8+ T cells were also partially restrained by Treg, although the mechanism(s) appear to be independent of PD-1 and Tim-3 [75]. Interestingly, our previously published data show that Treg from aged mice have significantly increased levels of IL-35p19, suggesting that IL-35 may contribute to suppression of MCMV-specific CD8+ T cell responses [76]. However, a recent paper showed that IL-35 produced by NK cells during acute MCMV infection promotes, rather than inhibits

117

IL-10 production [77]. Nonetheless, further studies are required to determine the mechanism by

which Treg restrain MCMV-specific CD8+ T cell effector functions and thus promote latent

infections.

It was very surprising that, while the depletion of Treg resulted in reduction in viral load

in the spleen, it was accompanied by an augmented reactivation and/or replication of the latent

viral pool in the SG. We envision three major potential mechanisms that may explain these

results. First, it is possible that under chronic infection such as that observed in MCMV infected

SG, Treg could lose immunosuppressive abilities and acquire the phenotype of a more

pathogenic or anti-viral Treg. There is precedence for Treg metamorphosis in tumor models and

under chronic inflammatory conditions [78-81]. This makes teleological sense as chronic

inflammation may favor Treg with more effector function to replace exhausted effector T cells.

However, we looked for Treg expression of pathogenic markers (i.e. T-bet, RORγt) on SG Treg

and failed to find expression of these markers. Second, it is possible that the kinetics of latency

establishment between the two tissues explains the differential effects on latent virus (clearance

& reactivation) in the two tissues. For example, because the virus established latency in the

spleen after three months but was latent in the SG after 7 months, perhaps clearance was easier to

achieve in the spleen due to a lower level of latent viral load. However, we performed several

experiments examining Treg depletion 2-5 months after primary infection and found that, similar to 8 months after infection, that loss of Treg drove significant decrease in MCMV replication in the spleen (Supplementary Fig.7). Thus, it is likely that the tissue microenvironment rather than the timing controls the tissue-specific role of Treg. Third, it is possible that Treg maintain their

suppressive capacity and instead of their “classical” role of inhibiting pro-inflammatory T cells,

it is possible that they similarly control anti-inflammatory T cells. If so, the result of suppressing

118

anti-inflammatory cells would be the promotion of an immune-suppressive environment which is permissive for viral reactivation/replication. Our data are most consistent with this latter possibility. In this regard, CD4+ T cell production of IL-10 was increased in both the spleen and

SG after Treg depletion. One obvious question is why didn’t this increase in IL-10 drive viral reactivation in the spleen? One likely explanation is that, in the spleen, where CD8+ T cells are critical to prevent lytic virus production, their levels of IL-10R are significantly decreased, making them insensitive to IL-10 [82].

Our data also show that Treg depletion drives viral reactivation in the SG and implicates

IL-10 in the process. However, current data in the literature suggest that IL-10 contributes more to viral replication than outright reactivation [83]. Indeed, prior work has shown that the specific production of IL-10 from Foxp3- CD4+ cells attenuates acute antiviral immune-responses and leads to persistent viral replication in the SG [27]. Thus, while our data clearly show that inhibition of IL-10R signaling restored viral control, more work is required to conclusively determine whether IL-10 promotes outright reactivation or promotes viral replication. However, in our study, we initiated IL-10R antibody treatment following the initiation of Treg depletion, thus, if virus had started to reactivate, it could have been effectively controlled. The effect of IL-

10 on viral control could be direct or indirect. IL-10 can directly affect CD4+ T cell production of effector cytokines like IFN-γ [84]. Indeed, IFN-γ is indispensable for the control of viral load in the SG [25, 85]. However, there was no diminution in IFN-γ production upon Treg depletion, instead IFN-γ production was actually increased (Supplementary Fig.8). Alternatively, IL-10 can indirectly compromise CD4+ T cell responses in the SG by interfering with the responsiveness of APC to IFN-γ. For example, it is well known that IL-10 inhibits the effect of

IFN-γ by interfering with IFN-γ induced genes, preventing the phosphorylation of STAT-1

119

molecules and activation of monocytes [86]. Thus, while our data clearly show a role for IL-10,

more work is required to determine the cellular targets of IL-10 that regulate

reactivation/replication. Notably, the cellular site(s) of MCMV latency in host tissues is a long

debated issue. Like HCMV, MCMV can establish a latent infection in cells of the myeloid

lineage and these cells are able to respond to IL-10. However, other, non-hematopoietic targets,

like endothelial cells cannot be excluded.

This divergent and tissue-specific role in controlling MCMV viral latency by Treg raises

the question as to whether Treg manipulation is a useful therapy in latent HCMV infection. In

HCMV, manipulating Treg is viewed as a promising therapeutic approach to control latent viral

reactivation in immune-compromised hosts like organ transplant patients. In a prior study, the

use of immune-suppressive drugs like daclizumab (anti-CD25), steroids and calcineurin

inhibitors in CMV sero-positive renal transplant patients, led to reduction of Treg levels that

correlated with enhanced levels of CMV-specific effector T cells, suggesting that modulation of

Treg favors maintenance of CMV-specific immunity [87]. However, Treg depletion may also

promote viral reactivation in sites such as SG that are not assessed in treated patients. Thus,

manipulating Treg could be a double-edged sword. Treg in CMV infection might exert different

functional activity depending on their localization within the infected host and the T-cell

responses that they regulate. Our data, suggesting that Treg could regulate another suppressor

CD4+Foxp3-IL-10+ cells in the SG and limit viral reactivation opposed to their conventional role in regulating effector T cells as observed in the spleen, is a novel concept. It is unclear whether the mechanisms employed by Treg to inhibit effector T cell responses vs. other regulatory cell populations are distinct. Understanding such mechanisms might be crucial however to enhance the suppressive effects of Treg on other immune suppressive cell

120

populations in some instances (chronic infections) or block the suppressive effect of Treg on

others (i.e. autoimmunity).

Methods:

Cells and Virus

NIH 3T3 cells (ATCC CRL1658) were grown in Dulbecco’s modified Eagle’s medium (DMEM,

Media tech, Herndon, VA) supplemented with 10 % fetal bovine serum (FBS, Hyclone, Logan,

UT), 7.5% Sodium Bicarbonate, 4 mM HEPES, 2 mM L-glutamine, and gentamicin in a

humidified 5% CO2 incubator at37ºC. Parent stocks of the wild type MCMV K181 (originally a kind gift from Dr. Ed Mocarski, Stanford University) were prepared in NIH 3T3 cells from a SG-

derived virus stock as previously described [65]. Virus titers were determined by plaque assay on

NIH 3T3 cells. All virus stocks were stored at -70oC and re-titered before use in experiments.

Mice and infection

Young C57BL/6 mice were purchased from Taconic Farms (Germantown, NY)

Foxp3-IRES-DTR-GFP knock-in C57BL/6 mice were a generous gift from Dr. A. Rudensky.

(Rockefeller University, NY). For depletion of Foxp3 Treg cells, 1µg DT was injected at day 0.

Followed by 0.25µg DT on day 3, 6. Mice were sacrificed on day 7. For Treg depletion and IL-

10R neutralization, mice were injected with 1µg DT at day 0, followed by 0.25µg DT on day 3, 6 and 8. In addition, mice received antiIL-10R blocking antibody (Clone: 1B1.3A BioXcell) or ratIgG1 isotype control (Clone: HRPN BioXcell) 500µg on day 2, 5, 8 and 250µg at day 8. Mice were sacrificed on day 10. All mice were injected by intraperitoneal route (i.p.).

For MCMV infection, five to six week-old mice were infected with MCMV K181 strain, via i.p.

inoculation with 1 × 106 PFU. Mice were sacrificed upon establishment of latent MCMV at

121

various times (> 6-7 months post infection in C57Bl/6 mice). Mice were maintained under

specific-pathogen-free conditions at Cincinnati Children's Hospital Medical Center.

Ethics Statement

All animal protocols were reviewed and approved by the Institutional Animal Care and Use

Committee at the Cincinnati Children’s Hospital Research Foundation(CCHRF) under

IACUC2016-0087 (1D03023).

The care and use of laboratory animals at CCHMC is in accordance with the principles and

standards set forth in their Principles for Use of Animals (NIH Guide for grants and Contracts),

the Guide for the care and Use of Laboratory Animals (Department of Health, Education, and

Welfare DHEW, Public Health Service PHS, National Institutes of Health NIH Publ. 8th edition,

Rev.2011). The provisions of the Animal Welfare Acts (P.L. 89-544 and its amendments), and other applicable laws and regulations.

Virological Methods

For plaque assays, dilutions of virus stocks, 10% (w/v) mouse tissue sonicates, and sonicated

leukocyte cell suspensions were adsorbed onto 70% confluent NIH 3T3 monolayers for one hour

at 37oC, and then overlaid with 1:1 carboxymethyl cellulose (CMC): 2X modified Eagle’s

medium as previously described[85]. At 6 days, the overlay was removed and the cells were

fixed with methanol and stained with Giemsa to determine the number of plaques. For

measurement of tissue virus replication following mouse infection, tissues were placed in pre-

weighed tubes or were homogenized in media to prepare cell suspensions, followed by

sonication on ice to disrupt cells and release free virus. Tissue homogenates were titered by

plaque assay similar to virus stocks as described above.

Reactivation Assays

122

Explant reactivation assays of tissues from latently infected mice were established as previously

described [45, 65]. After infection when virus was no longer replicating and establishment of

latency (> 6-7 months post infection in C57Bl/6 mice) and following DT treatment, the SGs and spleens were collected. In some experiments, other tissues such as lungs and liver were also collected for plaque assays. The SGs were sonicated and titered to detect persistent replicating virus as the SG is a major site of viral persistence. In some experiments, to analyze populations of infiltrating leukocytes in the SGs, the SGs were first homogenized to prepare a cell suspension, an aliquot was removed for sonication and plaque assay analysis, and the remaining sample was treated as described below under ‘cell isolation’. For the explant reactivation assays, the spleens were minced and primary spleen cultures were established as previously described

[45].In some studies, SGs and lungs were also analyzed by explant reactivation assay. Briefly, the spleen explant assay was established by dividing the spleens into three parts, with each part placed into a well of a 6-well tissue culture plate containing 5 ml of media. The cultures were followed for up to 6 weeks and culture media from each well were collected weekly, sonicated and titered in plaque assays to detect the presence of reactivating or replicating virus. Plaques detected in any wells were counted as a reactivation event.

PCR Analysis

Tissues were isolated from infected and uninfected mice using dissection tools pre-treated with

DNA Away (Molecular BioProducts). DNA was isolated from cell suspensions following tissue homogenization using QIAamp DNA Mini Kit (Qiagen #51306) according to manufacturer’s protocol. Quantitative PCR was performed as previously described[88] using the following primers: E1 forward primer 5’-TCGCCCATCGTTTCGAGA-3’ ; E1 reverse primer 5’-

123

TCTCGTAGGTCCACTGACGGA-3’ to yield an amplified 106bp product. The TAMRA

Taqman E1 probe was purchased from Applied Biosystems.

Cell isolation

Spleens were harvested and crushed through 100-µm filters (BD Falcon) to generate single-cell

suspensions. For the SG, tissue was digested with collagenase (4) or with liberase in media

(RPMI 1640 containing 0.5 mg/ml digestion enzyme (Sigma), 5mM CaCl2, 0.2 mg/ml of DNase

I (Sigma) and 5% FBS) incubated for 45minutes at 37°C, followed by a two-step

discontinuousPercoll gradient (Little Chalfont, UK). Gradient samples were centrifuged at

25000rpm, room temperature, for 25 min with the brake off. The lymphocytes were harvested at

the interface between 30% and 70% Percoll layers.

Flow Cytometry

A total of 1 -2 million single-cell suspensions from either spleen or SG surface stained with a

combination of the following tetramers and antibodies:

M45 –HGIRNASFI, m139 – TVYGFCLL, M38 – SSPPMFRV, IE3 – RALEYKNL andM25 –

NHLYETPISATAMVI. Tetramers were all synthesized by the NIH tetramer core facility. Cells were then incubated with Fc block and surface stained with

Surface Abs: αCD4, αCD8α, αTCRβ (BD Biosciences, San Diego, CA, USA), αCD44,

αKLRG1, αCD127, αCD69, αCD103, αCX3CR1, αCD16/32 and αLy6C (eBioscience, San

Diego, CA, USA). For CD8 T cell intracellular staining, cells were stained in media and fixed with 2%Methanol free formaldehyde (MF FA) for 1 hour and then intracellularly stained with

αKi67 (eBioscience) using eBio permeabilization buffer. For CD8 T cell peptide stimulation using M38, M139 at 2µg/ml (a gift from Dr.Edith Janssen, Cincinnati Children's Hospital

Medical Center, Cincinnati,OH) cells were stimulated for 5 h in the presence of anti-CD107a

124

antibody and in the presence of brefeldinA for the final 4 h. Cells were then fixed with 2%MF

FA for 1 hour and then in 0.05% saponin. Cells were stained for IFN-γ and TNF-α production

(all from eBioscience). For CD4 T cells, cells were stimulated with 50ng/ml PMA and 1µg/ml

ionomycin for 5 hours, in the presence of brefeldinA for the final 4 h and fixed with 2%MF FA

for 1 hour followed by intracellular staining for IL-10, IFN-γ, TNF-α (all from eBioscience) and

Foxp3 (eBioscience) staining was done according to eBioscience Foxp3 staining kit and protocol. Data were acquired on an LSRII flow cytometer (BD Biosciences) and analyzed using

FACSDiva software (BD Biosciences).

RT-PCR

Samples were homogenized and total cellular RNA was extracted and quantified. DNase-treated

RNA was then used to synthesize cDNA. The primer sequences used for detection of IL-10 were: 5′- GCTCTTACTGACTGGCATGAG -3′ and 5′- CGCAGCTCTAGGAGCATGTG -3′.

The primer sequences used for detection of β-actin as an internal control were

5′-GGCCCAGAGCAAGAGAGGTA-3′ and 5′-GGTTGGCCTTAGGTTTCAGG-3′.

Quantitative real-time PCR was performed with Roche LightCycler 480 SYBRGreen 1 Master

Mix using the Roche LightCycler 480 II instrument (Roche Diagnostics).

Statistical Analysis

Data were analyzed using GraphPad Prism software and Excel software. Statistical analysis was performed using either a Student’s t-test or aFisher’s exact test in different experiments. A p- value of <0.05 was considered significant.

125

Acknowledgements

Thanks to Dr. Ian Humphreys (Cardiff University) for helpful advice on the IL-10R

neutralization strategy. This work was supported by NIH grants AG033057 (to C.A.C and

D.A.H), AG033057-S1 (to M.A.), and NIH R01 AI087683-01A1, Steinmann Family Foundation

CMV Award (R.D.C).

References:

1. Medzhitov R, Janeway CA, Jr. Innate immune recognition and control of adaptive immune responses. Semin Immunol. 1998;10(5):351-3. PubMed PMID: 9799709. 2. Finlay BB, McFadden G. Anti-immunology: evasion of the host immune system by bacterial and viral pathogens. Cell. 2006;124(4):767-82. doi: 10.1016/j.cell.2006.01.034. PubMed PMID: 16497587. 3. Schmid-Hempel P. Immune defence, parasite evasion strategies and their relevance for 'macroscopic phenomena' such as virulence. Philos Trans R Soc Lond B Biol Sci. 2009;364(1513):85-98. doi: 10.1098/rstb.2008.0157. PubMed PMID: 18930879; PubMed Central PMCID: PMCPMC2666695. 4. Bloom BR. Games parasites play: how parasites evade immune surveillance. Nature. 1979;279(5708):21-6. PubMed PMID: 88015. 5. White DW, Suzanne Beard R, Barton ES. Immune modulation during latent herpesvirus infection. Immunol Rev. 2012;245(1):189-208. doi: 10.1111/j.1600-065X.2011.01074.x. PubMed PMID: 22168421; PubMed Central PMCID: PMCPMC3243940. 6. Gupta G, Oghumu S, Satoskar AR. Mechanisms of immune evasion in leishmaniasis. Adv Appl Microbiol. 2013;82:155-84. doi: 10.1016/B978-0-12-407679-2.00005-3. PubMed PMID: 23415155; PubMed Central PMCID: PMCPMC3697132. 7. Bogdan C, Rollinghoff M. The immune response to Leishmania: mechanisms of parasite control and evasion. Int J Parasitol. 1998;28(1):121-34. PubMed PMID: 9504340. 8. Crough T, Khanna R. Immunobiology of human cytomegalovirus: from bench to bedside. Clinical microbiology reviews. 2009;22(1):76-98, Table of Contents. doi: 10.1128/CMR.00034- 08. PubMed PMID: 19136435; PubMed Central PMCID: PMCPMC2620639. 9. Reddehase MJ, Simon CO, Seckert CK, Lemmermann N, Grzimek NK. Murine model of cytomegalovirus latency and reactivation. Curr Top Microbiol Immunol. 2008;325:315-31. PubMed PMID: 18637514. 10. Zheng Q, Tao R, Shang SQ. [Modulation of host immune defenses by cytomegalovirus: advanced insights from evolutionary game theory]. Bing Du Xue Bao. 2013;29(1):85-91. PubMed PMID: 23547385.

126

11. Rawlinson WD, Farrell HE, Barrell BG. Analysis of the complete DNA sequence of murine cytomegalovirus. J Virol. 1996;70(12):8833-49. PubMed PMID: 8971012; PubMed Central PMCID: PMCPMC190980. 12. Vossen MT, Westerhout EM, Soderberg-Naucler C, Wiertz EJ. Viral immune evasion: a masterpiece of evolution. Immunogenetics. 2002;54(8):527-42. doi: 10.1007/s00251-002-0493- 1. PubMed PMID: 12439615. 13. Hengel H, Brune W, Koszinowski UH. Immune evasion by cytomegalovirus--survival strategies of a highly adapted opportunist. Trends in microbiology. 1998;6(5):190-7. PubMed PMID: 9614343. 14. Oliveira SA, Park SH, Lee P, Bendelac A, Shenk TE. Murine cytomegalovirus m02 gene family protects against natural killer cell-mediated immune surveillance. J Virol. 2002;76(2):885-94. PubMed PMID: 11752177; PubMed Central PMCID: PMCPMC136824. 15. Hengel H, Reusch U, Gutermann A, Ziegler H, Jonjic S, Lucin P, et al. Cytomegaloviral control of MHC class I function in the mouse. Immunological reviews. 1999;168:167-76. PubMed PMID: 10399073. 16. Sinzger C, Digel M, Jahn G. Cytomegalovirus cell tropism. Curr Top Microbiol Immunol. 2008;325:63-83. PubMed PMID: 18637500. 17. Loewendorf A, Benedict CA. Modulation of host innate and adaptive immune defenses by cytomegalovirus: timing is everything. J Intern Med. 2010;267(5):483-501. doi: 10.1111/j.1365-2796.2010.02220.x. PubMed PMID: 20433576; PubMed Central PMCID: PMCPMC2902254. 18. Mitrovic M, Arapovic J, Traven L, Krmpotic A, Jonjic S. Innate immunity regulates adaptive immune response: lessons learned from studying the interplay between NK and CD8+ T cells during MCMV infection. Med Microbiol Immunol. 2012;201(4):487-95. doi: 10.1007/s00430-012-0263-0. PubMed PMID: 22965169; PubMed Central PMCID: PMCPMC3500700. 19. Su HC, Nguyen KB, Salazar-Mather TP, Ruzek MC, Dalod MY, Biron CA. NK cell functions restrain T cell responses during viral infections. Eur J Immunol. 2001;31(10):3048-55. doi: 10.1002/1521-4141(2001010)31:10<3048::AID-IMMU3048>3.0.CO;2-1. PubMed PMID: 11592081. 20. Krmpotic A, Bubic I, Polic B, Lucin P, Jonjic S. Pathogenesis of murine cytomegalovirus infection. Microbes and infection / Institut Pasteur. 2003;5(13):1263-77. PubMed PMID: 14623023. 21. Jonjic S, Mutter W, Weiland F, Reddehase MJ, Koszinowski UH. Site-restricted persistent cytomegalovirus infection after selective long-term depletion of CD4+ T lymphocytes. J Exp Med. 1989;169(4):1199-212. PubMed PMID: 2564415; PubMed Central PMCID: PMCPMC2189231. 22. Walton SM, Mandaric S, Torti N, Zimmermann A, Hengel H, Oxenius A. Absence of cross-presenting cells in the salivary gland and viral immune evasion confine cytomegalovirus immune control to effector CD4 T cells. PLoS Pathog. 2011;7(8):e1002214. doi: 10.1371/journal.ppat.1002214. PubMed PMID: 21901102; PubMed Central PMCID: PMCPMC3161985. 23. Jeitziner SM, Walton SM, Torti N, Oxenius A. Adoptive transfer of cytomegalovirus- specific effector CD4+ T cells provides antiviral protection from murine CMV infection. Eur J Immunol. 2013;43(11):2886-95. doi: 10.1002/eji.201343690. PubMed PMID: 23921569.

