<<

-Induced Changes in Hematopoiesis Arielle Glatman Zaretsky, Julie B. Engiles and Christopher A. Hunter This information is current as J Immunol 2014; 192:27-33; ; of September 29, 2021. doi: 10.4049/jimmunol.1302061 http://www.jimmunol.org/content/192/1/27 Downloaded from References This article cites 104 articles, 32 of which you can access for free at: http://www.jimmunol.org/content/192/1/27.full#ref-list-1

Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists

• Fast Publication! 4 weeks from acceptance to publication

*average by guest on September 29, 2021 Subscription Information about subscribing to The Journal of is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Th eJournal of Brief Reviews Immunology

Infection-Induced Changes in Hematopoiesis Arielle Glatman Zaretsky,* Julie B. Engiles,† and Christopher A. Hunter* The (BM) is an important site for the in- of lineage commitments. Lineageneg Sca-1+ c-kit+ cells (LSKs; terrelated processes of hematopoiesis, , reviewed extensively in Refs. 7–9) include the earliest hema- , and . A wide variety of mi- topoietic precursors in the BM with the potential to develop crobial challenges are associated with profound changes into multiple lineage-specific progenitors, such as common in this compartment that impact on hematopoietic dif- lymphoid and myeloid progenitors and or ferentiation and mobilization of a variety of cell types. erythrocytic precursors (Fig. 1). Notably, only a small per- This article reviews some of the key pathways that con- centage of LSKs are HSCs; the majority of the LSK population trol BM homeostasis, the infectious and inflammatory represents a variety of multipotent or lineage committed cells. At steady-state, this differentiation is a complex but well-ordered

processes that affect the BM, and how addressing the Downloaded from knowledge gaps in this area has the potential to widen process, leading to the development of , gran- our comprehension of immune homeostasis. The Journal ulocytes, and myeloid cells. of Immunology, 2014, 192: 27–33. Given the diverse functions of the BM, it is not surprising that this organ is composed of distinct anatomical compartments. For example, within the BM, HSCs are distributed primarily in or he bone marrow (BM) is a critical site of immune cell near the endosteal region or the interface between medullary http://www.jimmunol.org/ development and erythropoiesis, and provides a niche bone and stromal cells (Fig. 1). This is a site with a distinctive T for plasma cells and memory T cells. Although the cell microanatomic , although recent evidence populations and structural elements in the BM are typically indicates that perivascular niches also support HSC populations characterized by composition and morphology (1, 2), the ap- (6). The retention of HSCs in this environment is thought to plication of novel imaging technologies, such as intravital im- promote survival and/or maintain hematopoietic progenitors in aging and laser-scanning cytometry, has allowed the field to the quiescent G0 phase of the cell cycle, allowing these cells to better define the microenvironments within this complex organ. self-renew and offering a ready pool of cells for rapid emer- Similarly, the use of conditional knockout technologies has gence (10–12). The direct interactions between vascular stromal helped to clarify the factors that maintain stem cell populations cells and nestin-negative mesenchymal progenitors, and be- by guest on September 29, 2021 and support the development of hematopoietic precursors and tween osteoblastic cells and HSCs themselves, promote HSC immature B cells (3–6). Using these advances, recent subsetting survival and control niche size (5, 13). of stromal and precursor populations in the BM has provided Several and molecules, notably CXCL12 insights into their behavior in the endosteal and perivascular and VLA-4 (14), contribute to HSC localization and main- compartments (3–6). In addition to the central role of the BM in tenance, and the local production of by mes- maintaining immune homeostasis, the ability to generate and enchymal and perivascular stromal cells, as well as endothelial mobilize immune cells in response to infection is a key function cells, promotes the generation and maintenance of HSCs (6). of this system. Notably, emergency granulopoiesis and rapid Perhaps the best studied –receptor pair in this pro- mobilization of from the BM is key for resistance cess is CXCL12–CXCR4, and disruption of this pathway leads to many . Similarly, increased erythropoiesis can be to alterations in cellular retention in the BM, including mo- a physiological response to inflammation, but certain in- bilization of early lymphoid progenitors and HSCs (14, 15). fections lead to the depletion of erythroid precursors and the However, these cell types occupy distinct niches, populated by development of anemia. The overarching goal of this review is to discrete populations of CXCL12-producing cells (16). Thus, discuss the role of the BM niche in the host response to infection, expression of CXCL12 by endothelial cells, perivascular stromal illustrate the impact of infectious on this compart- cells, and osteoblasts supports specific cell types within dis- ment, and highlight some of the major questions in the field. tinct niches. For example, the use of lineage-specific deletions Hematopoiesis and the niche established that nestin-negative mesenchymal progenitors, not Hematopoiesis is the process by which hematopoietic stem CXCL12-abundant reticular cells (which make the majority of cells (HSCs) differentiate into immune cells through a series CXCL12) (12, 17, 18), are the critical source of CXCL12

*Department of Pathobiology, School of Veterinary Medicine, University of Pennsylva- 380 South University Avenue, Room 313 Hill Pavilion, Philadelphia, PA 19104. nia, Philadelphia, PA 19104; and †New Bolton Center, Kennett Square, PA 19348 E-mail address: [email protected] Received for publication August 2, 2013. Accepted for publication October 30, 2013. Abbreviations used in this article: BM, bone marrow; HSC, hematopoietic stem cell; LSK, lineageneg Sca-1+ c-kit+ cell; Treg, regulatory . This work was supported by the National Institutes of Health (Grants AI 042334 and T32 AI 055400), the American Foundation, and the Commonwealth of Pennsylvania. Copyright Ó 2013 by The American Association of Immunologists, Inc. 0022-1767/13/$16.00 Address correspondence and reprint requests to Dr. Christopher A. Hunter. Depart- ment of Pathobiology, School of Veterinary Medicine, University of Pennsylvania,

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1302061 28 BRIEF REVIEWS: INFECTION-INDUCED CHANGES IN HEMATOPOIESIS

FIGURE 1. HSC responses to infection. (A–C) Po- tential routes of sensing by HSCs. (A) HSCs in the BM can express pattern recognition receptors, such as TLRs; thus, when the BM is directly infected, these cells may recognize pathogen-derived Ag or immune products produced in response to infection within the BM. Alter- natively, circulating HSCs in the expressing pattern recognition receptors may recognize pathogen associated molecular patterns and traffic back to the BM to relay these signals. (B) HSCs in the BM may be directly infected by pathogens, such as JC and respond directly to this challenge. (C) produced at distal sites, such as IFN-g or type I IFNs, may enter the BM and signal HSCs to initiate BM responses. Infection-induced changes in the BM (D). HSCs are maintained by stromal cells and CXCL12–CXCR4 interactions. Although HSCs are de- picted in endosteal regions, there is also evidence for their

presence in perivascular niches. These cells can differ- Downloaded from entiate into myeloid or common lymphoid progenitors, which then undergo , granulopoiesis, and/or lymphopoiesis. The impact of infection on these processes is shown, with increased myelopoiesis and emergency granulopoiesis correlating with decreased lymphopoiesis. http://www.jimmunol.org/

required to maintain HSCs in the BM niche (11). This study Notably, some of the responses associated with diverse in- also demonstrated that other stromal cells are key sources of fections are context dependent, which likely reflects different CXCL12 required for survival of progenitors. Of note, host–pathogen interactions. Thus, whether a pathogen can pre-B, pro-B, and mature B cells, as well as , ex- establish infection in the BM and which cell types it infects are press high levels of CXCR4, although mature and immature relevant factors. This has led to an interest in understanding B cell subsets are the least responsive to CXCL12. These dif- whether HSCs are inherently resistant to infection (9); however, by guest on September 29, 2021 ferential sensitivities imply that later-stage B cell populations are many of these studies have used pathogens that do not com- poised to traffic out of the BM, whereas early-stage B cells are monly infect the BM or pathogens that typically require more receptive to signals that maintain their BM localization , a process that HSCs cannot perform. For in- (19). Understanding these events has led to the use of blocking stance, mycobacterial species can be isolated from the BM reagents for CXCL12 and CXCR4, which mobilize HSCs from under a variety of circumstances, which range from asymp- the BM, resulting in improved harvesting of stem cells for tomatic individuals to AIDS patients with overt clinical , transplantation (14, 17, 20). and a recent report highlighted that Mycobacterium Inflammation, infection, and hematopoiesis resides latently in mesenchymal stem cells, which are phagocytic (34). The importance of understanding these specifics is ex- It has long been recognized that systemic infection with a emplified by the disparate responses associated with different variety of bacterial, viral, and parasitic organisms can result in . Thus, the presence of the noncytolytic lymphocytic profound alterations in the BM, many of which appear to be choriomeningitis virus in the BM contributes to the reduced part of a conserved host response to microbial challenge (Fig. 1). For example, during malaria and toxoplasmosis (and other ability of HSCs to engraft in this site (35, 36). JC virus, the systemic challenges), there is an increase in granulocytes in the of progressive multifocal leukoencephalopathy in im- BM, but a transient decrease in the numbers of lymphocytes, munocompromised patients, can infect HSCs and B cells, erythrocytes, and , despite low parasite bur- and is known to persist within the BM. Notably, several Ab- mediated therapies in humans that target LFA-1, VLA-4, or dens in this site (21–24). Increased populations of LSKs + and/or HSCs in the BM are the hallmark of many experi- CD20 lead to mobilization of pre-BandBcellsandCD34 mental (25–31). Thus, challenge with Plasmodium HSC progenitors from the BM. It has been proposed that in chabaudi or Pneumocystis carinii, organisms not typically found the context of reduced immune surveillance, these events in the BM, leads to increased LSK and HSC populations in the promote the dissemination of JC virus to the brain (37, 38). BM and circulation, followed by an increase in multipotent CMV can also infect stromal and mononuclear cells within progenitor cells (25, 26). Similarly, murine ehrlichiosis results in the BM, which has been linked to a reduced ability to make increased numbers of LSKs, although these cells appear to have progenitor colonies (39). Although the examples discussed a defect in their ability to differentiate (32, 33). For many earlier focus on organisms found in the BM during infection, of these examples, it is unclear whether these alterations are reductions in precursor populations can also occur with in- secondary consequences of systemic inflammation or part of fectious challenges that do not establish in this site. For ex- a coordinated host response to limit infection. ample, marked decreases in CD34+ hematopoietic precursors The Journal of Immunology 29 have been reported during HIV and SIV infection without parasite of mice that is restricted to the gut, this challenge detection of local virus (40–43). leadstoepithelialcellproductionofthymicstromallym- Whether HSCs have an active role in immune sensing phopoietin that induces production in the BM (57). remains an open question (Fig. 1), and it has been proposed In contrast, increased numbers are characteristic that the expansion of HSC populations may serve as a com- of many bacterial infections, and G-CSF promotes “emergency ponent of the primary response, as well as a mechanism to granulopoiesis” in the BM (58, 59). At the molecular level, replenish depleted progenitor populations (9). In adults, small steady-state granulopoiesis is regulated through the transcrip- numbers of HSCs appear to traffic between the BM and cir- tion factor C/EBPa, whereas C/EBPb and STAT3 mediate G- culation, perhaps acting as a form of immune surveillance that CSF–dependent granulopoiesis (60, 61). Naive mice depleted can relay distal signals to the BM (44, 45). Evidence in favor of of neutrophils or injected with the adjuvant alum (which surveillance activity includes HSC expression of TLRs (Fig. 1) induces mobilization) exhibit emergency gran- (9). Furthermore, TLR signaling in LSKs and other hemato- ulopoiesis and proliferation of HSCs in a G-CSF– and poietic progenitors results in myeloid differentiation (46), C/EBPb–dependent manner (62). This body of work illustrates whereas TLR9 is required in a model of HSV-1 infection for the feedback mechanisms that allow cells in the BM to respond HSCs to produce dendritic cells (47). There are studies in rapidly to changes in the periphery, and suggests the presence which MyD88, the adaptor molecule involved in TLR and IL-1 of a density-sensing mechanism that regulates granulopoietic signaling, has been shown to be critical for infection-induced activity (62). granulopoiesis, myelopoiesis, and mobilization of these pop- The BM is also an active site of myelopoiesis, leading to the Downloaded from ulations. Thus, infection with vaccinia virus in vivo or culture production of and . BM macrophages of LSKs with Candida albicans in vitro led to increased num- have an important role in maintaining the HSC niche (63), yet bers of LSKs and differentiated myeloid cells in the BM in an the populations that are mobilized in response to infection MyD88-dependent manner (27, 28). However, the observa- have key roles in the development and resolution of inflam- tion that the increase in LSKs present in a model of bacterial mation, as well as acting as potent effectors or infection with aureus is MyD88 (64). In mice challenged with T. gondii or Listeria mono- http://www.jimmunol.org/ independent (30) highlights the gaps in our understanding of cytogenes, the CCR2-dependent mobilization of monocytes the cross talk between the peripheral immune response and out of the BM is essential to control these organisms (64–67). the BM compartment. Nevertheless, this literature provides a During Ehrlichia infection, IFN-g is required to activate direct link between pathogen recognition and mobilization macrophages to control this intracellular bacterium, but IFN-g of the appropriate innate populations required to control also contributes to the diminished hematopoietic progenitor infection. population in the BM (33). Indeed, the ability of IFN-g to induce SOCS3 in granulocyte- progenitors leads Erythropoiesis