127

24. Jonjic S, Pavic I, Lucin P, Rukavina D, Koszinowski UH. Efficacious control of cytomegalovirus infection after long-term depletion of CD8+ T lymphocytes. J Virol. 1990;64(11):5457-64. PubMed PMID: 1976821; PubMed Central PMCID: PMCPMC248597. 25. Lucin P, Pavic I, Polic B, Jonjic S, Koszinowski UH. Gamma interferon-dependent clearance of cytomegalovirus infection in salivary glands. J Virol. 1992;66(4):1977-84. PubMed PMID: 1312614; PubMed Central PMCID: PMCPMC288986. 26. Pahl-Seibert MF, Juelch M, Podlech J, Thomas D, Deegen P, Reddehase MJ, et al. Highly protective in vivo function of cytomegalovirus IE1 epitope-specific memory CD8 T cells purified by T-cell receptor-based cell sorting. J Virol. 2005;79(9):5400-13. doi: 10.1128/JVI.79.9.5400-5413.2005. PubMed PMID: 15827154; PubMed Central PMCID: PMCPMC1082747. 27. Humphreys IR, de Trez C, Kinkade A, Benedict CA, Croft M, Ware CF. Cytomegalovirus exploits IL-10-mediated immune regulation in the salivary glands. J Exp Med. 2007;204(5):1217-25. doi: 10.1084/jem.20062424. PubMed PMID: 17485516; PubMed Central PMCID: PMCPMC2118568. 28. Campbell AE, Cavanaugh VJ, Slater JS. The salivary glands as a privileged site of cytomegalovirus immune evasion and persistence. Med Microbiol Immunol. 2008;197(2):205- 13. doi: 10.1007/s00430-008-0077-2. PubMed PMID: 18259775. 29. Clement M, Marsden M, Stacey MA, Abdul-Karim J, Gimeno Brias S, Costa Bento D, et al. Cytomegalovirus-Specific IL-10-Producing CD4+ T Cells Are Governed by Type-I IFN- Induced IL-27 and Promote Virus Persistence. PLoS Pathog. 2016;12(12):e1006050. doi: 10.1371/journal.ppat.1006050. PubMed PMID: 27926930; PubMed Central PMCID: PMCPMC5142785. 30. Thom JT, Weber TC, Walton SM, Torti N, Oxenius A. The Salivary Gland Acts as a Sink for Tissue-Resident Memory CD8(+) T Cells, Facilitating Protection from Local Cytomegalovirus Infection. Cell Rep. 2015;13(6):1125-36. doi: 10.1016/j.celrep.2015.09.082. PubMed PMID: 26526997. 31. Li YN, Liu XL, Huang F, Zhou H, Huang YJ, Fang F. CD4+CD25+ regulatory T cells suppress the immune responses of mouse embryo fibroblasts to murine cytomegalovirus infection. Immunol Lett. 2010;131(2):131-8. doi: 10.1016/j.imlet.2010.03.011. PubMed PMID: 20381532. 32. Jost NH, Abel S, Hutzler M, Sparwasser T, Zimmermann A, Roers A, et al. Regulatory T cells and T-cell-derived IL-10 interfere with effective anti-cytomegalovirus immune response. Immunol Cell Biol. 2014;92(10):860-71. doi: 10.1038/icb.2014.62. PubMed PMID: 25070242. 33. Koffron AJ, Hummel M, Patterson BK, Yan S, Kaufman DB, Fryer JP, et al. Cellular localization of latent murine cytomegalovirus. J Virol. 1998;72(1):95-103. PubMed PMID: 9420204; PubMed Central PMCID: PMCPMC109353. 34. Henry SC, Hamilton JD. Detection of murine cytomegalovirus immediate early 1 transcripts in the spleens of latently infected mice. J Infect Dis. 1993;167(4):950-4. PubMed PMID: 8383725. 35. Collins T, Pomeroy C, Jordan MC. Detection of latent cytomegalovirus DNA in diverse organs of mice. J Infect Dis. 1993;168(3):725-9. PubMed PMID: 8394863. 36. Balthesen M, Messerle M, Reddehase MJ. Lungs are a major organ site of cytomegalovirus latency and recurrence. J Virol. 1993;67(9):5360-6. PubMed PMID: 8394453; PubMed Central PMCID: PMCPMC237936.

128

37. Bando K, Paradis IL, Komatsu K, Konishi H, Matsushima M, Keena RJ, et al. Analysis of time-dependent risks for infection, rejection, and death after pulmonary transplantation. J Thorac Cardiovasc Surg. 1995;109(1):49-57; discussion -9. PubMed PMID: 7815807. 38. Azevedo LS, Pierrotti LC, Abdala E, Costa SF, Strabelli TM, Campos SV, et al. Cytomegalovirus infection in transplant recipients. Clinics (Sao Paulo). 2015;70(7):515-23. doi: 10.6061/clinics/2015(07)09. PubMed PMID: 26222822; PubMed Central PMCID: PMCPMC4496754. 39. Stowe RP, Kozlova EV, Yetman DL, Walling DM, Goodwin JS, Glaser R. Chronic herpesvirus reactivation occurs in aging. Exp Gerontol. 2007;42(6):563-70. doi: 10.1016/j.exger.2007.01.005. PubMed PMID: 17337145; PubMed Central PMCID: PMCPMC1992441. 40. Kondo K, Kaneshima H, Mocarski ES. Human cytomegalovirus latent infection of granulocyte-macrophage progenitors. Proc Natl Acad Sci U S A. 1994;91(25):11879-83. PubMed PMID: 7991550; PubMed Central PMCID: PMCPMC45339. 41. Hahn G, Jores R, Mocarski ES. Cytomegalovirus remains latent in a common precursor of dendritic and myeloid cells. Proc Natl Acad Sci U S A. 1998;95(7):3937-42. PubMed PMID: 9520471; PubMed Central PMCID: PMCPMC19941. 42. Reeves M, Sinclair J. Aspects of human cytomegalovirus latency and reactivation. Curr Top Microbiol Immunol. 2008;325:297-313. PubMed PMID: 18637513. 43. Sinclair J. Human cytomegalovirus: Latency and reactivation in the myeloid lineage. J Clin Virol. 2008;41(3):180-5. doi: 10.1016/j.jcv.2007.11.014. PubMed PMID: 18164651. 44. Sinclair J, Sissons P. Latency and reactivation of human cytomegalovirus. J Gen Virol. 2006;87(Pt 7):1763-79. doi: 10.1099/vir.0.81891-0. PubMed PMID: 16760381. 45. Stoddart CA, Cardin RD, Boname JM, Manning WC, Abenes GB, Mocarski ES. Peripheral blood mononuclear phagocytes mediate dissemination of murine cytomegalovirus. J Virol. 1994;68(10):6243-53. PubMed PMID: 8083964; PubMed Central PMCID: PMCPMC237044. 46. Jarvis MA, Nelson JA. Human cytomegalovirus persistence and latency in endothelial cells and macrophages. Curr Opin Microbiol. 2002;5(4):403-7. PubMed PMID: 12160860. 47. Seckert CK, Renzaho A, Tervo HM, Krause C, Deegen P, Kuhnapfel B, et al. Liver sinusoidal endothelial cells are a site of murine cytomegalovirus latency and reactivation. J Virol. 2009;83(17):8869-84. doi: 10.1128/JVI.00870-09. PubMed PMID: 19535440; PubMed Central PMCID: PMCPMC2738169. 48. Polic B, Hengel H, Krmpotic A, Trgovcich J, Pavic I, Luccaronin P, et al. Hierarchical and redundant lymphocyte subset control precludes cytomegalovirus replication during latent infection. J Exp Med. 1998;188(6):1047-54. PubMed PMID: 9743523; PubMed Central PMCID: PMCPMC2212537. 49. Simon CO, Holtappels R, Tervo HM, Bohm V, Daubner T, Oehrlein-Karpi SA, et al. CD8 T cells control cytomegalovirus latency by epitope-specific sensing of transcriptional reactivation. J Virol. 2006;80(21):10436-56. doi: 10.1128/JVI.01248-06. PubMed PMID: 16928768; PubMed Central PMCID: PMCPMC1641801. 50. Lu X, Pinto AK, Kelly AM, Cho KS, Hill AB. Murine cytomegalovirus interference with antigen presentation contributes to the inability of CD8 T cells to control virus in the salivary gland. J Virol. 2006;80(8):4200-2. doi: 10.1128/JVI.80.8.4200-4202.2006. PubMed PMID: 16571839; PubMed Central PMCID: PMCPMC1440476.

129

51. Klenerman P, Oxenius A. T cell responses to cytomegalovirus. Nat Rev Immunol. 2016;16(6):367-77. doi: 10.1038/nri.2016.38. PubMed PMID: 27108521. 52. Liu XF, Wang X, Yan S, Zhang Z, Abecassis M, Hummel M. Epigenetic control of cytomegalovirus latency and reactivation. Viruses. 2013;5(5):1325-45. doi: 10.3390/v5051325. PubMed PMID: 23698401; PubMed Central PMCID: PMCPMC3712310. 53. Hummel M, Abecassis MM. A model for reactivation of CMV from latency. J Clin Virol. 2002;25 Suppl 2:S123-36. PubMed PMID: 12361763. 54. Hummel M, Zhang Z, Yan S, DePlaen I, Golia P, Varghese T, et al. Allogeneic transplantation induces expression of cytomegalovirus immediate-early genes in vivo: a model for reactivation from latency. J Virol. 2001;75(10):4814-22. doi: 10.1128/JVI.75.10.4814- 4822.2001. PubMed PMID: 11312353; PubMed Central PMCID: PMCPMC114236. 55. Li Z, Wang X, Yan S, Zhang Z, Jie C, Sustento-Reodica N, et al. A mouse model of CMV transmission following kidney transplantation. Am J Transplant. 2012;12(4):1024-8. doi: 10.1111/j.1600-6143.2011.03892.x. PubMed PMID: 22226173. 56. Fietze E, Prosch S, Reinke P, Stein J, Docke WD, Staffa G, et al. Cytomegalovirus infection in transplant recipients. The role of tumor necrosis factor. Transplantation. 1994;58(6):675-80. PubMed PMID: 7940686. 57. Docke WD, Prosch S, Fietze E, Kimel V, Zuckermann H, Klug C, et al. Cytomegalovirus reactivation and tumour necrosis factor. Lancet. 1994;343(8892):268-9. PubMed PMID: 7905100. 58. Simon CO, Seckert CK, Dreis D, Reddehase MJ, Grzimek NK. Role for tumor necrosis factor alpha in murine cytomegalovirus transcriptional reactivation in latently infected lungs. J Virol. 2005;79(1):326-40. doi: 10.1128/JVI.79.1.326-340.2005. PubMed PMID: 15596827; PubMed Central PMCID: PMCPMC538715. 59. Kurz SK, Reddehase MJ. Patchwork pattern of transcriptional reactivation in the lungs indicates sequential checkpoints in the transition from murine cytomegalovirus latency to recurrence. J Virol. 1999;73(10):8612-22. PubMed PMID: 10482614; PubMed Central PMCID: PMCPMC112881. 60. Grzimek NK, Dreis D, Schmalz S, Reddehase MJ. Random, asynchronous, and asymmetric transcriptional activity of enhancer-flanking major immediate-early genes ie1/3 and ie2 during murine cytomegalovirus latency in the lungs. J Virol. 2001;75(6):2692-705. doi: 10.1128/JVI.75.6.2692-2705.2001. PubMed PMID: 11222693; PubMed Central PMCID: PMCPMC115894. 61. Reddehase MJ, Podlech J, Grzimek NK. Mouse models of cytomegalovirus latency: overview. J Clin Virol. 2002;25 Suppl 2:S23-36. PubMed PMID: 12361754. 62. Hengel H, Lucin P, Jonjic S, Ruppert T, Koszinowski UH. Restoration of cytomegalovirus antigen presentation by gamma interferon combats viral escape. J Virol. 1994;68(1):289-97. PubMed PMID: 8254740; PubMed Central PMCID: PMCPMC236288. 63. Schwele S, Fischer AM, Brestrich G, Wlodarski MW, Wagner L, Schmueck M, et al. Cytomegalovirus-specific regulatory and effector T cells share TCR clonality--possible relation to repetitive CMV infections. Am J Transplant. 2012;12(3):669-81. doi: 10.1111/j.1600- 6143.2011.03842.x. PubMed PMID: 22081907. 64. Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol. 2007;8(2):191-7. doi: 10.1038/ni1428. PubMed PMID: 17136045.

130

65. Cardin RD, Schaefer GC, Allen JR, Davis-Poynter NJ, Farrell HE. The M33 chemokine receptor homolog of murine cytomegalovirus exhibits a differential tissue-specific role during in vivo replication and latency. J Virol. 2009;83(15):7590-601. doi: 10.1128/JVI.00386-09. PubMed PMID: 19439478; PubMed Central PMCID: PMCPMC2708650. 66. Smith CJ, Caldeira-Dantas S, Turula H, Snyder CM. Murine CMV Infection Induces the Continuous Production of Mucosal Resident T Cells. Cell Rep. 2015;13(6):1137-48. doi: 10.1016/j.celrep.2015.09.076. PubMed PMID: 26526996; PubMed Central PMCID: PMCPMC4648370. 67. Farrell HE, Abraham AM, Cardin RD, Molleskov-Jensen AS, Rosenkilde MM, Davis- Poynter N. Identification of common mechanisms by which human and mouse cytomegalovirus seven-transmembrane receptor homologues contribute to in vivo phenotypes in a mouse model. J Virol. 2013;87(7):4112-7. doi: 10.1128/JVI.03406-12. PubMed PMID: 23345521; PubMed Central PMCID: PMCPMC3624218. 68. Miroux C, Vausselin T, Delhem N. Regulatory T cells in HBV and HCV liver diseases: implication of regulatory T lymphocytes in the control of immune response. Expert opinion on biological therapy. 2010;10(11):1563-72. doi: 10.1517/14712598.2010.529125. PubMed PMID: 20932226. 69. Moreno-Fernandez ME, Presicce P, Chougnet CA. Homeostasis and function of regulatory T cells in HIV/SIV infection. J Virol. 2012;86(19):10262-9. doi: 10.1128/JVI.00993- 12. PubMed PMID: 22811537; PubMed Central PMCID: PMCPMC3457299. 70. Walton S, Mandaric S, Oxenius A. CD4 T cell responses in latent and chronic viral infections. Front Immunol. 2013;4:105. doi: 10.3389/fimmu.2013.00105. PubMed PMID: 23717308; PubMed Central PMCID: PMCPMC3651995. 71. Wang F, Chi J, Peng G, Zhou F, Wang J, Li L, et al. Development of virus-specific CD4+ and CD8+ regulatory T cells induced by human herpesvirus 6 infection. J Virol. 2014;88(2):1011-24. doi: 10.1128/JVI.02586-13. PubMed PMID: 24198406; PubMed Central PMCID: PMCPMC3911638. 72. Cardin RD, Abenes GB, Stoddart CA, Mocarski ES. Murine cytomegalovirus IE2, an activator of gene expression, is dispensable for growth and latency in mice. Virology. 1995;209(1):236-41. doi: 10.1006/viro.1995.1249. PubMed PMID: 7747475. 73. Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nature reviews Immunology. 2008;8(7):523-32. doi: 10.1038/nri2343. PubMed PMID: 18566595; PubMed Central PMCID: PMCPMC2665249. 74. Walker LS, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol. 2011;11(12):852-63. doi: 10.1038/nri3108. PubMed PMID: 22116087. 75. Dietze KK, Zelinskyy G, Liu J, Kretzmer F, Schimmer S, Dittmer U. Combining regulatory T cell depletion and inhibitory receptor blockade improves reactivation of exhausted virus-specific CD8+ T cells and efficiently reduces chronic retroviral loads. PLoS Pathog. 2013;9(12):e1003798. doi: 10.1371/journal.ppat.1003798. PubMed PMID: 24339778; PubMed Central PMCID: PMCPMC3855586. 76. Raynor J, Karns R, Almanan M, Li KP, Divanovic S, Chougnet CA, et al. IL-6 and ICOS Antagonize Bim and Promote Regulatory T Cell Accrual with Age. J Immunol. 2015;195(3):944-52. doi: 10.4049/jimmunol.1500443. PubMed PMID: 26109645; PubMed Central PMCID: PMCPMC4506860.

131

77. Jensen H, Chen SY, Folkersen L, Nolan GP, Lanier LL. EBI3 regulates the NK cell response to mouse cytomegalovirus infection. Proc Natl Acad Sci U S A. 2017;114(7):1625-30. doi: 10.1073/pnas.1700231114. PubMed PMID: 28143936; PubMed Central PMCID: PMCPMC5321027. 78. Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29(1):44-56. doi: 10.1016/j.immuni.2008.05.007. PubMed PMID: 18585065; PubMed Central PMCID: PMCPMC2630532. 79. Koenen HJ, Smeets RL, Vink PM, van Rijssen E, Boots AM, Joosten I. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood. 2008;112(6):2340-52. doi: 10.1182/blood-2008-01-133967. PubMed PMID: 18617638. 80. Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nature medicine. 2016;22(6):679-84. doi: 10.1038/nm.4086. PubMed PMID: 27111280. 81. Addey C, White M, Dou L, Coe D, Dyson J, Chai JG. Functional plasticity of antigen- specific regulatory T cells in context of tumor. Journal of immunology. 2011;186(8):4557-64. doi: 10.4049/jimmunol.1003797. PubMed PMID: 21389255. 82. Jones M, Ladell K, Wynn KK, Stacey MA, Quigley MF, Gostick E, et al. IL-10 restricts memory T cell inflation during cytomegalovirus infection. J Immunol. 2010;185(6):3583-92. doi: 10.4049/jimmunol.1001535. PubMed PMID: 20713884; PubMed Central PMCID: PMCPMC3655265. 83. Mandaric S, Walton SM, Rulicke T, Richter K, Girard-Madoux MJ, Clausen BE, et al. IL-10 suppression of NK/DC crosstalk leads to poor priming of MCMV-specific CD4 T cells and prolonged MCMV persistence. PLoS Pathog. 2012;8(8):e1002846. doi: 10.1371/journal.ppat.1002846. PubMed PMID: 22876184; PubMed Central PMCID: PMCPMC3410900. 84. Naundorf S, Schroder M, Hoflich C, Suman N, Volk HD, Grutz G. IL-10 interferes directly with TCR-induced IFN-gamma but not IL-17 production in memory T cells. Eur J Immunol. 2009;39(4):1066-77. doi: 10.1002/eji.200838773. PubMed PMID: 19266486. 85. Pavic I, Polic B, Crnkovic I, Lucin P, Jonjic S, Koszinowski UH. Participation of endogenous tumour necrosis factor alpha in host resistance to cytomegalovirus infection. J Gen Virol. 1993;74 ( Pt 10):2215-23. doi: 10.1099/0022-1317-74-10-2215. PubMed PMID: 8105025. 86. Ito S, Ansari P, Sakatsume M, Dickensheets H, Vazquez N, Donnelly RP, et al. Interleukin-10 inhibits expression of both interferon alpha- and interferon gamma- induced genes by suppressing tyrosine phosphorylation of STAT1. Blood. 1999;93(5):1456-63. PubMed PMID: 10029571. 87. Presser D, Sester U, Mohrbach J, Janssen M, Kohler H, Sester M. Differential kinetics of effector and regulatory T cells in patients on calcineurin inhibitor-based drug regimens. Kidney Int. 2009;76(5):557-66. doi: 10.1038/ki.2009.198. PubMed PMID: 19494797. 88. Snyder CM, Allan JE, Bonnett EL, Doom CM, Hill AB. Cross-presentation of a spread- defective MCMV is sufficient to prime the majority of virus-specific CD8+ T cells. PLoS One. 2010;5(3):e9681. doi: 10.1371/journal.pone.0009681. PubMed PMID: 20300633; PubMed Central PMCID: PMCPMC2837378.

132

Figures

Fig.1. Activated Treg are increased in the spleen during latent MCMV. Splenocytes were isolated

from naïve (9.5months, white bars), or aged matched MCMV-latently infected mice (black bars, 8 months post- inoculation with1× 10 pfu of MCMV). Cells were stained for CD4, CD69, and Foxp3 and analyzed by flow cytometry. Dot plots⁶ (A) and bar graph (B) show the frequency of Foxp3+ cells in total CD4+ cells from naïve and MCMV infected mice as described above (mean+SEM). Representative histograms (C) and bar graph (D) show the frequency of CD69+ on gated Foxp3+ cells from naïve and MCMV infected mice as described above (mean+SEM). Data is representative of two independent experiments. Naïve

(N=4), WT MCMV infected (N=6). Statistical analysis (**p ≤ 0.01, Student’s t-test).

133

Fig.2. Treg inhibit effector T cell responses during latent MCMV infection in the spleen. 5-6 week old WT C57BL/6 (white bars) and Foxp3DTR (black bars) mice were inoculated with1× 10 pfu of MCMV. 8 months post-MCMV infection, both groups were injected with Diphtheria toxin (DT) on⁶ day 0, 3, 6 and sacrificed on day 7. Splenocytes from infected mice were stained with M25 (CD4 T cell) and M45, IE3, m139, M38 (CD8 T cell) tetramers day 7 (+DT) and analyzed with flow cytometry. Bar graphs (A) show the total number of M25- specific CD4 T cells and M45-, IE3-, m139- and M38-specific CD8 T cells from

MCMV infected WT C57BL/6 (white bars, N=6-7) and Foxp3DTR (black bars, N= 6-8) mice (mean+SEM).

134

Data are pooled from two independent experiments. Spleen cells (2 x 106 cells/well) from MCMV infected

mice day 7 (+DT) were ex vivo stimulated with M38 peptide (5 h at 37°C) and treated with Brefeldin A and

were stained for CD8, and IFN-γ, TNF-α and analyzed by flow cytometry. Representative plots and bar graphs (B) show the average frequency and total number of CD8+ T lymphocytes producing IFN-γ and/or

TNF-α (mean+SEM). WT C57BL/6 (N=6), Foxp3DTR (N=6). Spleen cells from infected groups (+DT) were ex-vivo stimulated with MCMV-peptide (M38) for 5 hours during which CD107α antibodies were incubated with the cells. Representative plots and bar graphs (C) show the average frequency and total number of

CD8+T lymphocytes expressing CD107α (mean+SEM).WT C57BL/6 (N=11), Foxp3DTR(N=10). Statistical analysis, *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001 (Student’s t test).