to reduced G-CSF signaling andashiftfromneutrophil by guest on September 29, 2021 The suppression of erythropoiesis and development of anemia production to myeloid differentiation (68). In this context, it is characteristic of many infections (48). For organisms, such is tempting to speculate that systemic IFN-g (or direct TLR as Plasmodium and Babesia sp., that directly infect erythro- signaling) provides a mechanism to tailor BM output to the cytes, there is a clear link to erythrocyte destruction that can class of pathogen. However, the identification of an IFN-g– eventually lead to a depletion of erythroid precursor cells in dependent atypical progenitor population of IL-7R+c-kithi a chronic setting. For other pathogens, such as the African cells, with predominantly myeloid potential, that is involved Trypanosomes, the presence of parasites in the bloodstream in clearance of P. chabaudi (26) illustrates the broad effects of is associated with damage of erythrocytes, elevated eryth- IFN-g on myelopoiesis. rophagocytosis, and ultimately, decreased erythropoiesis (49). In other settings, severe anemia associated with the loss of B cell lymphopoiesis and homeostasis erythroid precursors also has an immune component (50, 51); The BM is also a site of B cell development, and many in- Ehrlichia muris and Toxoplasma gondii do not infect eryth- fections can profoundly impact this process (47, 69–74). rocytes, but these distinct challenges lead to a reduction in Challenge with influenza or lymphocytic choriomeningitis erythroid precursors and severe anemia (32). Although the virus results in a transient decrease in pro-, pre-, and imma- cytokines IFN-g, IL-6, and IL-15 are implicated in immune- ture B cells in the BM, which is, in part, dependent on TNF-a mediated anemia (24, 52), it remains unclear whether this and a (71, 72). In a model of bacterial sepsis, the response simply reflects an interesting epiphenomenon or is early depletion of B cell progenitors is delayed in MyD88- part of a conserved host response that limits availability of deficient mice, indicating a role for TLR or IL-1 family mem- host cells for organisms that do infect erythrocytes. bers (30). Although the physiological significance of these events remains to be defined, the block in B cell development Granulopoiesis and myelopoiesis in the BM correlates with reduced humoral responses to irrelevant Ags Increased granulopoiesis within the BM is a hallmark of acute (71, 72). Interestingly, under inflammatory conditions, there infection or inflammation in experimental and clinical settings is an inverse correlation between the induction of granulopoiesis that gives rise to short-lived neutrophils, , and and decreased lymphopoiesis in the BM (75). In one experi- (21, 23, 32, 53–56). It has long been recognized mental system, treatment of mice with IFA results in an increase that increased circulating levels of basophils and eosinophils in granulocyte numbers, but a decline in the numbers of B (and are characteristic of many helminth parasites, but how the T) lymphocytes in the BM (76). Similarly, during infection with communicates with the BM to promote this the bacterium E. muris, a transient decrease in B220+ cells in the process has been unclear. For Trichuris muris, a nematode BM is accompanied by an increase in granulocytes (32). Insight 30 BRIEF REVIEWS: INFECTION-INDUCED CHANGES IN HEMATOPOIESIS into how these events may be coordinated is provided by the quired for HSC self-renewal, leading to a reduction in the observation that, although lymphoid and granulocytic precursors number of HSCs (94). express CXCR4, the disruption of the CXCL12/CXCR4 axis Systemic levels of IL-1 and TNF are also characteristic of during inflammation leads to preferential loss of B cell precursors, many infectious challenges and have been linked to alter- potentially providing space to generate additional granulocytes ations in the BM. TNF-a treatment results in a reduction required for resistance to infection (75). These studies highlight in progenitor populations in the BM, whereas IL- the coordinated changes that occur in hematopoietic processes 1b elicits increased granulocyte precursors (76). Moreover, in the BM during infection, which are presumably required to CXCR4-deficient mice or mice treated with pertussis , allow the development of appropriate responses to different which blocks chemokine signaling, given IL-1 or TNF-a have classes of pathogen. increased B cell and myeloid progenitors in the circulation, in- dicating that CXCR4–CXCL12 interactions facilitate - BM as a niche for plasma cells and memory T cells mediated regulation of B cell and myeloid cell retention in the a The BM also provides a niche for long-lived BM (76). In addition, TNF- and IL-1 (and RANKL and populations that continually produce Abs against previously M-CSF) can induce differentiation from mononuclear encountered Ags. The ability of eosinophils, basophils, meg- precursor cells and subsequent inflammatory osteolysis, a com- akaryocytes, and stromal cells in this site to produce BLyS, plication in many infectious, inflammatory, and neoplastic dis- April, IL-6, and CXCL12 is required for the survival and eases (96–98). Additional studies are required to understand the retention of plasma cells (3, 4, 77–83). As noted earlier, in- physiological significance of these cytokine-mediated changes in Downloaded from fection can lead to alterations in many of these cell populations, the BM and whether they impact on the development of im- and there is evidence that without eosinophils, alternative mune responses that are tailored to specific pathogens. plasma cell survival niches can be established in the spleen BM as an immune-privileged site (78). Memory CD4+ and CD8+ T cells also reside within the As described earlier, the BM can be a site of infection, but it is BM, maintained by stromal cells that produce the cytokines sensitive to the systemic effects of microbial challenge at distal http://www.jimmunol.org/ IL-7 and IL-15 that act as survival and proliferative factors for locations, and it is likely that these processes could be det- these populations (84–89). In humans, it has been shown rimentaltoessentialstemcellpopulations.Consequently, that memory T cells in the BM are more highly activated mechanisms to temper the adverse effects of inflammation on and polyfunctional than those isolated from blood, although different stem cell niches may be necessary. Although the BM memory T cells from blood can develop a similar phenotype lacks a physical barrier to exclude immune cells, there are after culture with IL-15 (87, 90). Thus far, it is unclear whether elements of immune privilege in this compartment that may the profound infection-induced changes in the BM impact on protect progenitors from immune-mediated damage or in- the (ill-defined) memory T cell and plasma cell niches or on the flammatory signals that could lead to transformation of these function of these populations. Understanding how different by guest on September 29, 2021 long-lived pluripotent cells. Notably, as much as ∼25% of infections influence the homeostasis of memory cell pop- BM CD4+ TcellsareFoxp3+ regulatory T cells (Tregs), a ulations in the BM may provide opportunities to manipulate much higher frequency than the 5–10% typically present these niches, and thus aid in the design of vaccines that induce in other sites. Intravital imaging of the BM of Foxp3-GFP long-lasting . reporters revealed that Tregs are predominantly located in or near the endosteal region, with the majority of HSCs found Impact of systemic cytokine responses on the BM compartment in close proximity to or in contact with Tregs (99). The Although some changes that occur in the BM during certain significance of these populations in infection has not been infections may be attributed to the local presence of pathogens, addressed; however, in an allogeneic transfer model, depletion perhaps the most common scenario is that the production of of Tregs prevented engraftment of an allo-HSC progenitor cytokines at distal sites affects the BM (Fig. 1) (91). Thus, type population (99). After BM transplantation, BM T cells ex- I IFNs can shift HSCs out of cell-cycle arrest and induce press a unique profile of surface markers and cytokine pro- proliferation and differentiation, ultimately resulting in de- duction, with high expression of CD44, CD62L, and CD45RB, creased numbers of HSCs (92). In murine models of influenza higher levels of IFN-g,IL-4,andIL-10,anddecreasedIL-2 or Sendai virus, production of type I IFNs in the leads to secretion, compared with those in other sites, and an in- upregulation of antiviral genes in hematopoietic cells in the creased ability to protect from graft-versus-host disease (100). BM (93). Whether these factors are produced at sufficiently Similar findings were reported in human patients, wherein high levels in the to have systemic effects in the BM or increased Tregs positively correlated with lower incidence whether there is a mechanism to relay these signals to the BM of graft-versus-host disease (100, 101). More recently, in is unclear. As discussed earlier, IFN-g can modify myelo- two models of arthritic disease, Tregs in the BM inhibited poiesis (26, 33, 68), and other aspects of hematopoiesis (94) TNF-mediated bone damage (102), as well as plasma cell and the high systemic levels of IFN-g characteristic of many accumulation (103). Taken together, these studies suggest infections suggest it would have a major impact on the BM. that the Treg population in the BM creates a suppressive During chronic infection with Mycobacterium avium,this environment, which establishes a specialized niche for HSCs. production of IFN-g can activate HSCs from the quiescent Although Treg populations at other sites can be significantly state (95). In addition, IFN-g–mediated induction of SOCS1 altered during infection (104–106), how those in the BM (an inhibitor of cytokine signaling) inhibits the ability of the are influenced by inflammation or infection and whether cytokine thrombopoietin to activate STAT5, which is re- they preserve different niches or are resident or transient The Journal of Immunology 31 populations represent distinct gaps in our understanding of 2. Travlos, G. S. 2006. Normal structure, function, and histology of the bone marrow. Toxicol. Pathol. 34: 548–565. BM dynamics. 3. Tokoyoda, K., A. E. Hauser, T. Nakayama, and A. Radbruch. 2010. Organization of by bone marrow . Nat. Rev. Immunol. 10: 193– 200. Conclusions 4. Tokoyoda, K., T. Egawa, T. Sugiyama, B.-I. Choi, and T. Nagasawa. 2004. There have been significant advances in characterizing the Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity 20: 707–718. effects of infection and inflammation on the function of the 5. Nombela-Arrieta, C., G. Pivarnik, B. Winkel, K. J. Canty, B. Harley, BM, but major gaps remain in our understanding of whether J. E. Mahoney, S. Y. Park, J. Lu, A. Protopopov, and L. E. Silberstein. 2013. these changes impact the ability of a host to control pathogens. Quantitative imaging of haematopoietic stem and progenitor cell localization and hypoxic status in the bone marrow microenvironment. Nat. Cell Biol. 15: 533– Insomesituations,suchasemergencygranulopoiesisormono- 543. cyte mobilization, it is easy to link these processes with re- 6. Ding, L., T. L. Saunders, G. Enikolopov, and S. J. Morrison. 2012. Endothelial and perivascular cells maintain haematopoietic stem cells. 481: 457–462. sistance to infection. Alternatively, it is possible that some of 7. Krause, D. S., D. T. Scadden, and F. I. Preffer. 2013. The hematopoietic stem cell the global changes in BM cell populations reflect a shift in niche—home for friend and foe? Cytometry B Clin. Cytom. 84: 7–20. energetic resources from hematopoiesis to the support of ef- 8. Kondo, M. 2010. Lymphoid and myeloid lineage commitment in multipotent hematopoietic progenitors. Immunol. Rev. 238: 37–46. fector populations required for pathogen control. Interest- 9. King, K. Y., and M. A. Goodell. 2011. Inflammatory modulation of HSCs: ingly, diminished BM hematopoietic progenitor populations viewing the HSC as a foundation for the immune response. Nat. Rev. Immunol. 11: 685–692. during infection are frequently accompanied by the devel- 10. Me´ndez-Ferrer, S., T. V. Michurina, F. Ferraro, A. R. Mazloom, B. D. Macarthur, opment of extramedullary hematopoiesis in the spleen and S. A. Lira, D. T. Scadden, A. Ma’ayan, G. N. Enikolopov, and P. S. Frenette. (31, 33, 107), which may open niches and resources for 2010. Mesenchymal and haematopoietic stem cells form a unique bone marrow Downloaded from niche. Nature 466: 829–834. the process of granulopoiesis and myelopoiesis. In addition, 11. Greenbaum, A., Y. M. Hsu, R. B. Day, L. G. Schuettpelz, M. J. Christopher, the basis for and biological impact of the infection-induced J. N. Borgerding, T. Nagasawa, and D. C. Link. 2013. CXCL12 in early mes- enchymal progenitors is required for haematopoietic stem-cell maintenance. Na- blockade of B cell development in the BM remains unclear ture 495: 227–230. (108). In the context of infection, the presence of microbial 12. Sugiyama, T., H. Kohara, M. Noda, and T. Nagasawa. 2006. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone Ags in the BM while B cells are undergoing selection could marrow niches. Immunity 25: 977–988. lead to the development of B cells that are tolerant to path- 13. Zhang, J., C. Niu, L. Ye, H. Huang, X. He, W. G. Tong, J. Ross, J. Haug, http://www.jimmunol.org/ ogen Ags or even to the deletion of pathogen-specific B cells. T. Johnson, J. Q. Feng, et al. 2003. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425: 836–841. To the best of our knowledge, this idea has not been tested, 14. Motabi, I. H., and J. F. DiPersio. 2012. Advances in stem cell mobilization. Blood but this phenomenon may provide a mechanism to limit Rev. 26: 267–278. 15. Ma, Q., D. Jones, and T. A. Springer. 1999. The chemokine receptor CXCR4 is deleterious effects of infection on . required for the retention of B lineage and granulocytic precursors within the bone Regardless, despite dramatic changes in numerous resident marrow microenvironment. Immunity 10: 463–471. BM populations in response to inflammation, the BM niche 16. Ding, L., and S. J. Morrison. 2013. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature 495: 231–235. does appear to return to a normal steady-state. Whether this 17. Shiozawa, Y., and R. S. Taichman. 2012. Getting blood from bone: an emerging disruption impacts long-term hematopoiesis is not yet ap- understanding of the role that osteoblasts play in regulating hematopoietic stem by guest on September 29, 2021 cells within their niche. Exp. Hematol. 40: 685–694. preciated. Consequently, there are open questions about the 18. Sugiyama, T., and T. Nagasawa. 2012. Bone marrow niches for hematopoietic processes that lead to restoration of this environment and stem cells and immune cells. Inflamm. Targets 11: 201–206. whether they are different from those involved in the initial 19. Honczarenko, M., R. S. Douglas, C. Mathias, B. Lee, M. Z. Ratajczak, and L. E. Silberstein. 1999. SDF-1 responsiveness does not correlate with CXCR4 seeding of the BM. Understanding how restoration occurs may expression levels of developing human bone marrow B cells. Blood 94: 2990–2998. translate into more effective strategies to achieve BM recon- 20. Rettig, M. P., G. Ansstas, and J. F. DiPersio. 2012. Mobilization of hematopoietic stem and progenitor cells using inhibitors of CXCR4 and VLA-4. Leukemia 26: stitution after irradiation, infection, or other processes that 34–53. disrupt BM homeostasis. Indeed, because stem cells can 21. Villeval, J. L., A. Gearing, and D. Metcalf. 1990. Changes in hemopoietic and reside in the BM in a dormant state (109), incorrect reseeding regulator levels in mice during fatal or nonfatal malarial infections. II. Non- erythroid populations. Exp. Parasitol. 71: 375–385. of the BM or contamination of this immune-privileged site 22. Villeval, J. L., A. Lew, and D. Metcalf. 1990. Changes in hemopoietic and reg- could provide protected niches for these cells. Finally, al- ulator levels in mice during fatal or nonfatal malarial infections. I. Erythropoietic populations. Exp. Parasitol. 71: 364–374. though histological studies of the BM have provided the 23. Petakov, M., N. Stojanovic´,G.Jovcic´, D. Bugarski, V. Todorovic´,and foundation to characterize this compartment (2), improved O. Djurkovic´-Djakovic´. 2002. Hematopoiesis during acute Toxoplasma gondii infection in mice. Haematologia (Budap.) 32: 439–455. imaging technologies continue to improve our knowledge of 24. Chou, D. B., B. Sworder, N. Bouladoux, C. N. Roy, A. M. Uchida, M. Grigg, the cell–cell interactions involved in the regulation of the BM P. G. Robey, and Y. Belkaid. 2012. Stromal-derived IL-6 alters the balance of niche. Combined with infectious models that have different myeloerythroid progenitors during Toxoplasma gondii infection. J. Leukoc. Biol. 92: 123–131. effects on BM hematopoiesis, these technologies can be used 25. Shi, X., P. Zhang, G. D. Sempowski, and J. E. Shellito. 2011. Thymopoietic and to better understand the normal developmental processes that bone marrow response to murine Pneumocystis . Infect. Immun. 79: 2031–2042. occur in the BM. 26. Belyaev, N. N., D. E. Brown, A. I. Diaz, A. Rae, W. Jarra, J. Thompson, J. Langhorne, and A. J. Potocnik. 2010. Induction of an IL7-R(+)c-Kit(hi) mye- lolymphoid progenitor critically dependent on IFN-gamma signaling during acute Acknowledgments malaria. Nat. Immunol. 11: 477–485. We thank Deborah Argento for graphics assistance with figure design. 27. Singh, P., Y. Yao, A. Weliver, H. E. Broxmeyer, S. C. Hong, and C. H. Chang. 2008. Vaccinia virus infection modulates the hematopoietic cell compartments in the bone marrow. Stem Cells 26: 1009–1016. Disclosures 28. Ya´n˜ez, A., C. Murciano, J. E. O’Connor, D. Gozalbo, and M. L. Gil. 2009. Candida albicans triggers proliferation and differentiation of hematopoietic stem The authors have no financial conflicts of interest. and progenitor cells by a MyD88-dependent signaling. Microbes Infect. 11: 531– 535. 29. Zhang, P., S. Nelson, G. J. Bagby, R. Siggins, II, J. E. Shellito, and D. A. Welsh. References 2008. The lineage-c-Kit+Sca-1+ cell response to Escherichia coli bacteremia in 1. Yang, M., G. Bu¨sche, A. Ganser, and Z. Li. 2013. Morphology and quantitative Balb/c mice. Stem Cells 26: 1778–1786. composition of hematopoietic cells in murine bone marrow and spleen of healthy 30. Scumpia, P., K. Kelly-Scumpia, M. Delano, J. Weinstein, A. Cuenca, S. Al-Quran, subjects. Ann. Hematol. 92: 587–594. I. Bovio, S. Akira, Y. Kumagai, and L. Moldawer. 2010. Cutting edge: bacterial 32 BRIEF REVIEWS: INFECTION-INDUCED CHANGES IN HEMATOPOIESIS