135

Fig.3. Treg promote latent MCMV infection in the spleen. 5-6 week old WT control and Foxp3DTR mice were inoculated with 1× 10 pfu of MCMV (N=9/group). 8 months post-MCMV infection, both groups were injected with Diphtheria toxin⁶ (DT) on day 0, 3, 6 and sacrificed on day 7. Spleens were analyzed for reactivation from latency by spleen explant assay as described in methods. Supernatants were assayed for infectious virus by plaque assay weekly. Bar graph (A) indicates the percentage of mice positive for virus reactivation from the spleens in either WT control (white bars) and Foxp3DTR (black bars) mice, with the numbers of positive mice shown above the bars. Bar graph (B) represents average of virus titer of replicating virus in the supernatants of the spleen explant cultures of WT control (white bars) and

Foxp3DTR (black bars) MCMV infected mice day7 post Treg depletion (mean+SEM). Data include all of the mice in the experiment, including those mice with undetectable virus. A total of 1.5 ml of the explant cultures, representing ~37% of the total supernatant, was titered. Mice with undetectable amounts of virus were given a value of zero. Statistical analysis, (Student’s t test) *p ≤ 0.05.

136

Fig.4. Treg are required to prevent MCMV reactivation and MCMV-specific CD4+ T cells in the SG.

5-6 week old WT C57BL/6 and Foxp3DTR mice were inoculated with1× 10 pfu of MCMV. 8 months post-

MCMV infection, both groups were injected with Diphtheria toxin (DT) on day⁶ 0, 3, 6 and sacrificed on day

7. Bar graph (A) shows the percentage of WT C57BL/6 uninfected control (UI, N=2 day 0, N=2 day 7, gray bar), MCMV infected (N=3 day 0, N=11 day7 white bars) and Foxp3DTR (N=6 day 0, N=10 day 7,

black bars) mice positive for virus replication in the SGs before Treg depletion (indicated as day0) and

day7 post Treg depletion with the numbers of positive mice in each group shown above the bars. (B) Bar graph shows the average viral titer of individual SGs of MCMV infected mice before Treg depletion

(indicated as day0) and day7 post Treg depletion as described above (mean+SEM). The presence of replicating virus was detected by plaque assay. Samples with no detectable virus were assigned a titer of

0.7 log pfu/ml, the limit of detection for the plaque assay as indicated by the dashed line. Data is representative of five independent experiments. Statistical analysis, *p ≤ 0.05, **p ≤ 0.01 (Student’s t tests) comparing infected mice. Single cell suspensions were generated from the SGs of MCMV infected mice and were stained first with MCMV-tetramers, M38 (CD8), M25 (CD4) and then cells were surface

137 stained for CD8, CD4, and analyzed with flow cytometry. Bar graph (C) shows the total number of M38- specific CD8 T cells in the SGs (mean+SEM). WT C57BL/6 (N=4). Foxp3DTR (N=5). Bar graph (D) shows the total number of M25-specific CD4 T cells in the SGs (mean+SEM). WT C57BL/6 (N=6). Foxp3DTR

(N=8). Statistical analysis, *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001 (Student’s t test).

138

Fig.5. Treg suppress CD4+ Foxp3- IL-10+ cells in the SG and IL-10 is critical for limiting viral load in the SG after Treg depletion. 5-6 weeks old WT C57BL/6 (white bars) and Foxp3DTR (black bars) mice

were inoculated with 1× 10 pfu of MCMV. 8 months post-MCMV infection, both groups were injected with

Diphtheria toxin (DT) on day⁶ 0, 3, 6 and sacrificed on day 7. Bar graph (A) shows the average normalized

IL-10 mRNA level (mean+SEM) in SG extracts of MCMV-infected mice (day7) post Treg depletion. WT

C57BL/6 (N=11). Foxp3DTR (N=10). Single cell suspensions were generated from the SGs of MCMV

infected mice (day7 post Treg depletion). Cells were stained for TCRß, CD4, CD8, Foxp3 and IL-10

139

following stimulation with or without PMA and ionomycin for 5 hours, in the presence of brefeldinA. Bar

graph (B) shows the average of frequency of IL-10+ in Foxp3- CD4+ TCRβ+ and CD8+ TCRβ+ cells and

TCR-β− cells (mean+SEM) in the SG. WT C57BL/6 (N=5). Foxp3DTR (N=5). Statistical analysis, *p ≤ 0.05,

**p ≤ 0.01 (Student’s t test). (C) Schematic representation of diphtheria toxin and IL-10 neutralization treatment. Briefly, 5-6 week old WT C57BL/6 (N=24) and Foxp3DTR (N=26) mice were inoculated with 1×

10 plaque-forming unit (pfu, p.i.) of MCMV. 8-12 months post-MCMV infection, both groups were injected⁶ with DT on day 0, 3, 6, 8 and sacrificed on day 10. Groups were split and were treated with

500µg of either isotype control antibody or with anti–IL-10R neutralizing antibody on day 2, 5, 8 and were sacrificed on day10. Bar graph (D) indicates the percentage of mice positive for virus reactivation in the

SGs (day 10) post Treg depletion and IL-10R neutralization with the numbers of mice in each group shown above the bars. WT C57BL/6 uninfected (UI, N=6), WT C57BL/6 MCMV infected (N=24) and

Foxp3DTR (N=26). The presence of replicating virus was detected by plaque assay (data combined from two independent experiments). Statistics for percentage of mice with virus reactivation which was made using Fisher’s exact test (*P ≥ 0.05, ** P≥ 0.01, *** P≥ 0.001) comparing all groups.

140

Supporting information:

Day(0) Naïve WT Foxp3-DTR MCMV MCMV Spleen 0/2 0/3 0/6

Lung 0/2 0/3 0/6 Liver 0/2 0/3 0/6

Pancreas 0/2 0/3 0/6 Salivary 0/2 0/3 0/6 Gland

S1 Table. Establishment of latent MCMV infection. Table shows the number of mice with positive

MCMV titers (replicating virus) in the spleen, lung, liver, pancreas and salivary gland within the three groups: Naïve, WT control and Foxp3-DTR, 8 months post MCMV infection. Titers were quantified via plaque assay before Treg depletion, indicated here as Day0. 0/number of mice in each group indicates absence of actively replicating virus and confirms the establishment of latency in all tissues.

141

S2 Table. Absence of actively replicating virus in the spleen in controls and Foxp3-DTR mice 7 days post Treg depletion. Table shows the number of mice with positive MCMV titers (replicating virus) in the spleen within the two groups: WT control and Foxp3-DTR, 8 months post MCMV infection. Titers were quantified by plaque assay 7 days after Treg depletion, indicated here as Day7. 0/number of mice in each group indicates absence of actively replicating virus and confirms the establishment of latency.

(N=9/group).

142

S1 Fig. Treg in the spleen during latent MCMV. Splenocytes were isolated from naïve (9.5months), or aged matched MCMV-latently infected mice (8months p.i.). Cells were stained for CD4 and Foxp3 and analyzed by flow cytometry. Graph shows the number of Foxp3+ cells in total CD4+ cells. Naïve (N=4),

WT MCMV infected (N=6).

143

S2 Fig. Highly activated, proliferating MCMV-specific CD8+ T cells in the spleen post Treg depletion. 5-6 week old WT C57BL/6 and Foxp3DTR mice were inoculated with 1× 10 pfu of MCMV. 8 months post-MCMV infection, splenocytes were isolated from infected mice. Cells were⁶ stained for IE3, m139, M38 (CD8 T cell) tetramers day 0 (-DT) and analyzed with flow cytometry. A) Graph shows the total number of IE3-, m139- and M38-specific CD8 T from WT C57BL/6 (white bars, N=3) and Foxp3DTR

(black bars, N=6) mice. 5-6 week old C57BL/6 and Foxp3DTRmice were inoculated with1× 10 pfu of

MCMV. 8 months post-MCMV infection, both groups were injected with Diphtheria toxin (DT) on day⁶ 0, 3,

6 and sacrificed on day 7. Spleen cells were analyzed by flow cytometry. B) Bar graphs show the average

frequency and absolute number of CD4+ Foxp3+Treg in the spleen (mean+SEM). WT C57BL/6 (N=11).

Foxp3DTR (N=10). Spleen cells isolated from the two groups were stained with MCMV CD8-specific tetramers and then surface stained for expression of KLRG-1 and CD127 and intra-cellular expression of

Ki67. C) Bar graph shows the total numbers of effector subpopulations within gated m139-specific CD8 T cells (mean+SEM). WT C57BL/6 (N=6), Foxp3DTR (N=6). D) Bar graphs show the frequency and absolute number of Ki67+ cells within M45-, IE3-, m139- and M38-specific CD8 T (mean+SEM). WT C57BL/6

(N=6), Foxp3DTR(N=6). Statistical analysis, *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001 (Student’st-test).

144

S3 Fig. MCMV viral load in the spleen. Genomic DNA was isolated from the spleens of WT C57BL/6

and DTR mice at day 7 post treg depletion. MCMV E1 was detected by quantitative PCR, and data

expressed as genome copy number per 100 ng genomic DNA as described in Materials and Methods.

Results are pooled from 4 independent experiments (WT, N=20 and DTR, N=22) and show the

mean+SEM. The average reduction in viral load in DTR mice in 4 independent experiments was 62.7% ±

9.4 (one sample t-test p<0.007).

145

S4 Fig. Early MCMV viral reactivation post Treg depletion in the SG. 5-6 week old WT C57BL/6

(white) and Foxp3DTR (black) mice were inoculated with 1× 10 pfu of MCMV (N=8/group). 9 months post-

MCMV infection, both groups were injected with Diphtheria toxin⁶ (DT) on day 0, 3 and sacrificed on day4.

A) Bar graph shows the percentage of mice positive for virus reactivation of naïve (UI) and WT MCMV infected mice in the SGs day4 post Treg depletion with the numbers of mice in each group shown above the bars. The presence of replicating virus was detected by plaque assay. B) Bar graph shows viral titers of individually homogenized SGs of naïve and MCMV infected mice Day4 post Treg depletion

(mean+SEM).

146

S5 Fig. Efficiency of Treg depletion in the SG. 5-6 weeks old WT C57BL/6 and Foxp3DTR mice were

inoculated with1× 10 pfu of MCMV. 8 months post-MCMV infection, both groups were injected with

Diphtheria toxin (DT) ⁶on day 0, 3, 6 and sacrificed on day 7. A single cell suspension from the SGs of

naïve and MCMV infected mice was analyzed by flow cytometry. A) Bar graphs show the average

frequency and absolute number of CD4+ Foxp3+ Treg in the SG upon DT administration (mean+SEM).

WT C57BL/6 (N=7) Foxp3DTR (N=8). B) Bar graph shows the average frequency of tissue resident memory (CD103+CD69+) cells in the SG within the total M38-specific CD8 T cells (mean+SEM). C57BL/6

(N=4). Foxp3DTR (N=5). Statistical analysis, *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001 (Student’s t-test).

147

S6 Fig. Treg suppress CD4+ Foxp3- IL-10+ cells in the spleen. 5-6 weeks old WT C57BL/6 (white

bars) and Foxp3DTR (black bars) mice were inoculated with 1× 10 pfu of MCMV. 8 months post-MCMV

infection, both groups were injected with Diphtheria toxin (DT) on ⁶day 0, 3, 6 and sacrificed on day 7. A)

Single cell suspensions were generated from the spleen of MCMV infected WT C57BL/6 (N=11) and

Foxp3DTR (N=10) mice (day7) post Treg depletion. Cells were stained for TCRß, CD4, CD8, Foxp3 and IL-

10 following stimulation with or without PMA and ionomycin for 5 hours, in the presence of brefeldinA in

the final 4hrs. Bar graph shows the average of frequency of IL-10+ in Foxp3- CD4+ TCRβ+ and CD8+

TCRβ+ cells and TCR-β− cells (mean+SEM). B) Graph shows the average normalized IL-10 mRNA level in spleen of MCMV-infected WT C57BL/6 (N=10) and Foxp3DTR (N=9) mice (day7) post Treg depletion

(mean+SEM). Statistical analysis, *p ≤ 0.05, **p ≤ 0.01 (Student’s t test).

148

S7 Fig. Treg promote MCMV replication in the spleen. 5-6 week old WT C57BL/6 (white) and

Foxp3DTR (black) mice were inoculated with 1× 10 pfu of MCMV (N=8/group). 5 months post-MCMV infection, both groups were injected with Diphtheria toxin⁶ (DT) on day 0, 3, 6, 9,12 and sacrificed on day

14. A) Bar graph shows the percentage of mice positive for virus replication in the spleen day14 quantified by plaque assay post Treg depletion with the numbers of mice in each group shown above the bars. Viral titers were 18.6 pfu/ml +/- 15.5 in WT C57BL/6 mice and 2.4 pfu/ml +/- 2.24 in FoxP3-DTR mice; p=0.31. B) Genomic DNA was isolated from the spleens of WT C57BL/6 and DTR mice at day 14 post Treg depletion. MCMV E1 was detected by quantitative PCR, and data expressed as genome copy number per 100 ng genomic DNA as described in Materials and Methods (mean+SEM); p=0.36.

149

S8 Fig. IFN-ƴ production upon Treg depletion in the SG.

Single cell suspensions were generated from the SGs of MCMV infected mice (day7 post Treg depletion).

Cells were stained for CD4, Foxp3 and IFN-ƴ following stimulation with or without PMA and ionomycin for

5 hours, in the presence of brefeldinA. Bar graph shows the average of frequency of IFN-ƴ+ in Foxp3-

CD4+ (mean+SEM) in the SG. WT C57BL/6 (N=5). Foxp3DTR (N=5). Statistical analysis, *p ≤ 0.05

(Student’s t test).

150

Chapter4

IL-10 PRODUCING TFH CELLS COUNTER INFLAMMAGING BUT SUPPRESS HUMORAL IMMUNE RESPONSES

Maha Almanan1, Jana Raynor1, Ireti Ogunsulire2, Shibabrata Mukherjee1,4, Anna Malyshkina7, Jennifer Ingram3, Ankur Saini1,4, Markus M. Xie6, Theresa Alenghat1,4, George S. Deepe, Jr. 5, Senad Divanovic1, Harinder Singh1,4, Emily Miraldi1,4, Allan Zajac3, Dent, A.L.6, Christoph Hölscher2, Claire Chougnet1,4, and David A. Hildeman1,4¶* 1 Division of Immunobiology, Cincinnati Children's Hospital, University of Cincinnati College of

Medicine, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH

45229 and 4 Center for Systems Immunology, Cincinnati Children's Hospital, Cincinnati, OH 45229

2 Infection Immunology Research, Research Center Borstel, Borstel, Germany

3 Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL

5 Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267

6 Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN

7Institute of Virology, Essen University Hospital, Essen, Germany

*Corresponding authors Email: [email protected]

Running title:

IL-10-secreting T follicular helper cells Keywords: Treg, IL-10, Tfh, aging, CD4+

151

Abstract

Immune responses deteriorate with age and result in the decline of vaccine responsiveness.

Chronic low-level inflammation termed inflammaging is associated with the impairment of adaptive immune responses; however, the underlying mechanisms remain unclear. Here, we show that aged mice exhibit increased systemic IL-10 that is primarily produced by CD4+

FoxP3- T cells. These cells manifest a T follicular helper (Tfh) profile but express low levels of Bcl6 thereby enabling robust IL-10 expression. Consistent with their designation as Tfh10 cells, IL-6 and IL-21 are required for their accumulation with age. IL-21 promotes Tfh10 survival and maintains a systemic balance between IL-6 and IL-10. Importantly, blockade of

IL-10R signaling significantly restores Tfh-dependent antibody responses in aged mice.

Notably, we also found that Tfh10 cells are significantly increased in elderly humans. We propose that Tfh10 cells counter-regulate inflammaging but, in so doing, lead to impaired humoral responses with age.

152

Introduction Declining immune function is well described in the elderly, leading to increased risk and severity of infection, poorer control of cancer, and impaired responses to vaccination (Lord,

2013; Zhang et al., 2017). Despite such declining adaptive immune function, aging is also characterized by a persistent low-grade immune activation (so-called inflammaging). Such inflammaging is implicated in many deleterious processes in the elderly, including Alzheimer’s disease, cardiovascular diseases and general frailty (Giunta et al., 2008; North and Sinclair,

2012). Indeed, high levels of circulating, pro-inflammatory IL-6 are associated with increased morbidity and mortality among older individuals (Maggio et al., 2006).

Most inflammatory reactions incite potent anti-inflammatory feedback loops (Hanada and Yoshimura, 2002). Amongst anti-inflammatory mediators, IL-10 has broad-ranging and potent properties and can arise in response to or concurrently with inflammatory stimuli

(Couper et al., 2008). Recent work has shown a significant age-related increase in serum IL-

10 in a cross-sectional analysis of more than 465 subjects, ranging in age from 21 to 88 years

(Lustig et al., 2017). In addition, an IL-10 genotype (-21082GG) that is associated with high production of IL-10 in Caucasians was more prevalent in centenarians than in younger individuals (65-73 yrs) (Lio et al., 2002), and similarly more prevalent in middle-aged controls than in age-matched patients with myocardial infarction (Lio et al., 2004). Also, elderly men with the highest serum levels of inflammatory cytokines, or with the lowest levels of IL-10, had the highest risk of frailty-associated pathologies (Cauley et al., 2016). Thus, IL-10 levels, appears to play an important role in counter-regulation of inflammation to promote healthy aging.

In contrast to its beneficial roles in aging, IL-10 limits protective responses to pathogens. IL-10 plays a deleterious role in chronic infections, in mice and humans, limiting microbial clearance (Belkaid et al., 2001; Brooks et al., 2006). Indeed, we recently showed 153 that IL-10 fosters reactivation and replication of cytomegalovirus in the salivary glands of latently infected mice (Almanan et al., 2017). Further, IL-10 dampens responses to infections or vaccines (Dobber et al., 1995; McKinstry et al., 2009). In fact, in humans, the IFN-γ:IL-10 ratio appears to be a critical predictor to protective immunity in the elderly (McElhaney et al.,

2006). Despite these proposed roles for IL-10, little is known about its function, its cellular source, or its molecular control in aged animals or humans.

IL-10 can be produced by many cells, including those of the innate immune system

(notably multiple myeloid cell subsets), the adaptive immune system (T cells and B cells), and even non-immune cells (e.g., keratinocytes and hepatocytes) (Moore et al., 2001). At baseline in the spleen of young mice, the majority of IL-10 expression is localized to B cells and CD4+

T cells (CD25+ and CD25-) (Madan et al., 2009). In aging, a few studies have investigated the cellular sources of IL-10 and suggested a role for memory CD4 T cells. Indeed, early work showed increased IL-10 production by aged CD44hi CD4+ T cells (Hobbs et al., 1994). More importantly, the proportion of IL-10+ influenza-specific CD4+ T cells increases in aged mice

(Lefebvre et al., 2016b). In contrast, B cells capable of IL-10 production appear to be decreased in older subjects (van der Geest et al., 2016). Nonetheless, the major cellular source of IL-10 in aging remains unclear.

Here, we report that systemic levels of IL-10 are increased in aged mice and negatively impact vaccine responsiveness. Notably, we found that CD4+ FoxP3-, not classic FoxP3+, cells were required for increased systemic IL-10 levels in aging. Further, these IL-10- producing T cells bore markers of T follicular helper cells (Tfh), were present in both mice and humans, and they required IL-6 for their accumulation. Interestingly, IL-21, another promoter of Tfh homeostasis, was also required for the accrual of these cells, and, importantly, to regulate the systemic balance between IL-6 and IL-10. Mechanistically, we also found that the canonical Tfh transcription factor, BCL6, was downregulated with age in Tfh cells, permitting 154 their IL-10 production. Together, our data show that inflammation and anti-inflammation are linked via IL-21 production, which promotes accrual of IL-10-secreting Tfh (Tfh10) cells that function to dampen immune responses and IL-6-driven inflammaging.

Results: Aged mice have increased systemic levels of IL-10. While IL-10 levels have been shown to increase in aged humans (Lustig et al., 2017), it is unclear if IL-10 levels increase in aged mice. Using the sensitive in vivo cytokine capture assay (IVCCA) we found that steady-state levels of IL-10 in the serum were increased 2-3 fold in old compared to young mice (Figure 1A). To determine the potential sources of this enhanced IL-10, we examined various lymphoid and non-lymphoid tissues and found an increase in IL-10 mRNA in the epididymal white adipose tissue (WAT), lymph nodes, and spleen of aged, compared to young mice (Figure 1B). These data show that the systemic levels of IL-10 are increased with age and that secondary lymphoid organs appear to be major contributors of augmented IL-10 expression in aging.

CD4+ FoxP3- T cells are the major source of IL-10 in aged mice.