infection induces hematopoietic stem and progenitor cell expansion in the absence 56. Van Ginderachter, J. A., A. Beschin, P. De Baetselier, and G. Raes. 2010. of TLR signaling. J. Immunol. 184: 2247–2251. Myeloid-derived suppressor cells in parasitic infections. Eur. J. Immunol. 40: 31. Glatman Zaretsky, A., J. S. Silver, M. Siwicki, A. Durham, C. F. Ware, and 2976–2985. C. A. Hunter. 2012. Infection with Toxoplasma gondii alters lymphotoxin ex- 57. Siracusa, M. C., S. A. Saenz, D. A. Hill, B. S. Kim, M. B. Headley, T. A. Doering, pression associated with changes in splenic architecture. Infect. Immun. 80: 3602– E. J. Wherry, H. K. Jessup, L. A. Siegel, T. Kambayashi, et al. 2011. TSLP 3610. promotes -3-independent basophil and type 2 inflam- 32. MacNamara, K. C., R. Racine, M. Chatterjee, D. Borjesson, and G. M. Winslow. mation. Nature 477: 229–233. 2009. Diminished hematopoietic activity associated with alterations in innate and 58. Demetri, G. D., and J. D. Griffin. 1991. Granulocyte colony-stimulating factor adaptive immunity in a mouse model of human monocytic ehrlichiosis. Infect. and its receptor. Blood 78: 2791–2808. Immun. 77: 4061–4069. 59. Lieschke, G. J., D. Grail, G. Hodgson, D. Metcalf, E. Stanley, C. Cheers, 33. MacNamara, K. C., M. Jones, O. Martin, and G. M. Winslow. 2011. Transient K. J. Fowler, S. Basu, Y. F. Zhan, and A. R. Dunn. 1994. Mice lacking granulocyte activation of hematopoietic stem and progenitor cells by IFNg during acute colony-stimulating factor have chronic , granulocyte and macrophage bacterial infection. PLoS ONE 6: e28669. progenitor cell deficiency, and impaired neutrophil mobilization. Blood 84: 1737– 34. Das, B., S. S. Kashino, I. Pulu, D. Kalita, V. Swami, H. Yeger, D. W. Felsher, and 1746. A. Campos-Neto. 2013. CD271(+) bone marrow mesenchymal stem cells may 60. Hirai, H., P. Zhang, T. Dayaram, C. J. Hetherington, S. Mizuno, J. Imanishi, provideanichefordormantMycobacterium tuberculosis. Sci. Transl. Med 5: K. Akashi, and D. G. Tenen. 2006. C/EBPbeta is required for ‘emergency’ 170ra113. granulopoiesis. Nat. Immunol. 7: 732–739. 35. Bro-Jorgensen, K., and M. Volkert. 1972. Haemopoietic defects in mice infected 61. Panopoulos, A. D., L. Zhang, J. W. Snow, D. M. Jones, A. M. Smith, with lymphocytic choriomeningitis virus. 1. The enhanced x-ray sensitivity of virus K. C. El Kasmi, F. Liu, M. A. Goldsmith, D. C. Link, P. J. Murray, and infected mice. Acta Pathol. Microbiol. Scand. B Microbiol. Immunol. 80: 845–852. S. S. Watowich. 2006. STAT3 governs distinct pathways in emergency gran- 36. Petursson, S. R., P. A. Chervenick, and B. Wu. 1984. Megakaryocytopoiesis and ulopoiesis and mature neutrophils. Blood 108: 3682–3690. granulopoiesis after murine cytomegalovirus infection. J. Lab. Clin. Med. 104: 62. Cain, D. W., P. B. Snowden, G. D. Sempowski, and G. Kelsoe. 2011. Inflam- 381–390. mation triggers emergency granulopoiesis through a density-dependent feedback 37. Ferenczy, M. W., L. J. Marshall, C. D. Nelson, W. J. Atwood, A. Nath, K. Khalili, mechanism. PLoS ONE 6: e19957.

and E. O. Major. 2012. Molecular biology, epidemiology, and of 63. Winkler, I. G., N. A. Sims, A. R. Pettit, V. Barbier, B. Nowlan, F. Helwani, Downloaded from progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating I. J. Poulton, N. van Rooijen, K. A. Alexander, L. J. Raggatt, and J. P. Le´vesque. disease of the . Clin. Microbiol. Rev. 25: 471–506. 2010. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches 38. Jing, D., U. Oelschlaegel, R. Ordemann, K. Ho¨lig, G. Ehninger, H. Reichmann, and their depletion mobilizes HSCs. Blood 116: 4815–4828. T. Ziemssen, and M. Bornha¨user. 2010. CD49d blockade by in 64. Serbina, N. V., T. Jia, T. M. Hohl, and E. G. Pamer. 2008. -mediated patients with affects steady-state hematopoiesis and mobilizes defense against microbial pathogens. Annu. Rev. Immunol. 26: 421–452. progenitors with a distinct phenotype and function. Bone Marrow Transplant. 45: 65. Robben, P. M., M. LaRegina, W. A. Kuziel, and L. D. Sibley. 2005. Recruitment 1489–1496. of Gr-1+ monocytes is essential for control of acute toxoplasmosis. J. Exp. Med. 39. Apperley, J. F., C. Dowding, J. Hibbin, J. Buiter, E. Matutes, P. J. Sissons, 201: 1761–1769. M. Gordon, and J. M. Goldman. 1989. The effect of cytomegalovirus on he- 66. Kurihara, T., G. Warr, J. Loy, and R. Bravo. 1997. Defects in macrophage re- http://www.jimmunol.org/ mopoiesis: in vitro evidence for selective infection of marrow stromal cells. Exp. cruitment and host defense in mice lacking the CCR2 chemokine receptor. J. Exp. Hematol. 17: 38–45. Med. 186: 1757–1762. 40. Ganser, A., O. G. Ottmann, H. von Briesen, B. Vo¨lkers, H. Ru¨bsamen- 67. Serbina, N. V., and E. G. Pamer. 2006. Monocyte emigration from bone marrow Waigmann, and D. Hoelzer. 1990. Changes in the haematopoietic progenitor cell during bacterial infection requires signals mediated by chemokine receptor CCR2. compartment in the acquired immunodeficiency syndrome. Res. Virol. 141: 185– Nat. Immunol. 7: 311–317. 193. 68. de Bruin, A. M., S. F. Libregts, M. Valkhof, L. Boon, I. P. Touw, and 41. Isgro`, A., I. Mezzaroma, A. Aiuti, L. De Vita, F. Franchi, F. Pandolfi, C. Alario, M. A. Nolte. 2012. IFNg induces monopoiesis and inhibits neutrophil develop- F. Ficara, E. Riva, G. Antonelli, and F. Aiuti. 2000. Recovery of hematopoietic ment during inflammation. Blood 119: 1543–1554. activity in bone marrow from human immunodeficiency virus type 1-infected 69. Bockstal, V., P. Guirnalda, G. Caljon, R. Goenka, J. C. Telfer, D. Frenkel, patients during highly active antiretroviral therapy. AIDS Res. Hum. Retroviruses M. Radwanska, S. Magez, and S. J. Black. 2011. T. brucei infection reduces B