To identify cells with enhanced IL-10 production in aged mice, we took advantage of IL-

10-reporter (VertX) mice, which possess an IL-10-IRES-eGFP cassette in the endogenous IL-

10 locus (Madan et al., 2009). VertX mice allowed us to examine baseline IL-10-production directly ex vivo, in the absence of exogenous stimulation, as GFP levels in these mice directly correlate with IL-10-production (Madan et al., 2009). Flow cytometric analysis of spleen cells in aged versus young VertX mice revealed a significantly increased frequency of GFP+ (IL-

10+) cells in multiple cell types, but the largest increase was observed in CD4+ T cells (Figure

2A). As B cells are the predominant immune cell type in the spleen, the total numbers of

GFP+ B cells were increased with age. However, there was no significant difference in the 155 numbers of aged IL-10+ B cells compared to IL-10+ CD4+ T cells (Figure 2A). Instead, the largest increase in the frequency of IL-10-producing cells was in CD4+ T cells (Figure 2A). In addition, the level of IL-10 produced per cell was significantly higher in CD4+ T cells than in either CD8+ T cells, CD19- or CD19+ B cells (Figure 2A).

Because FoxP3+ regulatory T cells (Treg) are a well-known source of IL-10 in young mice, and their frequency is increased in old mice (Raynor et al., 2012), we next determined whether they were the major contributor to this increased IL-10 in aged mice. Staining for

FoxP3 in VertX mice while maintaining GFP expression is technically infeasible, so we sorted naive (CD4+CD44loCD62LhiFoxP3GFPneg), memory (CD4+CD44hiCD62LloFoxP3GFPneg) and regulatory (FoxP3GFPpos) T cells from young and aged FoxP3-DTR-GFP mice (Kim et al.,

2007), stimulated them with PMA and Ionomycin (P+I), and measured their production of IL-

10 by ELISA. As expected, naïve T cells produced little IL-10 whether they were from young or old mice (Figure 2B). IL-10 production from FoxP3+ Treg was slightly increased in aged mice (~2-fold), (Figure 2B). However, IL-10 production from aged FoxP3- memory T cells was increased >10-fold (Figure 2B). Similarly, flow cytometric analysis of spleen cells of WT mice showed that the frequency of IL-10-producing CD4+ FoxP3+ cells was increased slightly with age, while IL-10-producing FoxP3- CD4+ T cells were ~10-times more frequent with age

(Figure 2C). Again, Foxp3- CD4 T cells showed significantly higher expression of IL-10 per cell compared to their young counterparts and aged Foxp3+ cells (supplementary figure 1).

Together, these three independent approaches show that CD4+ FoxP3- cells have the highest capacity for IL-10-production in the spleens of aged mice. In addition, they are required for the increased systemic levels of IL-10, as depletion of >95% of CD4+ T cells in the spleens of old mice nearly returned the serum levels of IL-10 to levels observed in young mice (Figure 2D). In contrast, depletion of FoxP3+ T cells increased systemic IL-10 levels

156

(Figure 2D) and the frequency of IL-10-producing CD4+ T cells (Figure 2E). Thus, FoxP3-, but not FoxP3+, CD4+ T cells are required for the increased systemic levels of IL-10.

Accrual of IL-10-producing CD4+ FoxP3-T cells occurs in germ-free animals.

Recent work has shown that the microbiome changes with age (Odamaki et al., 2016;

Thevaranjan et al., 2017). Further, alterations in the microbiome can affect IL-10 production from CD4+ FoxP3+ and FoxP3- T cells (Mazmanian et al., 2008; Round and Mazmanian,

2010). To test whether the microbiome affects the accumulation of IL-10-producing CD4+ T cells, we aged several cohorts of mice in a germ-free facility. Interestingly, the accumulation of IL-10+ CD4+ FoxP3- cells was similar between age-matched mice housed under specific pathogen free conditions and germ-free animals across a range of ages (supplementary figure 2). Interestingly, we also show that age-driven changes to the microbiome do not appear to alter the accrual of IL-10-producing CD4+ T cells as these cells accumulate in germ-free mice. Further, the age-driven accrual of IL-10-producing CD4+ T cells occurred at four different institutions including: Cincinnati Children’s Hospital, Indiana University/Purdue

University Indianapolis, the University of Alabama-Birmingham, and the Research Center

Borstel in Germany. It is unlikely that the microbiomes of mice are the same at these different institutions. Therefore, the microbiome is not required for the accumulation of IL-10-producing

CD4+ FoxP3- T cells.

IL-10-producing CD4+ FoxP3- T cells in aged mice are predominantly Tfh cells.

Several distinct subsets of CD4+ FoxP3- T cells have been reported to produce IL-10, predominantly Th2, type I regulatory (TR1) T cells, “exTh17” cells, and exTreg cells (Gagliani et al., 2015; Roncarolo et al., 2014; Wang et al., 2005). Although they expressed LAG3, it is unlikely that the majority of the IL-10-producing cells were TR1 cells as they lacked expression of CD49b (supplementary figure 3A), an important marker on TR1 cells (Gagliani 157 et al., 2013). Very few IL-10+ CD4+T cells were capable of IL-4 or IL-17A co-production, ruling out the possibility that these were Th2 or Th17 cells (supplementary figure 3B). Next, analysis of IL-17A fate tracking mice (Hirota et al., 2011) revealed that the frequency of

“exTh17” cells within IL-10+ FoxP3- CD4+ T cells from aged mice was ~1% (supplementary figure 3C). Analysis of exTreg cells using FoxP3-CreRosaloxstoplox dTomato mice (Zhou et al.,

2008) (Madisen et al., 2010) revealed that ~20% of the IL-10+CD4+ T cells were dTomato+

GFP- “exTregs” in both young and old mice (supplementary figure 3D).Thus, neither Th2,

TR1, exTh17, nor exTreg make up the bulk of the IL-10-producing CD4+ T cells that accumulate in aged mice.

In our investigation of cytokine co-production by IL-10-producing CD4+ T cells, we found that the frequency and total numbers of IL-10+ cells that co-produced IL-21 was significantly increased in aged, compared to young, mice (Figure 3A). As IL-21 is typically produced by T follicular helper (Tfh) cells, we next assessed the frequency of IL-10+ CD4+ FoxP3- T cells that expressed CXCR5 and PD1, two canonical surface markers of Tfh cells, in conjunction with the transcription factor BCL6 (Haynes et al., 2007; Johnston et al., 2009). Strikingly, we found that the majority of IL-10+ FoxP3- CD4+ T cells were CXCR5+ and PD1+ in old mice

(Figure 3B). Further, there was a progressive age-related accrual of CXCR5+ PD1+ Tfh cells, including those that produce IL-10 (supplementary figure 4). Thus, the majority of the IL-10- producing T cells that accumulate with age bore markers of Tfh cells so for clarity, we will refer to them as Tfh10 cells.

IL-6 is required for Tfh10 development and for systemically increased IL-10 in aged mice.

We next examined the role of IL-6 in the accrual of Tfh10 T cells because IL-6: (i) controls Tfh development (Eto et al., 2011); (ii) promotes IL-10-production from CD4+ T cells (Jin et al.,

158

2013); and (iii) is a key inflammatory cytokine that is increased with age (Volpato et al., 2001).

To determine whether IL-6 promotes the accrual of Tfh10 cells with age, we aged IL-6-/- mice

≥16 mo and examined the proportion of Tfh10 cells. While no difference in the Tfh cells

(including those that produce IL-10) was observed between young WT and IL-6-/- mice, aged

IL-6-/- mice exhibited a dramatic reduction in the frequency and total number of Tfh and Tfh10 cells compared to aged WT mice (Figures 4A, 4B). Consistent with Tfh10 T cells being a major source of IL-10 in vivo, we found that systemic levels of IL-10 were significantly decreased in aged IL-6-/- mice (Figure 4C). To determine whether IL-6 was required for the development or survival of IL-10-producing FoxP3- CD4+ T cells, we blocked IL-6 after Tfh cells were formed and found that neutralization of IL-6 did not reduce the frequency or numbers of IL-10-producing cells (Figure 4D). Thus, IL-6 is required for the accrual of Tfh10 cells, likely by promoting their initial development.

IL-21 promotes accumulation of Tfh10 cells and regulates systemic IL-6/IL-10 balance.

As IL-21 is a critical cytokine produced by Tfh cells (Nurieva et al., 2007), we next examined whether IL-6 promoted IL-21 production by CD4+ T cells. As expected, and consistent with elevated Tfh cells with age, the proportion and absolute number of IL-21+ CD4+ T cells was significantly increased in aged, compared to young, mice (Figure 5A). However, in the absence of IL-6, the frequency and total numbers of IL-21-producing CD4+ T cells was completely abrogated (Figure 5A). As IL-21 is also critical for the development and homeostasis of Tfh cells (Vogelzang et al., 2008), we reasoned that IL-21 could contribute to the accrual of Tfh10 T cells with age. Similar to aged IL-6-/- mice, the loss of IL-21 prevented age-driven accrual of Tfh cells (Figure 5B) including those that produce IL-10 (Figure 5C).

Again, consistent with the loss of Tfh10 T cells, levels of systemic IL-10 were reduced in aged

159

IL-21-/- mice compared to aged wild type controls (Figure 5D). Strikingly, the levels of IL-6 were increased in IL-21-deficient aged mice (Figure 5E). Together, these data show that IL-

21 is critical to balance systemic inflammation (e.g IL-6/IL-10 levels), likely by promoting the accrual of Tfh10 cells. As IL-6 and IL-21 have been reported to increase ICOS which is critical for survival of Tfh cells (Akiba et al., 2005), we considered the possibility that increased levels of ICOS on aged Tfh cells could be contributing to their accumulation. Interestingly, we found a significant but marginal effect of ICOS-L neutralization on overall Tfh cell number and no effect on IL-10 producing cells (supplementary figure 5). These data show that IL-21 plays a key role in promoting accrual of Tfh10 cells with age, whose production of IL-10 likely feeds back to suppress IL-6.

IL-21 promotes repression of Bim in aged Tfh10 cells leading to their enhanced survival.

The accumulation of Tfh10 cells with age could be due to their increased proliferation and/or increased survival. The frequency of Tfh10 T cells that stained positive for the proliferation marker Ki-67+ actually decreased with age, ruling out the possibility that increased proliferation explains their accrual (Figure 6A). Given our and others previous data implicating the pro-apoptotic molecule Bim in T cell survival, (Chougnet et al., 2011;

Tsukamoto et al., 2010), we examined the role of Bim in the survival of IL-10-producing CD4+

T cells. First, Bim levels were reduced in IL-10-producing CD4+ T cells from aged compared to young mice (Figure 6B). Second, the frequency and total number of CD4+ FoxP3- T cells that were IL-10+ was significantly increased in Bim-deficient mice, as early as 6 months of age (Figure 6C). Given that IL-21 promotes accumulation of Tfh10 cells, we next determined whether IL-21 contributed to their reduced expression of Bim. Indeed, IL-21 was critical to suppress the levels of Bim within Tfh10 cells, which likely contributes to their increased

160 survival (Figure 6D). Together, these data suggest that IL-21-driven suppression of Bim contributes to the accumulation of IL-10-producing cells by enhancing their survival.

Tfh10 cells in aged mice manifest diminished levels of BCL6 thereby enabling IL-10 expression.

BCL6 is essential for Tfh differentiation and is induced by IL-6 and IL-21, so we examined

BCL6 levels in young versus aged Tfh cells. Interestingly, BCL6 levels were actually decreased in aged Tfh10 cells (Figure 7A). Further, decreased levels of BCL6 were associated with higher levels of IL-10 (Figure 7A). To determine whether BCL6 is required for the accrual of Tfh cells as well as their production of IL-10 in aged mice we utilized CD4Cre-

BCL6f/f mice that have a T cell-specific loss of BCL6 (Hollister et al., 2013). As expected, given that BCL6 is critical for promoting Tfh cells (Yu et al., 2009), CD4Cre-BCL6f/f mice had a significant loss of CXCR5 and PD1 expressing CD4+ Foxp3- T cells (Figure 7B). Strikingly, already by one year of age, the loss of BCL6 led to a significant increase in the frequency and total number of CD4+ FoxP3- cells that produced IL-10 (Figure 7C). As BCL6 has been reported to suppress expression of Blimp1 and Blimp1 has been shown to promote IL-10 expression from T cells (Neumann et al., 2014), we next examined Blimp1 levels in aged mice with and without BCL6. Interestingly, the levels of Blimp1 did not change in CD4+ FoxP3- cells that produced IL-10 from aged mice, whether or not BCL6 was present, making it unlikely that Blimp1 is promoting increased IL-10 expression in aged CD4+ FoxP3- T cells

(supplementary figure 6). Further, our prior data show that the additional loss of Blimp did not reduce IL10-producing CD4+ FoxP3- T cells in BCL6-deficient mice (Xie et al., 2017). Thus,

Bcl-6 suppresses IL-10 production, likely via a Blimp1-independent mechanism.

IL-10 limits Tfh-dependent vaccine responses in aged mice.

We next sought to determine the physiologic relevance of Tfh10 cells with age. As vaccine 161 responsiveness is a major problem in elderly humans and Tfh cells are critical regulators of vaccine responses (Crotty, 2011), we used a classic mouse model of a Tfh-dependent antibody response, immunization with NP-KLH. We reasoned that, if Tfh10 cells were important for regulating vaccine responses, then limiting IL-10 signaling should affect vaccine responsiveness. As reported before (Sage et al., 2015), we confirmed that old mice displayed a significantly lower level of anti-NP antibody production, as well as significantly lower frequency and total numbers of NP-specific B cells compared to young mice (Figure 8A).

Importantly, neutralization of IL-10R during NP-KLH immunization significantly restored anti-

NP antibody production as well as the frequency and numbers of anti-NP-specific B cells to levels close to those observed in young mice (Figure 8B). Thus, IL-10 limits Tfh-dependent B cell responses in aged mice.

Tfh10 cells accumulate during aging in humans.

Given the above data in mice implicating Tfh10 cells as regulators of vaccine responsiveness, and the fact that vaccine responses decline significantly in aged humans (Lord, 2013), we next determined whether Tfh10 cells accumulated in aged humans. The fact that Tfh cells are located, and function, in secondary lymphoid organs, required us to analyze their proportion in the spleens of young and old organ donors. Importantly, the frequency of Tfh cells (CXCR5+

PD-1+) was increased in aged humans (Figure 9A). Because flow cytometric analysis of cytokines is affected by cryopreservation, we FACS-sorted memory CD4+ T cells (CD45RO+) into Tfh (CD25-CD127+PD-1+CXCR5+), Treg (CD25+CD127-PD-1-CXCR5-) and other memory cells (CD25-CD127+PD-1-CXCR5-) and analyzed their production of IL-10 and IL-21 after in vitro re-stimulation with anti-CD3/CD28 beads. As expected, IL-21 production was largely limited to Tfh cells and was significantly increased with age (Figure 9B). Strikingly, the population with the highest production of IL-10 was the old Tfh cells (Figure 9C). Thus, similar

162 to mice, Tfh10 cells accumulate in aged humans and may explain the well-known age-related impairment in vaccine responsiveness.

DISCUSSION Our data show a novel linkage between two age-related phenomena, inflammaging and immune suppression. Notably, we found that IL-6 (a hallmark of inflammaging) is critical for the emergence of IL-10-expressing Tfh cells in aged mice. IL-10 is a critical as a feedback mechanism to dampen IL-6-driven inflammaging. However, the increased IL-10 also results in the impairment of vaccine responsiveness. Thus, further understanding of the mechanism(s) underlying the regulation and function of Tfh10 cells may provide insights and potential therapeutic approaches to harness the beneficial effects of IL-10 (minimizing systemic inflammation and organ dysfunction, (Couper et al., 2008) while reducing the harmful effects of IL-10, one of which is the well-known reduction in vaccine responses with age.

Although we showed that IL-10 limits GC B cell responses to an NP-KLH vaccine in aged mice, the targets of IL-10 remain unclear. The effects of IL-10 on B cells is controversial, some studies suggest that IL-10 promotes the survival of germinal center (GC) B cells (Levy and Brouet, 1994). On the other hand, others have shown that IL-10 limits GC B cell responses, consistent with our data (Cai et al., 2012). One potential explanation for these seemingly disparate observations may be due to the use of in vitro versus in vivo model systems. Indeed, much of the work showing positive role of IL-10 on GC B cells has been done in vitro; while work showing a negative role of IL-10 on GC B cells has been done in vivo. Nonetheless, more work is required to understand the cellular targets of IL-10 that are required to suppress antibody responses.

163

While cell intrinsic defects in adaptive immune cells clearly contribute to age-related immune suppression, our data show a substantial contribution of IL-10. These data suggest that cell intrinsic defects can be at least partially reversed by IL-10 neutralization or inhibition.

This is consistent with prior work showing that pro-inflammatory stimuli can restore vaccine responsiveness in aged mice (Maue et al., 2009). Similarly, in aged humans a strong pro- inflammatory adjuvant MF59 was able to significantly boost antibody responses to influenza

(Della Cioppa et al., 2012). In both studies, increases in IFN-γ producing CD4+ T cell responses was observed. All together, these data show that the aged immune system is amenable to at least partial restoration by manipulating the inflammatory environment.

Our work is also consistent with prior data showing an increase in Tfh cells in aged mice (Lefebvre et al., 2016a) (Lefebvre et al., 2016b). These prior studies suggested that subtle maturation defects in aged Tfh cells (slightly decreased levels of GL7, CXCR5 and

ICOS), accounted for their decreased ability to provide help to B cells. However, their homing to GCs was not significantly affected compared to young mice when the data were corrected for GC size (Lefebvre et al., 2016b). Further, although these authors observed an increase in

IL-10-producing CD4+ T cells after influenza infection in aged mice, this increase was not attributed to Tfh cells, but rather to Treg or T follicular regulatory cells (Lefebvre et al., 2016b).

However, in our aged cohorts of mice, neither Tfr nor Treg appear to be substantial contributors to IL-10 production in vivo. At baseline, without acute infection, we found that IL-

10-producing Tfh cells are required for elevated systemic levels of IL-10 in aged mice.

Notably, we show that IL-6 and IL-21 are individually required to drive the accumulation of Tfh10 cells, consistent with prior work suggesting a unique role for each cytokine in Tfh development and homeostasis (Choi et al., 2013). Our data also suggest a role for IL-6 in the development, but not the maintenance of Tfh10 cells. Indeed, we saw a precipitous loss of

Tfh10 cells in IL-6-deficient mice, but no change in Tfh10 cells when IL-6 was neutralized after 164 their formation, similar to a recent study in which IL-6 was neutralized during development of a Tfh response in aged mice (Tsukamoto et al., 2015). Part of this IL-6-driven developmental program involves induction of IL-21 production (Nurieva et al., 2008), which our and others data suggest may be required for the long-term maintenance of Tfh and Tfh10 cells

(Linterman et al., 2010). Our data also provide some molecular insight into the role of IL-21 on maintenance of Tfh10 cells in that we show it is required to suppress their expression of

Bim, which regulates their long-term survival. This concept of dual cytokines individually promoting Th cell development vs maintenance may be a common theme for Th cells as a similar phenomenon was recently observed in Th2 cells with TSLP and IL-4 (Rochman et al.,

2018).

Paradoxically, unlike the loss of IL-6 and IL-21, the loss of BCL6, a transcription factor essential for Tfh development, actually enhanced the accrual of IL-10-producing CD4+

FoxP3- T cells. Although we were unable to ascribe a Tfh status to these cells as the Tfh signature markers CXCR5 and PD1 were both substantially reduced in the absence of BCL6, our data clearly show that BCL6 is a major negative regulator of IL-10 production. This result likely reveals the dual nature of BCL6. On the one hand, BCL6 is critical for Tfh development and for expression of the canonical Tfh markers CXCR5 and PD1, while on the other hand,

BCL6 represses IL-10 expression. Indeed, we find little IL-10-production from Tfh cells in young mice, who maintain high expression of BCL6. However, with age, BCL6 levels decline and IL-10 production from Tfh cells increases.

Repression of IL-10 by BCL6 in Tfh10 cells could occur via two, non-mutually exclusive mechanisms. First, BCL6 could act indirectly, through repression of Prdm1 (Blimp1) expression (Johnston et al., 2009), as Blimp1 promotes IL-10 expression in both CD4+ and

CD8+ T cells (Neumann et al., 2014). However, we recently showed that the additional loss of Blimp1 did not reduce enhanced IL-10 production in the absence of BCL6 in young mice 165

(Xie et al., 2017). Second, BCL6 could act directly to repress IL-10 expression by binding to sites in the IL-10 locus (Hollister et al., 2013). Given the negative data implicated Blimp1, our data are more consistent with a scenario in which BCL6 represses IL-10 expression in young

Tfh cells, however, the loss of BCL6 with age de-represses IL-10 production from Tfh cells, although this remains to be rigorously tested.

Importantly, our data may provide an explanation for prior work in elderly humans showing that the antigen-stimulated IFN-γ:IL-10 ratio is directly correlated with cell-mediated immunity to influenza infection and inversely correlated with the severity of infection

(McElhaney et al., 2016) and also with seroconversion following influenza vaccination (Corsini et al., 2006). Our data shows that Tfh cells are a major source of T cell derived IL-10 in aged mice and possibly in elderly humans and that blockade of IL-10 signaling largely restored vaccine responses in mice. These data suggest that transient blockade of IL-10 could be a novel strategy to enhance vaccine responses in the elderly and, due to its transient nature, is unlikely to have untoward effects on autoimmunity, cardiovascular disease or frailty.

Materials and Methods: Mice: Young (6weeks-4months) C57BL/6 mice were purchased from Taconic Farms

(Germantown,NY). Old (≥16months) or Middle age (12-15months) C57BL/6 mice were from

National Institutes of Aging colony located at Charles River Laboratories (Wilmington, MA).