16: 1471–1479. lymphopoiesis in bone marrow and truncates compensatory splenic lymphopoiesis by guest on September 29, 2021 42. Thiebot, H., B. Vaslin, S. Derdouch, J. M. Bertho, F. Mouthon, S. Prost, G. Gras, through transitional B-cell . PLoS Pathog. 7: e1002089. P. Ducouret, D. Dormont, and R. Le Grand. 2005. Impact of bone marrow 70. Castro-Eguiluz, D., R. Pelayo, V. Rosales-Garcia, R. Rosales-Reyes, C. Alpuche- hematopoiesis failure on T-cell generation during pathogenic simian immunode- Aranda, and V. Ortiz-Navarrete. 2009. B cell precursors are targets for Salmonella ficiency virus infection in macaques. Blood 105: 2403–2409. infection. Microb. Pathog. 47: 52–56. 43. Thomsen, A. R., P. Pisa, K. Bro-Jørgensen, and R. Kiessling. 1986. Mechanisms of 71. Borrow, P., S. Hou, S. Gloster, M. Ashton, and L. Hyland. 2005. Virus infection- lymphocytic choriomeningitis virus-induced hemopoietic dysfunction. J. Virol. 59: associated bone marrow B cell depletion and impairment of humoral immunity to 428–433. heterologous infection mediated by TNF-alpha/LTalpha. Eur. J. Immunol. 35: 44. Boiko, J. R., and L. Borghesi. 2012. Hematopoiesis sculpted by pathogens: Toll- 524–532. like receptors and inflammatory mediators directly activate stem cells. Cytokine 57: 72. Sedger, L. M., S. Hou, S. R. Osvath, M. B. Glaccum, J. J. Peschon, N. van 1–8. Rooijen, and L. Hyland. 2002. Bone marrow B cell apoptosis during in vivo in- 45. Ciriza, J., H. Thompson, R. Petrosian, J. O. Manilay, and M. E. Garcı´a-Ojeda. fluenza virus infection requires TNF-alpha and lymphotoxin-alpha. J. Immunol. 2013. The migration of hematopoietic progenitors from the fetal liver to the fetal 169: 6193–6201. bone marrow: lessons learned and possible clinical applications. Exp. Hematol. 41: 73. Jyonouchi, H., P. W. Kincade, and R. A. Good. 1981. Immunosuppression of 411–423. marrow B lymphocytes by administration of Corynebacterium parvum in mice. J. 46. Nagai, Y., K. P. Garrett, S. Ohta, U. Bahrun, T. Kouro, S. Akira, K. Takatsu, and Immunol. 127: 2502–2507. P. W. Kincade. 2006. Toll-like receptors on hematopoietic progenitor cells 74. Nagaoka, H., G. Gonzalez-Aseguinolaza, M. Tsuji, and M. C. Nussenzweig. 2000. stimulate replenishment. Immunity 24: 801–812. Immunization and infection change the number of recombination activating gene 47. Welner, R. S., R. Pelayo, Y. Nagai, K. P. Garrett, T. R. Wuest, D. J. Carr, (RAG)-expressing B cells in the periphery by altering immature lymphocyte pro- L. A. Borghesi, M. A. Farrar, and P. W. Kincade. 2008. Lymphoid precursors are duction. J. Exp. Med. 191: 2113–2120. directed to produce dendritic cells as a result of TLR9 ligation during herpes in- 75. Ueda, Y., M. Kondo, and G. Kelsoe. 2005. Inflammation and the reciprocal fection. Blood 112: 3753–3761. production of granulocytes and lymphocytes in bone marrow. J. Exp. Med. 201: 48. Stein, B. L. 2012. The anemia of inflammation. J. Clin. Rheumatol. 18: 437–442. 1771–1780. 49. Nishimura, K., H. Nakaya, H. Nakagawa, S. Matsuo, Y. Ohnishi, and 76. Ueda, Y., K. Yang, S. J. Foster, M. Kondo, and G. Kelsoe. 2004. Inflammation S. Yamasaki. 2011. Effect of Trypanosoma brucei brucei on erythropoiesis in controls B lymphopoiesis by regulating chemokine CXCL12 expression. J. Exp. infected rats. J. Parasitol. 97: 88–93. Med. 199: 47–58. 50. Hunfeld, K. P., A. Hildebrandt, and J. S. Gray. 2008. Babesiosis: recent insights 77. O’Connor, B. P., V. S. Raman, L. D. Erickson, W. J. Cook, L. K. Weaver, into an ancient disease. Int. J. Parasitol. 38: 1219–1237. C. Ahonen, L. L. Lin, G. T. Mantchev, R. J. Bram, and R. J. Noelle. 2004. BCMA 51. Akinosoglou, K. S., E. E. Solomou, and C. A. Gogos. 2012. Malaria: a haemato- is essential for the survival of long-lived bone marrow plasma cells. J. Exp. Med. logical disease. Hematology 17: 106–114. 199: 91–98. 52. Mullarky, I. K., F. M. Szaba, L. W. Kummer, L. B. Wilhelm, M. A. Parent, 78. Chu, V. T., and C. Berek. 2013. The establishment of the plasma cell survival L. L. Johnson, and S. T. Smiley. 2007. Gamma suppresses erythro- niche in the bone marrow. Immunol. Rev. 251: 177–188. poiesis via interleukin-15. Infect. Immun. 75: 2630–2633. 79. Chu, V. T., A. Fro¨hlich, G. Steinhauser, T. Scheel, T. Roch, S. Fillatreau, J. J. Lee, 53. Hartmann, D. W., M. A. Entringer, W. A. Robinson, M. L. Vasil, C. J. Drebing, M. Lo¨hning, and C. Berek. 2011. Eosinophils are required for the maintenance of N. J. Morton, and L. True. 1981. Regulation of granulopoiesis and distribution of plasma cells in the bone marrow. Nat. Immunol. 12: 151–159. granulocytes in early phase of bacterial infection. J. Cell. Physiol. 109: 17–24. 80. Rodriguez Gomez, M., Y. Talke, N. Goebel, F. Hermann, B. Reich, and M. Mack. 54. Mungyer, G., L. G. Poels, C. Jerusalem, and R. Jerusalem. 1983. Plasmodium 2010. Basophils support the survival of plasma cells in mice. J. Immunol. 185: berghei: influence on granulopoiesis and macrophage production in BALB/c mice. 7180–7185. Exp. Parasitol. 56: 266–276. 81. Winter, O., K. Moser, E. Mohr, D. Zotos, H. Kaminski, M. Szyska, K. Roth, 55. Ojok, L., I. Kaeufer-Weiss, and E. Weiss. 2001. Bone marrow response to acute D. M. Wong, C. Dame, D. M. Tarlinton, et al. 2010. Megakaryocytes constitute and chronic Trypanosoma congolense infection in multimammate rats (Mastomys a functional component of a plasma cell niche in the bone marrow. Blood 116: coucha). J. Comp. Pathol. 124: 149–158. 1867–1875. The Journal of Immunology 33