Foxp3-IRES-DTR-GFP knock-in C57BL/6 mice (Kim et al., 2007), were a generous gift from

Dr. A. Rudensky and were aged in house. Bim-deficient (Bim knockout) mice were originally a kind gift from Drs. P. Bouillet and A. Strasser and were bred in-house. IL-6–deficient (IL-6 KO) mice on the C57BL/6 background were aged in-house. IL-10-reporter (VertX) mice which possess an IL-10-IRES-eGFP cassette in the endogenous IL-10 locus on the C57BL/6

166 background (Madan et al., 2009), were aged in-house. Young, middle age and old Germ-free mice on the C57BL/6 background were maintained in isolator units in the CCHMC Gnotobiotic

Mouse Facility. Young and old FoxP3-fate mapping mice (Foxp3CreRosa26dTomato) on the

C57BL/6 background were kindly provided by Dr. Sing S. Way (CCHMC). IL-17A fate tracking mice IL-17CreRosa26eYFP (Hirota et al., 2011) on the C57BL/6 background were bred and aged under specific-pathogen free conditions in the animal facility of the Research Centre

Borstel, Germany. Young and old IL-21–deficient (IL-21KO) mice on the C57BL/6 background were bred, maintained and aged in fully accredited facilities at the University of

Alabama at Birmingham. Spleens (controls and IL-21KO) were shipped overnight on ice and analyzed in Cincinnati. CD4Cre BCL6f/f mice on the C57BL/6 background were bred, maintained and aged in fully accredited facilities at the University of Indiana. Spleens (CD4Cre

BCL6f/f and control) were shipped overnight on ice and analyzed in Cincinnati. All animal protocols were reviewed and approved by the Institutional Animal Care and Use Committee at the Cincinnati Children’s Hospital Research Foundation (IACUC 2016-0087).

Immunization, neutralization and depletion treatments:

For depletion of Foxp3 Treg cells in old FoxP3-DTR mice, 1.25µg DT/mouse was injected i.p. and were sacrificed two days later. For CD4 T cell depletion, mice were injected with a single dose of 600ug /mouse of anti-CD4 i.p. (Clone: YTS191 BioXcell) or isotype control (Clone:

LFT-2 BioXcell) and were sacrificed two days later. For T cell–dependent immunization, mice were immunized intraperitoneally with 100 µg NP-KLH (Biosearch Technologies) mixed with

50% (vol/vol) alum (Thermo Scientific) and sacrificed 20 days later. For IL-10R neutralization, mice were injected with antiIL-10R blocking antibody (Clone: 1B1.3A BioXcell) or ratIgG1 isotype control (Clone: HRPN BioXcell) at day -1 (1mg), day 1 (250ug), day 3 (500ug), day 6

(500ug), day 8 (250ug) and were sacrificed 10 days after immunization. For IL-6 neutralization 167 mice were injected i.p. with 300ug α-IL-6 (Clone: MP5-20F3, BioXcell) or 300ug isotype control (Clone: HRPN BioXcell) on days 0 and sacrificed on day 2. For ICOS-L neutralization old C57BL/6 mice were injected i.p. with 150µg anti-ICOSL (HK5.3, BioXcell) or with rat

IgG2A isotype control (2A3, BioXcell), on days 0, 3, 6, 9 and sacrificed on day 12.

In vivo cytokine capture assay and ELISAs: IL-6 and IL-10 in vivo cytokine capture assay was performed as previously described

(Finkelman et al., 2003) employing biotinylated capture antibodies (Invitrogen). In brief, young

(1.5–4 mo) and old (≥16 mo) C57BL/6 mice were injected i.v. with 10 ug biotinylated anti–IL-6

(MP5-32C11; Invitrogen) and anti–IL-10 (JES5-16E3: Invitrogen)) capture antibodies; mice were bled within 24 h and serum was collected. A luminescent ELISA was performed using anti–IL-6 (MP5-20F3; Invitrogen) or anti-IL-10 (JES5-2A5: BD Biosciences) as the coating antibody. For NP-specific antibody titers, 96-well plates were coated overnight at 4 °C with

NP30-BSA (Biosearch), followed by blockade of nonspecific biding by incubation for 1-2 h at

25 °C with 5% BSA. Serum samples were loaded into plates with eight serial dilutions

(starting from 1:100 or 1:1000), followed by incubation for 2 h at 25 °C or overnight at 4 °C.

After samples were washed, horseradish peroxidase (HRP)-conjugated goat antibody to mouse IgG1 (PA1-74421; Thermo) was added to plates, followed by incubation for 2 h at 25

°C. The reactions were developed by incubation for 15 min at 37 °C with 50 µl TMB substrate

(BioLegend) and were stopped by the addition of 25 µl 10% H3PO4. The plates were read at

450 nm and 570 nm (for correction) with an enzyme-linked immunosorbent assay reader.

RT-PCR:

Samples from different tissues were homogenized and total cellular RNA was extracted and quantified. DNase-treated RNA was then used to synthesize cDNA. The primer sequences used for detection of IL-10 were: 5′- GCTCTTACTGACTGGCATGAG -3′ and 5′-

168

CGCAGCTCTAGGAGCATGTG -3′. Expression levels were normalized to S14 as internal control gene. The primer sequences used for S14 detection were 5′- GAG GAG TCT GGA

GAC GAC GA-3′ and 5′- TGG CAG ACA CCA AAC ACA TT-3′. Quantitative real-time PCR was performed with Roche LightCycler 480 SYBRGreen 1 Master Mix using the Roche

LightCycler 480 II instrument (Roche Diagnostics). Each reaction was performed in triplicate.

Flow cytometry and cell sorting: Human studies Spleen cells from young (median: 18.8, range 18-26 yrs, 3 males, 5 females) and old

(median: 62, range 60-67 yrs, 4 males, 4 females) organ donors with no immunological condition were rested overnight in RPMI medium supplemented with 10% fetal calf serum

(FCS), 1% penicillin, streptomycin, glutamine and 0.5% HEPES, at 37o C and 5% CO2. The cells were then washed with PBS+2%FCS and stained for CD4, CXCR5, PD-1, CD45RO

(Biolegend), CD25 (BD Bioscience), CD3 (Invitrogen), and CD127 (Beckman Coulter) for 30 mins in 4o C, fixed with 4% para-formaldehyde for 20 mins in 4o C. Cells were stained for

Foxp3 (Invitrogen) using Invitrogen Foxp3 permeabilization buffer and acquired on a flow cytometer. For sorting, CD4+ T cells were bead-purified by negative selection from spleen cells, surface stained with Abs against CD45RO, CD127, CD25, PD-1, CXCR5 and the following populations were sorted by FACS after gating on memory CD4+ T cells (CD45RO+)

: Tfh (CD25-CD127+PD-1+CXCR5+), Treg (CD25+CD127-PD-1-CXCR5-) and non Tfh memory cells (CD25-CD127+PD-1-CXCR5-). 10,000 cells were stimulated in vitro with anti-

CD3/CD28 beads at a 1:1 cell: bead ratio, or unstimulated. After 16h, supernatants were collected and analyzed by Luminex.

Mouse studies Spleens were harvested and crushed through100-mm filters (BD Falcon) to generate single- cell suspensions. A total of 2X106 cells plated incubated with Fc block and were surface 169 stained with a combination of the following Abs for surface staining: -CD4, -CD8α, -TCRβ, -

LAG3, -Fas (BD Biosciences), -CD19, -PD1, -CXCR5, -GL-7, CD49B (Invitrogen), B220,

IgG1(Biolegend). Cells were intracellularly stained with antibodies against Bim (Cell Signaling

Technology), Ki67, Foxp3 (Invitrogen), BCL6 (BD Biosciences). For cytokine staining, cells were stimulated with 25ng/ml PMA and 0.5μg/ml ionomycin for 5 hours, in the presence of brefeldinA for the final 4 h and fixed with 2%methanol-free formaldehyde for 1 hour followed by intracellular staining for IL-10, IFN-γ (Biolegend), IL-17, IL-4 (Invitrogen) using Invitrogen

Foxp3 permeabilization buffer. For IL-21 staining, cells were fixed, permeabilized with perm buffer from Invitrogen and incubated with IL-21R/Fc (R&D systems) chimera for 45mins-1hr at

4°C. Cells were then washed with perm buffer and stained with AF488 or AF-647-conjugated affinity-purified F(ab')2 fragment of goat anti–human Fcγ antibody (Jackson ImmunoResearch

Laboratories) for 45-1hr at 4°C. Data were acquired on an LSRII flow cytometer (BD

Biosciences) and analyzed using FACSDiva software (BD Biosciences) or FlowJo software

(FlowJo, Ashland, OR). For sorting, spleen cells from young (3 mo, n=3) and old (≥18 mo, n=4) Foxp3-IRES-DTR-GFP mice were enriched for CD4+ T cells using the negative selection

MACS CD4+ T cell isolation kit II (Miltenyi Biotec, San Diego, CA). Enriched cells were stained with anti-CD4, -CD44, -CD62L Abs, and the following populations were sorted by a

FACSAria (BD Biosciences): CD4+ Foxp3GFP+ (Treg), CD4+ Foxp3-GFP- CD44lo CD62Lhi

(naive CD4+), CD4+ Foxp3-GFP- CD44hi CD62Llo (memory CD4+). All three populations were stimulated with 50ng/ml PMA and 1μg/ml ionomycin and supernatants were collected after 15 hours.

Single cell RNA-seq:

Young and old CD4+GFP+ CD25-CD69- cells were sorted and post sort analysis showed that

<1% of the sorted cells were FoxP3+. Single cells were isolated on a FLUIDIGM C1 170 instrument, cDNA isolated, amplified, barcoded, and subjected to high throughput sequencing.

Following transcript pseudoalingment and normalization using Kallisto in AltAnalyze, predominant gene expression identified from individual cells by ICGS. ICGS provides the sensitivity to detect rare and common cell populations as well as possible transitional cell states. Four major cell populations were reported along with statistically enriched Gene

Ontology terms for gene clusters.

Acknowledgements:

This work was supported by funds from the National Institute of Health Grant AG033057 (to

D.A.H. and C.A.C.). We thank members of the Hildeman and Chougnet laboratories (Sarah

Jones, Cyd Castro Rojas, Rachel Walters, Sharmila Shanmuganad, Kun-Po Li, Courtney

Jackson) for their suggestions and help with mouse experiments. The authors would also like to thank Drs. Jeremy M. Kinder and Sing Sing Way for their help with the FoxP3-fate mapping mice and Calvin Chan for his help with adipose tissue IL-10 gene studies.

REFERENCES

171

Akiba, H., Takeda, K., Kojima, Y., Usui, Y., Harada, N., Yamazaki, T., Ma, J., Tezuka, K.,

Yagita, H., and Okumura, K. (2005). The role of ICOS in the CXCR5+ follicular B helper T cell maintenance in vivo. J Immunol 175, 2340-2348.

Almanan, M., Raynor, J., Sholl, A., Wang, M., Chougnet, C., Cardin, R.D., and Hildeman, D.A.

(2017). Tissue-specific control of latent CMV reactivation by regulatory T cells. PLoS Pathog

13, e1006507.

Belkaid, Y., Hoffmann, K.F., Mendez, S., Kamhawi, S., Udey, M.C., Wynn, T.A., and Sacks,

D.L. (2001). The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure. J

Exp Med 194, 1497-1506.

Brooks, D.G., Trifilo, M.J., Edelmann, K.H., Teyton, L., McGavern, D.B., and Oldstone, M.B.

(2006). Interleukin-10 determines viral clearance or persistence in vivo. Nature medicine 12,

1301-1309.

Cai, G., Nie, X., Zhang, W., Wu, B., Lin, J., Wang, H., Jiang, C., and Shen, Q. (2012). A regulatory role for IL-10 receptor signaling in development and B cell help of T follicular helper cells in mice. J Immunol 189, 1294-1302.

Cauley, J.A., Barbour, K.E., Harrison, S.L., Cloonan, Y.K., Danielson, M.E., Ensrud, K.E.,

Fink, H.A., Orwoll, E.S., and Boudreau, R. (2016). Inflammatory Markers and the Risk of Hip and Vertebral Fractures in Men: the Osteoporotic Fractures in Men (MrOS). J Bone Miner Res

31, 2129-2138.

Choi, Y.S., Eto, D., Yang, J.A., Lao, C., and Crotty, S. (2013). Cutting edge: STAT1 is required for IL-6-mediated Bcl6 induction for early follicular helper cell differentiation. J

Immunol 190, 3049-3053.

172

Chougnet, C.A., Tripathi, P., Lages, C.S., Raynor, J., Sholl, A., Fink, P., Plas, D.R., and

Hildeman, D.A. (2011). A major role for Bim in regulatory T cell homeostasis. J Immunol 186,

156-163.

Corsini, E., Vismara, L., Lucchi, L., Viviani, B., Govoni, S., Galli, C.L., Marinovich, M., and

Racchi, M. (2006). High interleukin-10 production is associated with low antibody response to influenza vaccination in the elderly. J Leukoc Biol 80, 376-382.

Couper, K.N., Blount, D.G., and Riley, E.M. (2008). IL-10: the master regulator of immunity to infection. J Immunol 180, 5771-5777.

Crotty, S. (2011). Follicular helper CD4 T cells (TFH). Annual review of immunology 29, 621-

663.

Della Cioppa, G., Nicolay, U., Lindert, K., Leroux-Roels, G., Clement, F., Castellino, F., Galli,

G., Groth, N., and Del Giudice, G. (2012). Superior immunogenicity of seasonal influenza vaccines containing full dose of MF59 ((R)) adjuvant: results from a dose-finding clinical trial in older adults. Hum Vaccin Immunother 8, 216-227.

Dobber, R., Tielemans, M., and Nagelkerken, L. (1995). The in vivo effects of neutralizing antibodies against IFN-gamma, IL-4, or IL-10 on the humoral immune response in young and aged mice. Cellular immunology 160, 185-192.

Eto, D., Lao, C., DiToro, D., Barnett, B., Escobar, T.C., Kageyama, R., Yusuf, I., and Crotty,

S. (2011). IL-21 and IL-6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS One 6, e17739.

Finkelman, F., Morris, S., Orekhova, T., and Sehy, D. (2003). The in vivo cytokine capture assay for measurement of cytokine production in the mouse. Curr Protoc Immunol Chapter 6,

Unit 6 28.

Gagliani, N., Amezcua Vesely, M.C., Iseppon, A., Brockmann, L., Xu, H., Palm, N.W., de

Zoete, M.R., Licona-Limon, P., Paiva, R.S., Ching, T., et al. (2015). Th17 cells 173 transdifferentiate into regulatory T cells during resolution of inflammation. Nature 523, 221-

225.

Gagliani, N., Magnani, C.F., Huber, S., Gianolini, M.E., Pala, M., Licona-Limon, P., Guo, B.,

Herbert, D.R., Bulfone, A., Trentini, F., et al. (2013). Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nature medicine 19, 739-746.

Giunta, B., Fernandez, F., Nikolic, W.V., Obregon, D., Rrapo, E., Town, T., and Tan, J.

(2008). Inflammaging as a prodrome to Alzheimer's disease. J Neuroinflammation 5, 51.

Hanada, T., and Yoshimura, A. (2002). Regulation of cytokine signaling and inflammation.

Cytokine & growth factor reviews 13, 413-421.

Haynes, N.M., Allen, C.D., Lesley, R., Ansel, K.M., Killeen, N., and Cyster, J.G. (2007). Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation. J Immunol 179, 5099-5108.

Hirota, K., Duarte, J.H., Veldhoen, M., Hornsby, E., Li, Y., Cua, D.J., Ahlfors, H., Wilhelm, C.,

Tolaini, M., Menzel, U., et al. (2011). Fate mapping of IL-17-producing T cells in inflammatory responses. Nat Immunol 12, 255-263.

Hobbs, M.V., Weigle, W.O., and Ernst, D.N. (1994). Interleukin-10 production by splenic

CD4+ cells and cell subsets from young and old mice. Cellular immunology 154, 264-272.

Hollister, K., Kusam, S., Wu, H., Clegg, N., Mondal, A., Sawant, D.V., and Dent, A.L. (2013).

Insights into the role of Bcl6 in follicular Th cells using a new conditional mutant mouse model.

J Immunol 191, 3705-3711.

Jin, J.O., Han, X., and Yu, Q. (2013). Interleukin-6 induces the generation of IL-10-producing

Tr1 cells and suppresses autoimmune tissue inflammation. J Autoimmun 40, 28-44.

Johnston, R.J., Poholek, A.C., DiToro, D., Yusuf, I., Eto, D., Barnett, B., Dent, A.L., Craft, J., and Crotty, S. (2009). Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science 325, 1006-1010. 174

Kim, J.M., Rasmussen, J.P., and Rudensky, A.Y. (2007). Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol 8, 191-197.

Lefebvre, J.S., Lorenzo, E.C., Masters, A.R., Hopkins, J.W., Eaton, S.M., Smiley, S.T., and

Haynes, L. (2016a). Vaccine efficacy and T helper cell differentiation change with aging.

Oncotarget 7, 33581-33594.

Lefebvre, J.S., Masters, A.R., Hopkins, J.W., and Haynes, L. (2016b). Age-related impairment of humoral response to influenza is associated with changes in antigen specific T follicular helper cell responses. Sci Rep 6, 25051.

Levy, Y., and Brouet, J.C. (1994). Interleukin-10 prevents spontaneous death of germinal center B cells by induction of the bcl-2 protein. The Journal of clinical investigation 93, 424-

428.

Linterman, M.A., Beaton, L., Yu, D., Ramiscal, R.R., Srivastava, M., Hogan, J.J., Verma, N.K.,

Smyth, M.J., Rigby, R.J., and Vinuesa, C.G. (2010). IL-21 acts directly on B cells to regulate

Bcl-6 expression and germinal center responses. J Exp Med 207, 353-363.

Lio, D., Candore, G., Crivello, A., Scola, L., Colonna-Romano, G., Cavallone, L., Hoffmann,

E., Caruso, M., Licastro, F., Caldarera, C.M., et al. (2004). Opposite effects of interleukin 10 common gene polymorphisms in cardiovascular diseases and in successful ageing: genetic background of male centenarians is protective against coronary heart disease. J Med Genet

41, 790-794.

Lio, D., Scola, L., Crivello, A., Colonna-Romano, G., Candore, G., Bonafe, M., Cavallone, L.,

Franceschi, C., and Caruso, C. (2002). Gender-specific association between -1082 IL-10 promoter polymorphism and longevity. Genes Immun 3, 30-33.

Lord, J.M. (2013). The effect of ageing of the immune system on vaccination responses. Hum

Vaccin Immunother 9, 1364-1367.

175

Lustig, A., Liu, H.B., Metter, E.J., An, Y., Swaby, M.A., Elango, P., Ferrucci, L., Hodes, R.J., and Weng, N.P. (2017). Telomere Shortening, Inflammatory Cytokines, and Anti-

Cytomegalovirus Antibody Follow Distinct Age-Associated Trajectories in Humans. Frontiers in immunology 8, 1027.

Madan, R., Demircik, F., Surianarayanan, S., Allen, J.L., Divanovic, S., Trompette, A., Yogev,

N., Gu, Y., Khodoun, M., Hildeman, D., et al. (2009). Nonredundant roles for B cell-derived IL-

10 in immune counter-regulation. J Immunol 183, 2312-2320.

Madisen, L., Zwingman, T.A., Sunkin, S.M., Oh, S.W., Zariwala, H.A., Gu, H., Ng, L.L.,

Palmiter, R.D., Hawrylycz, M.J., Jones, A.R., et al. (2010). A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci 13, 133-140.

Maggio, M., Guralnik, J.M., Longo, D.L., and Ferrucci, L. (2006). Interleukin-6 in aging and chronic disease: a magnificent pathway. The journals of gerontology. Series A, Biological sciences and medical sciences 61, 575-584.

Maue, A.C., Eaton, S.M., Lanthier, P.A., Sweet, K.B., Blumerman, S.L., and Haynes, L.

(2009). Proinflammatory adjuvants enhance the cognate helper activity of aged CD4 T cells. J

Immunol 182, 6129-6135.

Mazmanian, S.K., Round, J.L., and Kasper, D.L. (2008). A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453, 620-625.

McElhaney, J.E., Kuchel, G.A., Zhou, X., Swain, S.L., and Haynes, L. (2016). T-Cell Immunity to Influenza in Older Adults: A Pathophysiological Framework for Development of More

Effective Vaccines. Frontiers in immunology 7, 41.

McElhaney, J.E., Xie, D., Hager, W.D., Barry, M.B., Wang, Y., Kleppinger, A., Ewen, C.,

Kane, K.P., and Bleackley, R.C. (2006). T cell responses are better correlates of vaccine protection in the elderly. J Immunol 176, 6333-6339.

176

McKinstry, K.K., Strutt, T.M., Buck, A., Curtis, J.D., Dibble, J.P., Huston, G., Tighe, M.,

Hamada, H., Sell, S., Dutton, R.W., and Swain, S.L. (2009). IL-10 deficiency unleashes an influenza-specific Th17 response and enhances survival against high-dose challenge. J

Immunol 182, 7353-7363.

Moore, K.W., de Waal Malefyt, R., Coffman, R.L., and O'Garra, A. (2001). Interleukin-10 and the interleukin-10 receptor. Annual review of immunology 19, 683-765.

Neumann, C., Heinrich, F., Neumann, K., Junghans, V., Mashreghi, M.F., Ahlers, J., Janke,

M., Rudolph, C., Mockel-Tenbrinck, N., Kuhl, A.A., et al. (2014). Role of Blimp-1 in programing Th effector cells into IL-10 producers. J Exp Med 211, 1807-1819.