82. Slifka, M. K., R. Antia, J. K. Whitmire, and R. Ahmed. 1998. Humoral immunity 97. Chakravarti, A., M. A. Raquil, P. Tessier, and P. E. Poubelle. 2009. Surface due to long-lived plasma cells. Immunity 8: 363–372. RANKL of Toll-like receptor 4-stimulated human neutrophils activates osteoclastic 83. Manz, R. A., A. Thiel, and A. Radbruch. 1997. Lifetime of plasma cells in the bone resorption. Blood 114: 1633–1644. bone marrow. Nature 388: 133–134. 98. Bussard, K. M., C. V. Gay, and A. M. Mastro. 2008. The bone microenvironment 84. Tokoyoda, K., and A. Radbruch. 2012. Signals controlling rest and reactivation of in metastasis; what is special about bone? Cancer Metastasis Rev. 27: 41–55. T helper memory lymphocytes in bone marrow. Cell. Mol. Life Sci. 69: 1609– 99. Fujisaki, J., J. Wu, A. L. Carlson, L. Silberstein, P. Putheti, R. Larocca, W. Gao, 1613. T. I. Saito, C. Lo Celso, H. Tsuyuzaki, et al. 2011. In vivo imaging of Treg cells 85. Tokoyoda, K., S. Zehentmeier, A. N. Hegazy, I. Albrecht, J. R. Gru¨n, providing immune privilege to the haematopoietic stem-cell niche. Nature 474: M. Lo¨hning, and A. Radbruch. 2009. Professional memory CD4+ T lymphocytes 216–219. preferentially reside and rest in the bone marrow. Immunity 30: 721–730. 100. Zeng, D., P. Hoffmann, F. Lan, P. Huie, J. Higgins, and S. Strober. 2002. Unique 86. Kondrack, R. M., J. Harbertson, J. T. Tan, M. E. McBreen, C. D. Surh, and patterns of surface receptors, cytokine secretion, and immune functions distinguish T cells in the bone marrow from those in the periphery: impact on allogeneic bone L. M. Bradley. 2003. regulates the survival and generation of marrow transplantation. Blood 99: 1449–1457. memory CD4 cells. J. Exp. Med. 198: 1797–1806. 101. Nguyen, V. H., R. Zeiser, and R. S. Negrin. 2006. Role of naturally arising 87. Herndler-Brandstetter, D., K. Landgraf, B. Jenewein, A. Tzankov, R. Brunauer, regulatory T cells in hematopoietic cell transplantation. Biol. Blood Marrow S. Brunner, W. Parson, F. Kloss, R. Gassner, G. Lepperdinger, and B. Grubeck- Transplant. 12: 995–1009. Loebenstein. 2011. Human bone marrow hosts polyfunctional memory CD4+ and 102. Zaiss, M. M., B. Frey, A. Hess, J. Zwerina, J. Luther, F. Nimmerjahn, K. Engelke, CD8+ T cells with close contact to IL-15-producing cells. J. Immunol. 186: 6965– G. Kollias, T. Hu¨nig, G. Schett, and J. P. David. 2010. Regulatory T cells protect 6971. from local and systemic bone destruction in . J. Immunol. 184: 7238– 88. Becker, T., S. Coley, E. Wherry, and R. Ahmed. 2005. Bone marrow is a preferred 7246. site for homeostatic proliferation of memory CD8 T cells. J. Immunol. 174: 1269– 103. Jang, E., W. S. Cho, M. L. Cho, H. J. Park, H. J. Oh, S. M. Kang, D. J. Paik, and 1273. J. Youn. 2011. Foxp3+ regulatory T cells control humoral by 89. Parretta, E., G. Cassese, P. Barba, A. Santoni, J. Guardiola, and F. Di Rosa. 2005. suppressing the development of long-lived plasma cells. J. Immunol. 186: 1546– CD8 cell division maintaining cytotoxic memory occurs predominantly in the 1553. bone marrow. J. Immunol. 174: 7654–7664. 104. Hall, A. O., D. P. Beiting, C. Tato, B. John, G. Oldenhove, C. G. Lombana, 90. Palendira, U., R. Chinn, W. Raza, K. Piper, G. Pratt, L. Machado, A. Bell, G. H. Pritchard, J. S. Silver, N. Bouladoux, J. S. Stumhofer, et al. 2012. The Downloaded from N. Khan, A. D. Hislop, R. Steyn, et al. 2008. Selective accumulation of virus- cytokines and interferon-g promote distinct Treg cell populations specific CD8+ T cells with unique homing phenotype within the human bone required to limit infection-induced . Immunity 37: 511–523. marrow. Blood 112: 3293–3302. 105. Oldenhove, G., N. Bouladoux, E. A. Wohlfert, J. A. Hall, D. Chou, L. Dos 91. Lo´pez, C. B., and T. Hermesh. 2011. Systemic responses during local viral Santos, S. O’Brien, R. Blank, E. Lamb, S. Natarajan, et al. 2009. Decrease of infections: type I IFNs sound the alarm. Curr. Opin. Immunol. 23: 495–499. Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal 92. Sato, T., N. Onai, H. Yoshihara, F. Arai, T. Suda, and T. Ohteki. 2009. Interferon infection. Immunity 31: 772–786. 106. Koch, M. A., G. Tucker-Heard, N. R. Perdue, J. R. Killebrew, K. B. Urdahl, and regulatory factor-2 protects quiescent hematopoietic stem cells from type I interferon-