North, B.J., and Sinclair, D.A. (2012). The intersection between aging and cardiovascular disease. Circ Res 110, 1097-1108.

Nurieva, R., Yang, X.O., Martinez, G., Zhang, Y., Panopoulos, A.D., Ma, L., Schluns, K., Tian,

Q., Watowich, S.S., Jetten, A.M., and Dong, C. (2007). Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature 448, 480-483.

Nurieva, R.I., Chung, Y., Hwang, D., Yang, X.O., Kang, H.S., Ma, L., Wang, Y.H., Watowich,

S.S., Jetten, A.M., Tian, Q., and Dong, C. (2008). Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity 29,

138-149.

Odamaki, T., Kato, K., Sugahara, H., Hashikura, N., Takahashi, S., Xiao, J.Z., Abe, F., and

Osawa, R. (2016). Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol 16, 90.

Raynor, J., Lages, C.S., Shehata, H., Hildeman, D.A., and Chougnet, C.A. (2012).

Homeostasis and function of regulatory T cells in aging. Curr Opin Immunol 24, 482-487.

177

Rochman, Y., Dienger-Stambaugh, K., Richgels, P.K., Lewkowich, I.P., Kartashov, A.V.,

Barski, A., Khurana Hershey, G.K., Leonard, W.J., and Singh, H. (2018). TSLP signaling in

CD4(+) T cells programs a pathogenic T helper 2 cell state. Sci Signal 11.

Roncarolo, M.G., Gregori, S., Bacchetta, R., and Battaglia, M. (2014). Tr1 cells and the counter-regulation of immunity: natural mechanisms and therapeutic applications. Curr Top

Microbiol Immunol 380, 39-68.

Round, J.L., and Mazmanian, S.K. (2010). Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A 107, 12204-

12209.

Sage, P.T., Tan, C.L., Freeman, G.J., Haigis, M., and Sharpe, A.H. (2015). Defective TFH

Cell Function and Increased TFR Cells Contribute to Defective Antibody Production in Aging.

Cell Rep 12, 163-171.

Thevaranjan, N., Puchta, A., Schulz, C., Naidoo, A., Szamosi, J.C., Verschoor, C.P., Loukov,

D., Schenck, L.P., Jury, J., Foley, K.P., et al. (2017). Age-Associated Microbial Dysbiosis

Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell

Host Microbe 21, 455-466 e454.

Tsukamoto, H., Huston, G.E., Dibble, J., Duso, D.K., and Swain, S.L. (2010). Bim dictates naive CD4 T cell lifespan and the development of age-associated functional defects. J

Immunol 185, 4535-4544.

Tsukamoto, H., Senju, S., Matsumura, K., Swain, S.L., and Nishimura, Y. (2015). IL-6- mediated environmental conditioning of defective Th1 differentiation dampens antitumour immune responses in old age. Nat Commun 6, 6702. van der Geest, K.S., Lorencetti, P.G., Abdulahad, W.H., Horst, G., Huitema, M., Roozendaal,

C., Kroesen, B.J., Brouwer, E., and Boots, A.M. (2016). Aging-dependent decline of IL-10

178 producing B cells coincides with production of antinuclear antibodies but not rheumatoid factors. Experimental gerontology 75, 24-29.

Vogelzang, A., McGuire, H.M., Yu, D., Sprent, J., Mackay, C.R., and King, C. (2008). A fundamental role for interleukin-21 in the generation of T follicular helper cells. Immunity 29,

127-137.

Volpato, S., Guralnik, J.M., Ferrucci, L., Balfour, J., Chaves, P., Fried, L.P., and Harris, T.B.

(2001). Cardiovascular disease, interleukin-6, and risk of mortality in older women: the women's health and aging study. Circulation 103, 947-953.

Wang, Z.Y., Sato, H., Kusam, S., Sehra, S., Toney, L.M., and Dent, A.L. (2005). Regulation of

IL-10 gene expression in Th2 cells by Jun proteins. Journal of immunology 174, 2098-2105.

Xie, M.M., Koh, B.H., Hollister, K., Wu, H., Sun, J., Kaplan, M.H., and Dent, A.L. (2017). Bcl6 promotes follicular helper T-cell differentiation and PD-1 expression in a Blimp1-independent manner in mice. Eur J Immunol 47, 1136-1141.

Yu, D., Rao, S., Tsai, L.M., Lee, S.K., He, Y., Sutcliffe, E.L., Srivastava, M., Linterman, M.,

Zheng, L., Simpson, N., et al. (2009). The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity 31, 457-468.

Zhang, X., Meng, X., Chen, Y., Leng, S.X., and Zhang, H. (2017). The Biology of Aging and

Cancer: Frailty, Inflammation, and Immunity. Cancer J 23, 201-205.

Zhou, X., Jeker, L.T., Fife, B.T., Zhu, S., Anderson, M.S., McManus, M.T., and Bluestone, J.A.

(2008). Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. J Exp

Med 205, 1983-1991.

179

Figures

Figure 1. Aged mice have increased systemic levels of IL-10. (A) Young (white bar, n=6) and old

(black bar, n=5) C57BL/6 mice were i.v. injected with biotinylated anti–IL-10 capturing Abs. Serum was collected 24 h later, and IL-10 levels were measured by ELISA. Graph shows the average serum IL-10

(mean±SEM). Data are representative of at least two independent experiments. (B) IL-10 mRNA gene expression was measured by real-time RT-PCR on cDNA isolated from the spleen, liver, gut, lymph nodes, (inguinal, epididymal) white and brown adipose tissue, from individual young (n=4-8) and old

(n=5-9) mice. Graph shows the mean fold change in IL-10 mRNA expression calculated by dividing the individual expression level in old mice by the average expression level of the young mice

(mean±SEM). Dashed line represents equal level of expression in young and old mice. Data pooled from two independent experiments. *p ≤0.05, **p ≤0.01, ***p ≤0.001, ****p ≤0.0001, Student’s t-test.

180 Figure 2. CD4+FoxP3- T cells are the major source of IL-10 in aged mice. (A) Splenocytes from young (n=3) and old (n=5) IL-10gfp (Vertex) mice were stained with Abs against CD4, CD8, TCRβ and

CD19 and analyzed by flow cytometry. The representative plots show the gating strategy and

181 frequencies of CD4+, CD8+, CD19+ and CD19- that are GFP+. Graphs show the total number (upper) and frequency (middle) of cells that are GFP+ in young (white bar) and old (black bar) mice

(mean±SEM). The lower graph shows the level of GFP expression in old CD4+, CD8+, CD19+ and

CD19- that are GFP+ (mean±SEM). **p ≤0.01, Student’s t-test. Data are representative of at least two independent experiments. (B) Splenocytes from young (3mo, n=4) and old (≥18mo, n=4) FoxP3-IRES-

DTR-GFP mice were enriched for CD4+ cells using CD4 T cell isolation kit (Miltenyi) and stained with

Abs against CD4, CD62L and CD44. CD4+FoxP3GFP+, CD4+FoxP3GFP− CD44hi CD62Llo (memory) and

CD4+FoxP3GFP− CD44lo CD62Lhi (naïve) cells were FACS sorted. Purified cells (3 × 105) were stimulated with PMA and Ionomycin (P+I) for 15hrs and IL-10 levels in culture supernatants quantified by ELISA. Graph shows the average IL-10 levels (mean±SEM). Data pooled from two independent experiments. (C) Splenocytes from young (n=4) and old (n=4) C57BL/6 mice were stimulated with

(P+I), stained with Abs against TCRβ, CD8, Foxp3 and IL-10, and assessed for cytokine production by flow cytometry. The representative plots show the gating strategy and frequencies of Foxp3+ or

Foxp3- that are IL-10+ from young or old mice. Graphs show the frequency and the total number of cells that are IL-10+ in young (white bar) and old (black bar) mice (mean±SEM). (D) Young (n=6) and old (n=14) C57BL/6 mice were treated with a single dose (600 μg) of anti-CD4 depleting Ab or isotype control (n=16) at d0. Old Foxp3-DTR C57BL/6 mice (n=6) were treated with a single dose (1.25μg) of

Diphtheria Toxin (DT). Mice were injected i.v. with biotinylated α-IL-10 capturing Ab at d1. Serum was collected at d2 to measure IL-10 levels by ELISA. Graph shows the average serum IL10 levels

(mean±SEM) and representative data pooled from two independent experiments. (E) Splenocytes from isotype control old mice (n=8) and old Foxp3DTR C57BL/6 mice treated with a single dose

(1.25μg) of Diphtheria Toxin (DT) (n=6) were stimulated as above (C), stained with Abs against TCRβ,

CD8, Foxp3 and IL-10 and analyzed by flow cytometry. Graph shows the frequency of Foxp3- cells that are IL-10+ (mean±SEM). *p ≤0.05, **p ≤0.01, ***p ≤0.001, Student’s t-test.

182 Figure 3. IL-10-producing FoxP3- CD4+ T cells in aged mice are predominantly Tfh cells. (A)

Splenocytes from young (n=6) and old (n=6) C57BL/6 mice were stimulated with (P+I), stained with

Abs against TCRβ, CD8, Foxp3, IL-10, IL-21 and analyzed by flow cytometry. The representative histograms and graphs show the frequencies and total numbers of IL-21+ cells within Foxp3- that are

IL-10+ (mean±SEM). *p ≤0.05, **p ≤ 0.01, Student’s t-test. Data are representative of at least two independent experiments. (B) Splenocytes from young (n=4) and old (n=4) C57BL/6 mice were stimulated as above and stained with Abs against TCRβ, CD8, Foxp3, CXCR5, PD-1 and IL-10, and analyzed by flow cytometry. The representative plots and graphs show the frequencies and total numbers of indicated subsets within Foxp3- that are IL-10+ (mean±SEM). *p ≤0.05, **p ≤ 0.01,

Student’s t-test. Data are representative of at least two independent experiments.

183 Figure 4. IL-6 is required for Tfh10 cells and for systemic levels of IL-10 in aged mice. (A, B)

Splenocytes from young and old C57BL/6 or IL-6-/- mice (n≥4/group) were stimulated with (P+I), stained with Ab against TCRß, CD8, CXCR5, PD1, Foxp3 and IL-10, and analyzed by flow cytometry.

184 The representative plots and bar graphs show the frequency and total number of Foxp3- that CXCR5+

PD1+ and their IL-10 production (mean±SEM). (C) Old C57BL/6 (n=8) and IL-6-/- (n=8) mice were i.v. injected with biotinylated anti–IL-10 Abs, serum was collected 24 hrs later, and IL-10 levels were measured by ELISA. Graph shows the average serum IL-10 (mean±SEM). *p ≤0.05, **p ≤0.01,

Student’s t-test. (D) Old C57BL/6 (19mo) mice were treated with isotype control (n=6) or α-IL-6 blocking antibody (n=8) on day0 and sacrificed on day2. Splenocytes were stimulated with (P+I), stained with Ab against TCRß, CD8, Foxp3 and IL-10 and analyzed with flow cytometry. The representative bar graph shows the frequency of Foxp3- that are IL-10+ (mean±SEM).

185 186 Figure 5. IL-21 contributes to accrual of Tfh10 cells and regulates the systemic IL-6/IL-10 balance. (A) Splenocytes from young and old C57BL/6 or IL-21-/- mice (n≥4/group) were stimulated with (P+I), stained with Ab against TCRß, CD8, Foxp3 and IL-21 and analyzed by flow cytometry. Bar graphs show the frequency and total number of Foxp3- that are IL-21+ (mean±SEM). (B, C)

Splenocytes from old C57BL/6 or IL-21-/- mice (n≥3/group) were stimulated as above, stained with Ab against TCRß, CD8, CXCR5, PD1, Foxp3 and IL-10, and analyzed with flow cytometry. The representative plots and bar graphs show the frequency and total number of Foxp3- CXCR5+ PD1+ and their IL-10 production (mean±SEM). Data are pooled from two independent experiments. (D, E)

Old C57BL/6 (n=4) and old IL-21-/- (n=3) mice were i.v. injected with biotinylated anti–IL-10 and anti-IL-

6 capturing Abs, serum was collected 24 h later, and IL-10 and IL-6 levels were measured by ELISA.

Graphs show the average serum IL-10 and IL-6 (mean±SEM)

187 Figure 6. IL-21 driven repression of Bim in aged Tfh10 cells results in their enhanced survival.

Splenocytes from young (n=4) and old (n=4) mice were stimulated with (P+I), stained with Abs against

TCRß, CD8, Foxp3, IL-10, Ki67, Bim and analyzed by flow cytometry. (A) Graph shows the frequency

188 of Foxp3- IL-10+ cells that are Ki67+( mean±SEM). (B) Graph shows the level of expression of Bim in

Foxp3- IL-10+ cells (mean±SEM). (C) Splenocytes from 6month old wild-type and Bim-/- mice

(n=6/group) were stimulated as above, stained with Ab against TCRß, CD4, Foxp3 and IL-10 and analyzed by flow cytometry. Plots and bar graphs show the frequency and total number of Foxp3- that are IL-10+ (mean±SEM). ). (D) Splenocytes from C57BL/6 or IL-21-/- mice (n≥3/group) were stimulated as above, stained with Ab against TCRß, CD8, Foxp3, IL-10 and Bim and analyzed with flow cytometry. Graph shows the level of expression of Bim in Foxp3- that are IL-10+ cells (mean±SEM). *p

≤0.05, **p ≤0.01, ***p ≤0.001, Student’s t-test. *p ≤0.05, **p ≤ 0.01, Student’s t-test.

189 Figure 7. Tfh10 cells in aged mice manifest diminished levels of BCL6 thereby enabling IL-10 expression. (A) Splenocytes from young and old C57BL/6 mice (n=4/group) were stimulated with

(P+I), stained with Ab against, CD8, CXCR5, PD1, Foxp3 and IL-10, and analyzed with flow cytometry.

The representative bar graphs show the level of expression of BCL6 and IL-10 in Foxp3- CXCR5+ PD-

1+ that are IL-10+ (mean±SEM). (B) Splenocytes from middle age wild-type or CD4Cre BCL6f/f mice

(n≥3/group) were stained with Ab against CXCR5, PD1and Foxp3. The representative plot and bar graph show the frequency of Foxp3- cells that are CXCR5+ PD1+ (mean±SEM). (C) Splenocytes from middle age wild-type or CD4Cre BCL6f/f mice (n≥3/group) were stimulated as above (A), stained with

Abs against TCRβ, CD8, Foxp3 and IL-10 and analyzed with flow cytometry. The representative histograms and bar graphs show the frequency and total number of Foxp3- cells that are IL-10+

(mean±SEM). *p ≤0.05, **p ≤0.01, Student’s t-test.

190 Figure 8. IL-10 limits Tfh-dependent vaccine responses in aged mice. (A) Young (n=6) and old

(n=5) mice were immunized with NP-KLH in Alum and sacrificed 20 days later. Splenocytes were stained with Abs against CD19, B220, GL7 and Fas and analyzed by flow cytometry. Representative

191 plots identifying GC B cells NP-specific as Fashi GL7hi that are IgG1+ NP tetramer+. Graphs show the frequency and the total number of splenic B cells that are IgG1+ NP+ (mean±SEM), as well as serum levels of immunoglobulin specific for NP (IgG1) of young vs old mice obtained 20 days after immunization (mean±SEM). (B) Mice were immunized as above d0 and then treated with isotype (n=7) or anti–IL-10R neutralizing Ab (n=8) on day -1, 1, 3, 6, 9 and sacrificed on day10. Representative plots display the frequency of GC B cells (NP-specific). Graphs show the frequency and the total number of

B cells that are IgG1+ NP+ (mean±SEM), as well as serum levels of immunoglobulin specific for NP

(IgG1) in mice with or without IL-10R neutralization obtained 10 days after immunization (mean±SEM).

*p ≤0.05, **p ≤ 0.01, Student’s t-test.

192 Figure 9. Tfh10 cells accumulate during aging in humans. Human spleen cells from young (n=8) and old (n=8) individuals were surface stained with Abs against CD3, CD4, CD45RO, CXCR5, PD-1 and Foxp3 and analyzed with flow cytometry. (A) Graph shows the frequency of CD3+ CD4+

CD45RO+ Foxp3- that are CXCR5+ PD-1+ (mean±SEM). (B, C) CD4+ T cells were bead-purified by negative selection, FACS-sorted memory CD4+ T cells (CD45RO+) into Tfh (CD25-CD127+PD-

1+CXCR5+), Treg (CD25+CD127-PD-1-CXCR5-) and other memory cells (CD25-CD127+PD-1-CXCR5-).

10,000 cells were stimulated in vitro with anti-CD3/CD28 beads at a 1:1 cell: bead ratio, or unstimulated. After 16h, supernatants were collected and analyzed by Luminex. Cytokines were undetectable in unstimulated cultures. Each individual is represented by a symbol, Y (young), O (old)

(mean±SEM). ***p ≤0.005, Student’s t-test.

193 *** 8000 Young *** O ld 6000

4000 IL-10 MFI IL-10 2000

0 FOXP3+ FOXP3-

Supplementary Fig.1. Splenocytes from young (n=4) and old (n=4) C57BL/6 mice were stimulated with (P+I), stained with Abs against TCRβ, CD8, Foxp3 and IL-10 and analyzed by flow cytometry.

Graph shows the mean level of IL-10 in Foxp3+ IL-10+ and Foxp3- IL-10+ cells (mean±SEM). ***p ≤

0.001, Student’s t-test.

Supplementary Fig.2. Splenocytes from microbiota-replete conventionally-housed (Con) (n=2-

4/group) or germ free (GF) (n=2-5/group) mice were stimulated with (P+I), stained with Abs against

TCRβ, CD8, Foxp3 and IL-10 and analyzed by flow cytometry. Graph shows frequency of Foxp3- cells that are IL-10+ (mean±SEM). Data pooled from 3 independent experiments. 194 Supplementary Fig.3. (A) Splenocytes from young (n=8) and old (n=8) mice were stimulated with

(P+I), stained with Abs against TCRβ, CD8, CD49B, LAG3 and IL-10, and analyzed by flow cytometry.

The plots and graph show the frequency of indicated subsets in Foxp3- IL-10+ cells (mean±SEM). (B)

Splenocytes from young (n =4) and old (n =4) mice were stimulated as above, stained with Abs 195 against TCRβ, CD8, IL-10, IL-17, IL-4 and analyzed by flow cytometry. The plots and graph show the frequency of indicated subsets in Foxp3- cells (mean±SEM). (C) Spleen cells from young (2mo, n=5) and middle-age (12mo, n=5) IL-17Acre Rosa26 YFP (R26YFP) mice were stimulated as above, stained with Abs against TCRβ, CD8, IL-10, IL-17 and analyzed with flow cytometry. Bar graph shows the

(mean±SEM), within the total FOXP3- IL-10+ cells, the frequency of cells producing IL-17 (IL-17+YFP+, gray), exTh17 (IL-17-YFP+, white) or those that never produced IL- 17 (IL-17-YFP-, black). (D) Spleen cells from middle-age (12mo, n=4) and young (6weeks, n=3) Foxp3CreRosa26dTomato mice were stimulated with (P+I), stained with Abs against TCRβ, CD8, IL-10 and Foxp3, and analyzed with flow cytometry. Plots and bar graph show, within the total Foxp3- IL-10+ cells, the frequency of exTreg cells

(Foxp3- IL-10+ dTomato+). Data pooled from two independent experiments (mean±SEM).

196 Supplementary Fig.4. Splenocytes from young, middle age and old C57BL/6 mice (n≥4/group) were stimulated with (P+I) and stained with Ab against TCRß, CD8, CXCR5, PD1, Foxp3 and IL-10. The representative bar graphs show the frequency of Foxp3- that CXCR5+ PD1+ and those that produce

IL-10 (mean±SEM). *p ≤0.05, **p ≤0.01, Student’s t-test.

197 Supplementary Fig.5. Old C57BL/6 mice were treated with isotype control (n=6) or α-ICOSL blocking antibody (n=5) on day (0, 3, 6, 9) and sacrificed on day 12. Splenocytes were stimulated with (P+I), stained with Ab against TCRß, CD4, CD8, CXCR5, PD1, Foxp3 and IL-10, and analyzed by flow cytometry. The representative plots and bar graphs show the frequency of Foxp3- that CXCR5+ PD1+ and the frequency of Foxp3- that IL-10+ (mean±SEM). 198 1200

900

600

300

IL-10+ BLIMP-1 MFIIL-10+ 0 WT BCL6 cKO

Supplementary Fig.6. Splenocytes from middle age wild-type or CD4Cre Bcl-6f/f mice (n≥3/group) were stimulated with P+I and stained with Abs against TCRβ, CD8, Foxp3 and IL-10 and Blimp-1 and analyzed by flow cytometry. Graph shows the mean fluorescence intensity (MFI) for Blimp-1 in IL-10+

Foxp3- cells (mean±SEM).

199 Chapter 5

Summary

The world is experiencing an extraordinary increase in elderly population. The average life expectancy will reach 84 years old by 2050 compared to 68 years old in 1950 (1).

Such an increase in longevity will have a huge impact on health care system. A significant part of this burden on health care is the age-driven loss of immune competence (2-6). Indeed, aging is associated with quantitative and qualitative defects in T and B cells resulting in adverse health outcomes and an increase in the susceptibility to acquire infections and a failure to respond to vaccines (7-10).

Another feature of the dysregulated immune responses with age is the emergence of chronic low-level inflammation termed inflammaging (11-13). Inflammaging is thought to be a major driver for serious age-related diseases, such as atherosclerosis, cardiovascular diseases, Alzheimer's disease, rheumatoid arthritis, cancer, and frailty

(11, 12). One major focus in the field of aging is to address possible compensatory regulatory mechanisms that can combat this inflammation with age favoring reduction in age related pathologies and improving the quality of life for elderly.

Human CMV (HCMV) is a ubiquitous infection, infecting 40–90% of world’s population

(14). HCMV is never cleared and persists in human hosts for life (15). Such persistence is thought to accelerate age-related immune dysregulation and inflammaging with a detrimental effect on host immunity (16). Understanding the interplay between aging, latent CMV and the mechanisms regulating its persistence and reactivation might

200 provide novel interventions to control the adverse effects of age-related CMV infection.

In chapter 3 we aimed at dissecting the regulatory mechanisms that shaped the immune

responses towards latent CMV. Surprisingly, we found that the relationship between

CD4+ Foxp3- IL-10 producing T cells and regulatory Foxp3+ T (Treg) cells was more

complex and intertwined than previously appreciated. Additionally, the role of Treg and

Foxp3- IL-10 producing cells was highly integrated resulting in differential control of

CMV reactivation/replication in the spleen and salivary glands. A brief summary of our

findings from chapter 3 and potential future directions for this work will be addressed in

the next section.

Inflammaging is characterized by an increase in plasma levels of pro-inflammatory

cytokines such as Interleukin-6 (IL-6) (17), which is a major predictor of mortality and

morbidity with age (18). Given our findings with IL-10 and control of latent MCMV

reactivation/replication, we reasoned that the production of anti-inflammatory cytokines could be a potential mechanism to counterbalance such inflammaging. Indeed, we observed that increased production of IL-6 was counterbalanced by a concomitant

production of large quantities of IL-10 with age. To further understand the role of IL-10

with age, we investigated its cellular sources, the molecular mechanisms driving its

production and its role in regulating immune responses with age. A summary of our

findings from chapter 4 and future directions will be addressed in the next section.

201

Major findings and future directions

Chapter 3

1- Foxp3+ Treg cells play differential roles in the spleen and salivary glands during latent MCMV infection.

Following primary infection with MCMV, the adaptive immune response clears most replicating virus, however the virus establishes latency within multiple tissues (19). CD8

T cells are critical for controlling both acute and latent MCMV infection (20). Indeed,

persistent MCMV infection in the spleen is known to induce the formation of activated, highly differentiated memory CD8 T cells (21). However, the regulatory mechanisms controlling CD8 T cells memory responses to MCMV and their potential contribution to controlling the latent viral pool are not clear. Our data provided evidence that, CD8 T cells are negatively regulated by Foxp3+ Treg cells. Depletion of Treg cells resulted in enhanced effector CD8 T cell responses in the spleen and a reduction in latent viral pool. The data clearly highlight an unappreciated in vivo role for regulatory T cells during latent MCMV.

Interestingly, while Treg depletion resulted in reduced latent viral pool in the spleen, it enhanced viral reactivation/replication in the salivary gland (SG). The SG represents a major site of cytomegalovirus replication and transmission (22). Furthermore, the production of IL-10 from Foxp3- CD4+ T cells attenuated antiviral responses and promoted viral replication in the SG (23). Interestingly, the reactivation of MCMV in the

SG was accompanied by the emergence of IL-10+ Foxp3- T cells. The data implicates

IL-10 from Foxp3- cells in viral replication/reactivation.

202

Notably, TNF-α, is an important cytokine in the control of MCMV reactivation/replication

(24, 25). Indeed, we observed significant increases in the expression of TNF-α in the

SG in the absence of Treg cells. Thus, our data is consistent with a scenario in which

IL-10+ Foxp3- cells and their production of IL-10 might enhance viral replication following TNF-α mediated MCMV reactivation.

While we saw an increase in viral reactivation/replication in the SG we observed reduced viral load in the spleen. One question that arose from our results is why the viral load in the spleen was reduced despite the elevated levels of IL-10-producing

CD4+ T cells in that organ? A possible explanation for this scenario is that upon Treg depletion expansion of MCMV-specific cytotoxic CD8 T cells could detect and kill cells harboring this reactivating virus. In contrast to the spleen, CD8 T cells are incapable of reducing virus in the SG due to loss of expression of MHC class I molecules in virally- infected cells in that organ.

These data also raise the question regarding the mechanism by which regulatory T cells are controlling CD8 T cells in the spleen. One possibility is that Treg might utilize TGF-ß,

IL-35 or CTLA-4 to suppress CD8 T cells (26). One way to test this possibility is through the individual neutralization of potential regulatory factors (Table1) in groups of MCMV latently infected mice. One group of control mice would undergo Treg depletion, while another control group would be injected with an isotype control antibody. The spleen

PCR for viral load and an explant assay would be utilized to examine the reactivation of the virus and flow cytometry will be utilized to investigate the levels of antigen- specific

CD8 T cells and their effector responses (cytokine production and cytotoxicity).

203

Table 1

Regulatory molecule Neutralizing Ab

IL-35 anti-IL-35

TGF-ß anti-TGF-ß

CTLA-4 CTLA-4Ig

IL-35/CTLA-4/TGF-ß anti-IL-35/ anti-TGF-ß/ CTLA-4Ig

It is possible that we would not see an effect of the neutralization of the regulatory factors separately relative to Treg depleted mice. For example, Treg depleted mice would experience enhanced CD8 T cell responses and reduction in viral load in the spleen. However, infected mice with neutralization of (e.g. IL-35) would show no effect on CD8 T cells and viral load. This result would suggest possible redundancy in the regulatory mechanisms utilized by Treg cells in controlling CD8+ T cell responses. Such possibility would require testing by combined neutralization of all three potential mechanisms (Table 1) which might be the best way to recapitulate the in vivo effect that we see with total depletion of Treg cells. One caveat that might arise from the experiment in the spleen is the maintenance of the neutralization effect ex vivo in the spleen explants. However, we expect that if single neutralization of one regulatory marker or all of them was efficient to enhance the CD8 T cells responses in vivo then spleen PCR would show significant reduction in viral load between neutralized mice compared to the control group which would reflect reduction in latent viral pool and hence delayed viral reactivation in spleen explants as well.

204

2- IL-10 is critical for MCMV replication in the salivary gland (SG).

Neutralization of IL-10R resulted in reduction in reactivation/replication of virus in the

SG after depletion of Treg. However, we did not distinguish whether the effects of IL-

10R blockade were to limit outright viral reactivation or to limit viral replication.

Nonetheless, our data are consistent with IL-10 promoting viral replication more than outright viral reactivation as three key pieces of data suggest that IL-10 is not involved in direct viral reactivation. First, IL-10 has been shown to regulate viral replication during primary infection rather than reactivation (27). Second, the neutralization of IL-10R was done two days after Treg depletion. Thus, it is possible that reactivation of the virus has already been established before the treatment. Lastly, we attempted to induce viral reactivation in our in vitro explant assay and found that the addition of IL-10 to the assay was not sufficient to exacerbate viral reactivation. While we cannot conclusively rule out the role of IL-10 in viral reactivation, the data suggests the presence of other stimuli driving viral reactivation in the absence of Treg cells.

Interestingly, we found an increase in TNF-α after Treg depletion in the SG. Notably, prior in vitro studies have shown a pivotal role for inflammatory cytokines, specifically of

TNF-α in the reactivation of latent MCMV via the activation of MIE enhancer (24, 28).

Thus, one possibility is that TNF-α can induce the first stage of reactivation followed by

IL-10 enhancing the replication. We hypothesize that TNF-α is controlling viral reactivation in the SG upon Treg depletion. Interestingly, our preliminary data neutralizing TNF-α showed a significant reduction in SG viral titers (Figure1), consistent with the concept that TNF-α is controlling viral reactivation. The results from TNF-α neutralization and IL-10R neutralization experiments support a scenario where Treg

205

depletion would lead to the production TNF-α, the activation of MIE enhancer and

subsequently viral reactivation. This would constitute the first checkpoint, while

simultaneous IL-10 production upon Treg depletion provides a second checkpoint

resulting in further enhancement of viral replication. Notably, the neutralization of TNF-α

in latent MCMV infected mice was done simultaneously with Treg depletion, while IL-

10R neutralization was done after two days of Treg depletion. This might also support

our proposed scenario that TNF-α controlled the reactivation of the virus which is happening very early once Treg are depleted while IL-10 controlled the replication after two days of reactivation.

We propose to investigate the role of TNF-α in MCMV reactivation in the SG through a series of experiments, some of which involve the tissue explant reactivation assay.

Notably, the explant assay is a functional assay to assess reactivation from latency. If the virus is typically replicating in the tissue at the time of isolation, it will be detected by seven days post explants. However, if virus is latent in the tissue at the time of isolation, it will take longer and usually will reactivate after day7. We anticipate that SG tissue

cultured with TNF-α will exacerbate the reactivation and therefore result in increased

viral production relative to culture in the absence of TNF-α. Such results would provide

strong evidence that TNF-α is controlling reactivation rather than replication of the virus.

However, if we see no evidence for differences in the reactivation between the cultures

with and without TNF-α, this would suggest that either TNF-α is not involved in

reactivation per se and is either redundant with other factors controlling reactivation or

TNF-α contributes to viral replication in the SG upon Treg depletion.

206

Major findings and future directions

Chapter4

1- A novel population of Tfh cells accumulates with age and is the dominant

source of IL-10 in aged mice.

In chapter 4 we describe a novel population of T follicular helper cells that accumulated

in aged mice and produced IL-10 (Tfh10). In agreement with previous reports

supporting the role of IL-21 and IL-6 in Tfh differentiation (29, 30), we found that both IL-

6 and IL-21 were required for the accumulation of Tfh10 cells with age. Notably, we did

not observe any change in Tfh10 cells following IL-6 neutralization which suggested that

IL-21 is required for their maintenance rather than IL-6. Indeed, previous reports have

shown that IL-6 can be the first inducer for IL-21 production from Tfh cells and IL-21 is

supporting their maintenance (31) (32). Our data also suggested that IL-21 regulated

the survival of Tfh10 cells via down regulation of Bim. However, the detailed molecular

mechanism(s) underlying the survival effect under IL-21/Bim axis were still not clear.

Mechanistically, IL-21 signaling may promote Tfh survival through activation of the

PI3K/Akt/Foxo3a axis which plays a major role in negatively regulating Bim expression

(33, 34).

Indeed, a previous study showed that the direct binding of IL-21 to its receptor on T cells leads to the activation of the PI3K signaling pathway and increased cell survival

(35). However, the authors attributed the IL-21 driven survival to the up-regulation of the survival molecule BCL-2 and the effect of IL-21/PI3K axis on Bim levels was not

207

addressed. To investigate the role of IL-21/Bim axis in the maintenance and survival of

Tfh10 cells we would propose several experiments.

To assess whether IL-21 is critical for the maintenance/survival of Tfh10 cells, we would

block IL-21R signaling in vivo using an IL-21R neutralizing antibody. We expect to see

lower levels of Tfh10 cells in aged IL-21R neutralized compared to isotype mice. We

would also expect to see higher levels of Bim in the remaining Tfh10 cells upon IL-21R

neutralization. The data would suggest that IL-21 is required to control levels of Bim in

Tfh10 cells and their homeostasis. Second, we would breed Bim-/- to IL-21-/- mice and assess their phenotype. We expect to see restoration in Tfh10 cells compared to their loss in Bim+/+ IL-21-/- mice. The results would further support the role of Bim in IL-21-

driven Tfh10 survival.

Third, to test the role of IL-21/PI3K/Bim in the survival of Tfh10 cells we would sort old

Tfh10 cells (CXCR5+ PD1+ Foxp3 RFP- IL-10 GFP+) from Foxp3-RFP IL-10-GFP reporter mice and culture the cells with IL-21. Expression of Bim with and without stimulation would be analyzed with flow cytometry. We expect to see lower levels of Bim after culture with IL-21. To further test the role of PI3K on IL-21 driven suppression of

Bim, we would use PI3K inhibitor. In this experiment we expect that PI3K inhibition would abrogate the down-regulatory effect of IL-21 on Bim levels, which would implicate

IL-21-driven PI3K on Bim expression and potentially in the survival of Tfh10 cells with age. However, no change in Bim levels under IL-21 stimulatory conditions might suggest an indirect effect of IL-21 on Bim in Tfh10 cells. In other words, IL-21 might need to indirectly signal to other cells that control the survival of Tfh10 cells and their

208

Bim levels. This scenario would require targeted deletion of IL-21R on different cells

such as B cells, T cells and dendritic cells (DCs).

2- IL-10 is increased with age and counterbalances inflammaging.

Inflammaging is characterized by a large increase in the production of IL-6 which is a

trigger for serious inflammatory diseases and frailty with age (17). Several critical questions have arisen in the field of aging regarding the compensatory mechanisms that would enable the human body to cope with and regulate the persistent production of IL-

6 with age. Indeed, experimental evidence through utilizing IL-10-/- mice (IL-10 deficient mice in pathogen free environment) indicated that IL-10 is able to counteract the appearance of major physical and biological characteristics of human frailty related to

IL-6 such as sarcopenia, muscular weakness, weight loss as well as age-related

increase of serum IL-6 (36). Thus, IL-10 might represent a key to counterbalance IL-6

inflammation with age. However, the role of IL-10 in regulating inflammaging and its

cellular sources are still unclear.

In chapter 4, we reported that IL-10 expression from different tissues implicated

secondary lymphoid organs (spleen, peripheral lymph nodes) as major site for

increased IL-10 production with age. One scenario that we considered for this enhanced

IL-10 production initially was the accrual of aged Treg cells which are known to produce

IL-10. Although IL-10 production from Treg cells was increased with age, the production

of IL-10 was far greater in Foxp3- CD4+ T cells in aged mice. More importantly,

depletion studies provided strong evidence that Foxp3- cells were the predominant

source of systemic IL-10 in aged mice.

209

Interestingly, similar to our data with IL-10, we found an increase in IL-6 expression in

the spleen and lymph nodes of old mice. More importantly, when IL-10 was decreased

in the absence of IL-21, IL-6 levels were increased with age. The findings suggest that

the increase in IL-10 production from Tfh10 cells with age is a counter-regulatory

mechanism for the increased IL-6 production with age. However, this needs to be

tested.

We propose to test the role of IL-10 in counterbalancing IL-6 with age through series of

experiments. First, we will use IL-21R neutralizing antibody in aged mice as mentioned

earlier and we will measure serum IL-10 and IL-6 levels via IVCCA. We expect an

increase in systemic IL-6 and decrease in IL-10 levels upon IL-21R neutralization similar

to what we see in aged IL-21-/- mice. We will follow these experiments with additional

transfer experiments. For example, we will transfer old Tfh10 cells into a group of aged

IL-21 deficient mice. We expect that the transfer of aged Tfh10 cells would restore the homeostatic IL-6/IL-10 balance. Such findings would support the role of IL-10 in counterbalancing IL-6 with age. However, absence of expected results would suggest that IL-21 is directly controlling IL-6/IL-10 balance with age.

3- BCL6 limits IL-10 production with age.

Our study highlights a major role for BCL6 in controlling IL-10 production with age.

Indeed, we found that BCL6 is naturally downregulated in aged Tfh10 cells and the

specific loss of BCL6 in T cells resulted in a significant increase in IL-10 production from

Foxp3- CD4+ T cells, with the caveat that these IL-10+ cells no longer expressed

markers of Tfh cells. Nonetheless, these data strongly implicate BCL6 as a physiologic

210 suppressor of IL-10. We envision a few mechanisms for how BCL6 may promote IL-10 expression in Tfh10 cells. 1- Through the lack of direct repression on IL-10 due to reduction of BCL6 expression. 2- Loss of repression due to other transcription factors that promote IL-10 expression.

To examine the first scenario that BCL6 expression levels are regulating IL-10 expression in aged Tfh cells we will restore expression of BCL6 in aged Tfh cells and assess its effects on IL-10 production. Aged Tfh10 cells from Foxp3-RFP IL-10-GFP double reporter mice will be transducer with a BCL6 retrovirus or an empty vector as a control. The cells will be stimulated subsequently to examine the production of IL-10 under PMA and Ionomycin. We anticipate that over expression of BCL6 in aged Tfh10 cells would result in decreased production of IL-10 compared to control cells transduced with empty vector. Such results would provide evidence that the lower expression of

BCL6 in aged Tfh10 cells is regulating their IL-10 production.

These results would be followed with more experiments to determine the mechanisms that could be regulating the reduction in BCL6 levels in Tfh10 cells with age. Notably, both IL-6 and IL-21 are inducers of STAT3 activation, while IL-21 also induces STAT5.

More importantly, a previous study, provided evidence for STAT5 outcompeting STAT3 for binding at the BCL6 locus and directly repressing BCL6 expression in the absence of

STAT3 (37, 38). Thus, we propose that dysregulated signaling of STAT5/STAT3 in

Tfh10 cells under the effect of IL-21 and IL-6 with age would result in reduced BCL6 levels and enhanced IL-10 production. Mechanistically, expression of SOCS molecules

(e.g. SOCS3, SOCS1) following STAT3 activation in aged Tfh10 cells can result in a

211 negative feedback loop and reduction in STAT3 phosphorylation via interfering with

JAK/STAT signaling pathway in the presence of gp130 chain of IL-6R (39).

We will examine the levels of p-STAT3 and p-STAT5 in a group of young (3 month) and old (17 month) mice (Foxp3-RFP IL-10-GFP double reporter mice) to sort on CXCR5+

PD1+ RFP- GFP+ (Tfh10) cells. Sorted cells will be cultured under IL-21/IL-6 stimulatory conditions and the level of p-STAT3, p-STAT5 will be analyzed with flow cytometry. We expect that in response to IL-21/IL-6, aged Tfh10 cells would have reduced levels of p-STAT3 and higher levels of p-STAT5 compared to young counterparts. Further, we expect that this stimulation with IL-21/IL-6 will lead to reduced expression of BCL6. Together, these data would suggest that a shift in p-STAT5/p-

STAT3 under IL-21/IL-6 is regulating the reduced levels of BCL6 in Tfh10 cells with age.

This could be further tested using STAT5 inhibitor which would result in restoration of levels of BCL6 in aged Tfh10 relative to young Tfh10 cells under IL-21/IL-6 conditions.

It is possible that we would not observe a change in p-STAT3/p-STAT5 under stimulation but reduction in BCL6 levels. This result might implicate dyregulation in p-

STAT1 in aged Tfh10 cells. Notably, STAT1 can also regulate BCL6 induction downstream of IL-6R (29). Therefore, a reduction in p-STAT1 levels under IL-6 stimulatory conditions can result in the reduced expression of BCL6 in Tfh10 cells with age. Further, multiple signaling pathways contribute to BCL6 induction in Tfh cells. For example, the transcription factor BATF has been shown to positively regulate the expression of BCL6 in Tfh cells (40), while FOXO1, which is regulated by PI3K pathway, may negatively regulate BCL6 expression (41). Should we fail to observe involvement of

212

altered STAT5 signaling on BCL6 expression, we would investigate the roles of these

other pathways that may contribute to age-related loss of BCL6 expression.

It is also possible that the decrease in BCL6 may not contribute significantly to the

increased production of IL-10 in aged Tfh cells. In this case, another possible scenario

regulating IL-10 expression in aged Tfh10 cells is an increase in other transcription

factors that positively regulate IL-10 expression. Indeed, we do observe an increase in

c-Maf in Tfh10 cells with age (Figure 2, A). c-Maf is involved in the differentiation of Tfh

cells as well as IL-10 production from CD4 T cells (42, 43). Interestingly, we observed a

significant reduction in c-Maf expression in aged Tfh10 cells in the absence of IL-

21(Figure2, B). Hence, c-Maf might play a role in the production of IL-10 from Tfh cells

with age. To assess the role of c-Maf in Tfh10 with age, we propose to examine the

levels of Tfh10 cells and their production of IL-10 in old mice through inducible deletion

of c-Maf in CD4 T cells using CD4ERCre+BCL6 f /f mice. Group of old CD4ERCre+BCL6 f /f and CD4ERCre-BCL6 f /f control mice will be treated with tamoxifen to induce c-Maf deletion. Expression of c-Maf in CD4 T cells will be first assessed by flow cytometry to assess efficiency of deletion. We will also measure the level of Tfh10 cells and their IL-

10 expression via flow cytometry. We anticipate that the loss of c-Maf in CD4 T cells to

result in reduction in Tfh cells with age and there production of IL-10 compared to

control group. To further investigate the intrinsic role of c-Maf in the expression of IL-10

in Tfh10 cells, we propose to knock down c-Maf in aged Tfh10 and determine the level

of expression of IL-10 in Tfh10 cells. We anticipate that c-Maf knock down would result

in the loss of IL-10 expression from aged Tfh cells compared to control group. The data

213

would strongly support a positive intrinsic regulatory role for c-Maf in IL-10 expression in

Tfh10 cells with age.

4- IL-10 limits Tfh-dependent vaccine responses in aged mice.

Age-related increases in IL-10 can result in unwanted side effects, such as reducing

vaccine responses. Indeed, previous studies in humans have shown that lower

antibody titer to influenza A virus correlated with increased IL-10 production (44), and

levels of IFN-γ relative to IL-10 correlated with vaccine protection with age (45). A

previous study by Lefebvre et al, reported an increase in antigen specific IL-10

producing CD4 T cells which correlated with loss of vaccine responses in aged mice

(46). Although the authors attributed this increase in IL-10 production to the

accumulation of Foxp3+ Tfr cells, it was also possible that IL-10 from Tfh10 cells might

be hampering the vaccine responses in aged mice. Hence, the increase in systemic IL-

10 and the accrual of Tfh10 might hamper vaccination responses with age.