D. J. Campbell. 2009. The transcription factor T-bet controls http://www.jimmunol.org/ dependent exhaustion. Nat. Med. 15: 696–700. homeostasis and function during type 1 inflammation. Nat. Immunol. 10: 595– 93. Hermesh, T., B. Moltedo, T. M. Moran, and C. B. Lo´pez. 2010. Antiviral in- 602. struction of bone marrow leukocytes during respiratory viral infections. Cell Host 107. Rossi, M. I., H. S. Dutra, M. C. El-Cheikh, A. Bonomo, and R. Borojevic. 1999. Microbe 7: 343–353. Extramedullar B lymphopoiesis in liver schistosomal : presence of the 94. de Bruin, A. M., O. Demirel, B. Hooibrink, C. H. Brandts, and M. A. Nolte. early stages and inhibition of the full B cell differentiation. Int. Immunol. 11: 509– 2013. Interferon-g impairs proliferation of hematopoietic stem cells in mice. Blood 518. 121: 3578–3585. 108. Rowland, S. L., K. Tuttle, R. M. Torres, and R. Pelanda. 2013. and 95. Baldridge, M. T., K. Y. King, N. C. Boles, D. C. Weksberg, and M. A. Goodell. cytokine receptor signals guide the development of the naı¨ve mature B cell rep- 2010. Quiescent haematopoietic stem cells are activated by IFN-gamma in re- ertoire. Immunol. Res. 55: 231–240. sponse to chronic infection. Nature 465: 793–797. 109. Stefanovic, S., F. Schuetz, C. Sohn, P. Beckhove, and C. Domschke. 2013. Bone 96. Teitelbaum, S. L. 2006. ; culprits in inflammatory osteolysis. Arthritis marrow microenvironment in cancer patients: immunological aspects and clinical

Res. Ther. 8: 201. implications. Cancer Metastasis Rev. 32: 163–178. by guest on September 29, 2021