Based on these data, we hypothesized that inhibiting IL-10 would serve to enhance humoral and germinal center B cell responses with age. Our data with IL-10R

neutralization provided evidence for a negative regulation imposed by IL-10 on antigen

specific B cell responses with age. Our findings have important implications for the

development of therapeutic strategies to augment vaccination responses with age via

targeting IL-10 or Tfh10 cells. Indeed, although there are T and B cell intrinsic defects

with age, our data with IL-10R neutralization suggest that these defects are not

permanent and are amenable to manipulation.

214

Notably, our findings can be further bolstered by additional experiments that would

enable us to identify the antigen specific Tfh and Tfh10 cells and their localization during

infection relative to B cells in the germinal center. Moreover, the effect of the

neutralization of IL-10 was very promising, yet it is unclear if such effect is sufficient to

provide future protection from infection. Thus, we propose to use a Tfh dependent

vaccination model using A/PR/8/34 influenza nucleoprotein (NP) which would enable us

to track antigen specific Tfh and B cells similar to a previous study (47). Mice will be

vaccinated with recombinant NP in alum and then challenged. We will utilize four groups

of young and old Foxp-3 RFP IL-10 GFP reporter mice (Table2), which would allow us

to specifically identify via flow cytometry three populations of interest that are antigen

specific, Tfh10 (CXCR5+ PD1+ RFP- GFP+), Tfh (CXCR5+ PD1+ RFP- GFP-) and Tfr

(CXCR5+ PD1+ RFP+ GFP-). NP-specific Tfh, Tfh10 and Tfr will be detected with I-Ab –

NP311-325 Tetramer and NP-specific GC B cells will be detected by staining with PE

labeled B cell tetramer in lungs and spleen.

Table 2 Treatment NP vaccine Influenza challenge PROTECTION Young iso + Young α-IL-10R + Old iso - Old α-IL-10R + iso+ α-IL-10R d -1, 3, 6, 8, 10, Vaccine d 0, Influenza infection d21 All mice will be vaccinated with recombinant NP in alum d0 and then infected with PR8 influenza d21 post vaccination. Similar to our previous model, young and old mice will be injected with anti-IL-10R neutralizing antibody at Day-1, 3, 6, 8 and 10 or isotype control. A group of young and old mice will be used as control unvaccinated mice.

Vaccinated mice will be infected intranasally with 400 EID50 of PR8 influenza and

215 sacrificed at day 5 post infection. Second, we will monitor the weight and survival of mice throughout the experiment. Third, we will determine the level of antibody titers via

ELISA. Finally, we will utilize flow cytometry as mentioned above.

Based on our previous results, we expect that the vaccinated young mice would have the highest protection (increased survival, weight and antibody titers) compared to vaccinated aged mice. We expect the vaccinated aged mice that received anti-IL-10R neutralizing antibody to have almost similar level of protection compared to young mice and higher levels compared to aged isotype control mice. If the vaccinated aged mice are not protected that might be due to the quality of neutralizing antibody. In other words, aged mice might have acquired similar levels of antigen specific antibodies compared to the young mice with anti-IL-10R, but they are still not potent enough to provide protection.

Notably, although an NP vaccine induces strong CD8 T cell protective immunity, the protein is weak at inducing neutralizing antibodies. Additionally, previous work has shown differential expression of antibody isotypes upon IL-10R neutralization (48).

Therefore, it is possible that the effect for blocking of IL-10 signaling is selective, increasing specific isotypes which might be involved in protection. Indeed, a previous study has shown that anti-IL-10 treatment of young and aged mice caused a significant increase in influenza-specific IgM and IgG1 titers and a decrease in IgG2b and IgG3.

The study did not examine the role of these isotypes in the protection against infection which would have been very informative. Although the differential effect of IL-10R neutralization on antibody isotypes in our experiment might depend on model of vaccination this possibility should be taken into consideration.

216

Targeting IL-10 with age: Clinical implications

Several approaches have been used to improve vaccine efficacy for the elderly. For

example, TLR4 agonist as a vaccine adjuvant improved the cell-mediated immune

response with age through the stimulation of inflammatory cytokines (IL-6 and TNFα) in

myeloid DCs which was associated with reduction in IL-10 levels (49). Moreover,

another TLR4 agonist (GLA-SE), in combination with the influenza split-virus vaccine

increased significantly the ratio of IFN-γ:IL-10 produced by the aged PBMCs to levels similar to those obtained in young cells (50). Thus, our results in mice strongly suggest that interfering with IL-10 could have an adjuvant-like effect on bolstering immune responses in aged people. However, given that IL-10 deficiency in mice and humans results in significant autoimmunity, there is a concern that targeting IL-10 might results in unwanted side effects, especially in the intestine or a flare of an autoimmune disease which is a major concern. Therefore, safety of IL-10 inhibition or IL-10R blockade is important.

Fortunately, a recent study, demonstrated that blocking of IL-10 signaling at the time of immunization does not induce unwanted side effects in the intestine or result in increased risk of the development of autoimmune disease in mice (51). Indeed, results using human papillomavirus (HPV) immunization model with neutralization of IL-10 receptor during a period of two weeks indicated that the neutralization of IL-10R does

not increase the development of autoimmune disease in NOD mice or the infiltration of inflammatory cells into the intestine using inducible model of colitis, suggesting that blocking of IL-10R is relatively safe and well tolerated.

217

Notably, the effect of IL-10 neutralization on vaccination responses might depend on the different roles of antibody isotypes in antiviral immunity. Indeed, previous studies provided evidence for distinct contributions of vaccine-induced IgG1 and IgG2a antibodies to protective immunity against influenza. Stimulation of IgG2a antibodies has been associated with increased efficacy of influenza vaccination and IgG2a antibodies are more efficient at clearing the virus compared to IgG1 isotype (52). Indeed, Fc portion of IgG2a antibodies are more efficient in interacting with complement components and Fc receptors than IgG1. This interaction strongly activates Fc receptor- mediated effector functions such as antibody-dependent cell-mediated cytotoxicity and opsonophagocytosis by macrophages (53, 54). Thus, utilization of IL-10R neutralization strategy in vaccination with age might depend on its effect on the different isotypes involved in control of the virus and protection. However, the effect of IL-10 or IL-10R neutralization might largely depend on the nature of the immune response. Hence, knowledge concerning the isotypes that are most efficient in the clearance of the virus is imperative.

218

Integrated model for the regulators with age

In this dissertation we focused on understanding the molecular mechanisms that

underlie the accumulation of Tfh10 cells in aged mice. We propose a model (Figure 3)

for the homeostasis of Tfh10 with age where IL-6 is required for the initial development

of Tfh10 cells and the subsequent production of IL-21 is maintaining and regulating the

survival of Tfh10 cells by repressing Bim expression with age. More importantly, our

findings summarize the recent advance in our understanding of the crosstalk between

pro and anti-inflammatory factors with age and propose novel therapeutic strategies that

can be designed to specifically bolster immune responses with age. Indeed, work from

this dissertation provided compelling evidence for a unique interaction between aging,

CMV infection, Treg and Tfh10 cells with age. Our findings support the presence of a

strong interconnected network between the immune regulators with age. Disrupting this network can result in unforeseen consequences. Thus, therapeutic intervention on the basis of Treg and IL-10 manipulation with age should be approached with extreme caution. A considerable risk resulting from Treg manipulation in CMV infected individual is viral reactivation in the salivary glands and an increase in Tfh10 cells and production of IL-10 in the spleen thus hampering vaccination responses (Figure 4).

219

1.5 * * 11/13 1.0 2/12 3/14 3/15 0/4

0.5

0.0

Virus Titer (Log10 pfu/ml) (Log10 Titer Virus UI

D T+ ISO D T+ ISO

DT+ Anti-TNF-a DT+ Anti-TNF-a WT DTR

Figure1. 5–6 week old WT C57BL/6 and Foxp3-DTR mice were inoculated with 1× 106 plaque-forming unit (pfu, p.i.) of MCMV. 8-9 months post-MCMV infection, both groups were injected with DT on day 0, 3, 6 and sacrificed on day 7. Groups were split and were treated with 500μg of either isotype control antibody or with anti–TNF-α neutralizing antibody on day 0, 2, 5 and were sacrificed on day7. Graph shows the average viral titer of individual salivary glands (SGs) of MCMV infected mice day7 post

Treg depletion and numbers on graph indicates the number of mice positive for virus in the SG. The presence of replicating virus was detected by plaque assay. Samples with no detectable virus were assigned a titer of 0.7 log pfu/ml, the limit of detection for the plaque assay as indicated by the dashed line. Statistical analysis, *p ≤ 0.05, **p ≤ 0.01

(Mann-Whitney test)

220

Figure 2. Splenocytes from young (n=4) and old (n=4) mice were stimulated with PMA and Ionomycin. Cells were stained for TCRß, CD8, CXCR5, PD1, Foxp3, IL-10 and c-

Maf, and analyzed by flow cytometry. (A) Graph shows the level of expression of c-Maf in CXCR5+ PD1+ that are IL-10+ cells (mean±SEM). (B) Splenocytes from old WT and

IL-21-/- were stimulated and stained as above. Graph shows the level of expression of c-

Maf in CXCR5+ PD1+ that are IL-10+ cells (mean±SEM). *p ≤0.05, ***p ≤ 0.001,

Student’s t-test.

221 Figure 3. Molecular events leading to the generation and maintenance of Tfh10 cells with age. In vivo production of IL-6 induces the differentiation of Tfh10 cells and their production of IL-21 and IL-10. Subsequent expression of IL-21 by differentiated Tfh10 cells enables them to maintain their phenotype in the absence of IL-6 in a self- sustainable manner. IL-21 produced by aged Tfh10 cells is required to regulate their survival by negatively regulating the expression of pro-apoptotic molecule Bim independent of conversion and proliferation mechanisms.

222

Figure 4. Diagram describing implications for targeting Treg Foxp3+ T cells in elderly

CMV-seropositive individual. Targeting Treg in an elderly individual can result in reactivation/replication of the virus due to the effect of IL-10 production from Foxp3- IL-

10 producing cells. In the spleen, targeting Treg can result in increased levels of Tfh10 cells. Enhanced IL-10 production from Tfh10 cells in the spleen would have negative impact on B and Tfh cells in the germinal centers resulting in reduced efficacy to vaccination.

223

Refrences

1. UN. un.org/en/development/desa/population/publications/pdf/ageing/WPA2017

2017.

2. Miller RA. Effect of aging on T lymphocyte activation. Vaccine.

2000;18(16):1654-60.

3. Miller RA, Garcia G, Kirk CJ, Witkowski JM. Early activation defects in T lymphocytes from aged mice. Immunological reviews. 1997;160:79-90.

4. Tamir A, Eisenbraun MD, Garcia GG, Miller RA. Age-dependent alterations in the assembly of signal transduction complexes at the site of T cell/APC interaction. Journal of immunology. 2000;165(3):1243-51.

5. Dorshkind K, Montecino-Rodriguez E, Signer RA. The ageing immune system: is it ever too old to become young again? Nature reviews Immunology. 2009;9(1):57-62.

6. Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nature reviews Immunology. 2013;13(12):875-87.

7. Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nat Immunol. 2004;5(2):133-9.

8. Eaton SM, Burns EM, Kusser K, Randall TD, Haynes L. Age-related defects in

CD4 T cell cognate helper function lead to reductions in humoral responses. J Exp Med.

2004;200(12):1613-22.

9. Albright JW, Albright JF. Ageing alters the competence of the immune system to control parasitic infection. Immunol Lett. 1994;40(3):279-85.

224 10. Kovaiou RD, Herndler-Brandstetter D, Grubeck-Loebenstein B. Age-related

changes in immunity: implications for vaccination in the elderly. Expert Rev Mol Med.

2007;9(3):1-17.

11. Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, et al.

Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity

emerged from studies in humans. Mechanisms of ageing and development.

2007;128(1):92-105.

12. Giunta B, Fernandez F, Nikolic WV, Obregon D, Rrapo E, Town T, et al.

Inflammaging as a prodrome to Alzheimer's disease. J Neuroinflammation. 2008;5:51.

13. Fulop T, Larbi A, Witkowski JM, McElhaney J, Loeb M, Mitnitski A, et al. Aging,

frailty and age-related diseases. Biogerontology. 2010;11(5):547-63.

14. Pass RF. Epidemiology and transmission of cytomegalovirus. The Journal of infectious diseases. 1985;152(2):243-8.

15. Reddehase MJ, Podlech J, Grzimek NK. Mouse models of cytomegalovirus latency: overview. J Clin Virol. 2002;25 Suppl 2:S23-36.

16. Mekker A, Tchang VS, Haeberli L, Oxenius A, Trkola A, Karrer U. Immune senescence: relative contributions of age and cytomegalovirus infection. PLoS Pathog.

2012;8(8):e1002850.

17. Maggio M, Guralnik JM, Longo DL, Ferrucci L. Interleukin-6 in aging and chronic disease: a magnificent pathway. The journals of gerontology Series A, Biological sciences and medical sciences. 2006;61(6):575-84.

225 18. Harris TB, Ferrucci L, Tracy RP, Corti MC, Wacholder S, Ettinger WH, Jr., et al.

Associations of elevated interleukin-6 and C-reactive protein levels with mortality in the

elderly. The American journal of medicine. 1999;106(5):506-12.

19. Sinclair J, Sissons P. Latency and reactivation of human cytomegalovirus. J Gen

Virol. 2006;87(Pt 7):1763-79.

20. Simon CO, Holtappels R, Tervo HM, Bohm V, Daubner T, Oehrlein-Karpi SA, et

al. CD8 T cells control cytomegalovirus latency by epitope-specific sensing of

transcriptional reactivation. J Virol. 2006;80(21):10436-56.

21. Snyder CM, Cho KS, Bonnett EL, van Dommelen S, Shellam GR, Hill AB.

Memory inflation during chronic viral infection is maintained by continuous production of

short-lived, functional T cells. Immunity. 2008;29(4):650-9.

22. Jonjic S, Mutter W, Weiland F, Reddehase MJ, Koszinowski UH. Site-restricted

persistent cytomegalovirus infection after selective long-term depletion of CD4+ T lymphocytes. J Exp Med. 1989;169(4):1199-212.

23. Humphreys IR, de Trez C, Kinkade A, Benedict CA, Croft M, Ware CF.

Cytomegalovirus exploits IL-10-mediated immune regulation in the salivary glands. J

Exp Med. 2007;204(5):1217-25.

24. Cook CH, Trgovcich J, Zimmerman PD, Zhang Y, Sedmak DD.

Lipopolysaccharide, tumor necrosis factor alpha, or interleukin-1beta triggers reactivation of latent cytomegalovirus in immunocompetent mice. J Virol.

2006;80(18):9151-8.

226

25. Pavic I, Polic B, Crnkovic I, Lucin P, Jonjic S, Koszinowski UH. Participation of endogenous tumour necrosis factor alpha in host resistance to cytomegalovirus infection. J Gen Virol. 1993;74 ( Pt 10):2215-23.

26. Sakaguchi S, Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T. Regulatory T cells: how do they suppress immune responses? International immunology.

2009;21(10):1105-11.

27. Mandaric S, Walton SM, Rulicke T, Richter K, Girard-Madoux MJ, Clausen BE, et al. IL-10 suppression of NK/DC crosstalk leads to poor priming of MCMV-specific CD4 T cells and prolonged MCMV persistence. PLoS Pathog. 2012;8(8):e1002846.

28. Stein J, Volk HD, Liebenthal C, Kruger DH, Prosch S. Tumour necrosis factor alpha stimulates the activity of the human cytomegalovirus major immediate early enhancer/promoter in immature monocytic cells. J Gen Virol. 1993;74 ( Pt 11):2333-8.

29. Choi YS, Eto D, Yang JA, Lao C, Crotty S. Cutting edge: STAT1 is required for

IL-6-mediated Bcl6 induction for early follicular helper cell differentiation. J Immunol.

2013;190(7):3049-53.

30. Vogelzang A, McGuire HM, Yu D, Sprent J, Mackay CR, King C. A fundamental role for interleukin-21 in the generation of T follicular helper cells. Immunity.

2008;29(1):127-37.

31. Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or

17 cell lineages. Immunity. 2008;29(1):138-49.

227

32. Linterman MA, Beaton L, Yu D, Ramiscal RR, Srivastava M, Hogan JJ, et al. IL-

21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses.

J Exp Med. 2010;207(2):353-63.

33. Tang ED, Nunez G, Barr FG, Guan KL. Negative regulation of the forkhead transcription factor FKHR by Akt. J Biol Chem. 1999;274(24):16741-6.

34. Gigoux M, Shang J, Pak Y, Xu M, Choe J, Mak TW, et al. Inducible costimulator promotes helper T-cell differentiation through phosphoinositide 3-kinase. Proc Natl Acad

Sci U S A. 2009;106(48):20371-6.

35. Ostiguy V, Allard EL, Marquis M, Leignadier J, Labrecque N. IL-21 promotes T lymphocyte survival by activating the phosphatidylinositol-3 kinase signaling cascade. J

Leukoc Biol. 2007;82(3):645-56.

36. Walston J, Fedarko N, Yang H, Leng S, Beamer B, Espinoza S, et al. The physical and biological characterization of a frail mouse model. The journals of gerontology Series A, Biological sciences and medical sciences. 2008;63(4):391-8.

37. Ray JP, Marshall HD, Laidlaw BJ, Staron MM, Kaech SM, Craft J. Transcription factor STAT3 and type I interferons are corepressive insulators for differentiation of follicular helper and T helper 1 cells. Immunity. 2014;40(3):367-77.

38. Walker SR, Nelson EA, Yeh JE, Pinello L, Yuan GC, Frank DA. STAT5 outcompetes STAT3 to regulate the expression of the oncogenic transcriptional modulator BCL6. Molecular and cellular biology. 2013;33(15):2879-90.

39. Fujimoto M, Naka T. Regulation of cytokine signaling by SOCS family molecules.

Trends in immunology. 2003;24(12):659-66.

228

40. Ise W, Kohyama M, Schraml BU, Zhang T, Schwer B, Basu U, et al. The transcription factor BATF controls the global regulators of class-switch recombination in both B cells and T cells. Nat Immunol. 2011;12(6):536-43.

41. Kerdiles YM, Stone EL, Beisner DR, McGargill MA, Ch'en IL, Stockmann C, et al.

Foxo transcription factors control regulatory T cell development and function. Immunity.

2010;33(6):890-904.

42. Andris F, Denanglaire S, Anciaux M, Hercor M, Hussein H, Leo O. The

Transcription Factor c-Maf Promotes the Differentiation of Follicular Helper T Cells.

Front Immunol. 2017;8:480.

43. Pot C, Jin H, Awasthi A, Liu SM, Lai CY, Madan R, et al. Cutting edge: IL-27 induces the transcription factor c-Maf, cytokine IL-21, and the costimulatory receptor

ICOS that coordinately act together to promote differentiation of IL-10-producing Tr1 cells. J Immunol. 2009;183(2):797-801.

44. Corsini E, Vismara L, Lucchi L, Viviani B, Govoni S, Galli CL, et al. High interleukin-10 production is associated with low antibody response to influenza vaccination in the elderly. J Leukoc Biol. 2006;80(2):376-82.

45. McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, et al. T cell responses are better correlates of vaccine protection in the elderly. J Immunol.

2006;176(10):6333-9.

46. Lefebvre JS, Masters AR, Hopkins JW, Haynes L. Age-related impairment of humoral response to influenza is associated with changes in antigen specific T follicular helper cell responses. Sci Rep. 2016;6:25051.

229

47. Lefebvre JS, Lorenzo EC, Masters AR, Hopkins JW, Eaton SM, Smiley ST, et al.

Vaccine efficacy and T helper cell differentiation change with aging. Oncotarget.

2016;7(23):33581-94.

48. Dobber R, Tielemans M, Nagelkerken L. The in vivo effects of neutralizing antibodies against IFN-gamma, IL-4, or IL-10 on the humoral immune response in young and aged mice. Cellular immunology. 1995;160(2):185-92.

49. Mast BT, Azar AR, Murrell SA. The vascular depression hypothesis: the influence of age on the relationship between cerebrovascular risk factors and depressive symptoms in community dwelling elders. Aging Ment Health. 2005;9(2):146-52.

50. Behzad H, Huckriede AL, Haynes L, Gentleman B, Coyle K, Wilschut JC, et al.

GLA-SE, a synthetic toll-like receptor 4 agonist, enhances T-cell responses to influenza vaccine in older adults. The Journal of infectious diseases. 2012;205(3):466-73.

51. Ni G, Liao Z, Chen S, Wang T, Yuan J, Pan X, et al. Blocking IL-10 signalling at the time of immunization does not increase unwanted side effects in mice. BMC

Immunol. 2017;18(1):40.

52. Huber VC, McKeon RM, Brackin MN, Miller LA, Keating R, Brown SA, et al.

Distinct contributions of vaccine-induced immunoglobulin G1 (IgG1) and IgG2a antibodies to protective immunity against influenza. Clin Vaccine Immunol.

2006;13(9):981-90.

53. Huber VC, Lynch JM, Bucher DJ, Le J, Metzger DW. Fc receptor-mediated phagocytosis makes a significant contribution to clearance of influenza virus infections.

J Immunol. 2001;166(12):7381-8.

230 54. Neuberger MS, Rajewsky K. Activation of mouse complement by monoclonal mouse antibodies. Eur J Immunol. 1981;11(12):1012-6.

231