Page 1 of 39 Diabetes

Restructuring of the gut microbiome by intermittent fasting prevents retinopathy

and prolongs survival in db/db mice

Eleni Beli1*, Yuanqing Yan2*, Leni Moldovan3, Cristiano P. Vieira4, Ruli Gao5, Yaqian

Duan6, Ram Prasad4, Ashay Bhatwadekar7, Fletcher A. White8, Steven Townsend9,

Luisa Chan10, Caitlin N. Ryan10, Daniel Morton10, Emil G. Moldovan11, FangI Chu12,

Gavin Y. Oudit13, Hartmut Derendorf14, Luciano Adorini15, Xiaoxin X. Wang16, Carmella

EvansMolina1,6,17, Raghavendra G. Mirmira1,6,17, Michael E. Boulton4, Mervin C.

Yoder1, Qiuhong Li18, Moshe Levi19, Julia V. Busik20, and Maria B. Grant4

1Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN,

USA

2Department of Neurosurgery, The University of Texas Health Science Center at

Houston, Houston, TX, USA

3Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA

4Department of Ophthalmology, University of Alabama, Birmingham, AL, USA

5Department of Genetics, The University of Texas MD Anderson Cancer Center,

Houston, TX, USA

6Department of Cellular and Integrative Physiology, Indiana University School of

Medicine, Indianapolis, IN, USA

7Department of Ophthalmology, Indiana University School of Medicine, Indianapolis,

IN, USA

8Department of Anesthesia, Indiana University, Indianapolis, IN, USA

9Department of Chemistry, Vanderbilt University, Nashville, TN, USA

10Second Genome, Inc., San Francisco, CA, USA

11Northeastern University, Boston, MA, USA

12Department of Radiation Oncology, University of California, Los Angeles, CA, USA

1

Diabetes Publish Ahead of Print, published online April 30, 2018 Diabetes Page 2 of 39

13Department of Medicine, University of Alberta, Edmonton, Alberta, Canada

14Department of Pharmaceutics, University of Florida, Gainesville, FL, USA

15Intercept Pharmaceuticals, New York, NY, USA

16Department of Medicine, University of Colorado, Denver, CO, USA

17Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA

18Department of Ophthalmology, University of Florida, Gainesville, FL, USA

19Department of Biochemistry and Molecular & Cellular Biology, Georgetown

University, Washington, DC, USA

20Department of Physiology, Michigan State University, East Lansing, MI, USA

*coauthors contributed equally to the manuscript

Correspondence:

Maria B. Grant M.D.

University of Alabama

VH 490 | 1720 2nd Ave S | Birmingham, AL 352940001

[email protected]

office: 2059968673

RUNNING TITLE: Intermittent fasting affects gut-retina axis in diabetes

Total Word Count of the Manuscript: 4,329

Figure count: 8

Keywords:

1. Intermittent Fasting

2. Diabetes

2 Page 3 of 39 Diabetes

3. Diabetic Retinopathy

4. Microbiome/ Microbiota

5. TUDCA

6. TGR5

3 Diabetes Page 4 of 39

Abstract

Intermittent fasting (IF) protects against the development of metabolic diseases and cancer, but whether it can prevent diabetic microvascular complications is not known. In db/db mice, we examined the impact of longterm IF on diabetic retinopathy

(DR). Despite no change in glycated hemoglobin, db/db mice on the IF regimen displayed significantly longer survival and a reduction in DR endpoints, including acellular capillaries and leukocyte infiltration. We hypothesized that IF mediated changes in the gut microbiota would produce beneficial metabolites and prevent the development of DR. Microbiome analysis revealed increased levels of and decreased

Bacteroidetes and Verrucomicrobia. Compared to db/db mice on adlibitum (AL) feeding, changes in the microbiome of the db/db mice on IF were associated with increases in gut mucin, goblet cell number and villi length and reductions in plasma peptidoglycan.

Consistent with the known modulatory effects of Firmicutes on bile acid (BA) metabolism, measurement of BAs demonstrated a significant increase of tauroursodeoxycholate (TUDCA), a neuroprotective BA, in db/db on IF but not in db/db on AL feeding. TGR5, the TUDCA receptor, was found in neural cells of the retina primary ganglion cells. Expression of TGR5 did not change with IF or diabetes.

However, IF reduced retinal TNFα mRNA, which is a key downstream target of TGR5 activation. Pharmacological activation of TGR5 using INT767 prevented DR in a second diabetic mouse model. These findings support the concept that IF prevents DR by restructuring the microbiota towards producing TUDCA and subsequent retinal protection by TGR5 activation.

4 Page 5 of 39 Diabetes

Introduction

Diabetesrelated blindness is a personal catastrophe for the individual and

affects over 4.2 million Americans (1). Although DR is commonly classified as a

microvascular complication of diabetes, it is now recognized that changes in the neural

cells of the retina occur early in the natural history of the disease and these changes

precede alterations in the microvasculature (2; 3). Current therapies for DR, such as

laser photocoagulation, injection of antiVEGF antibodies, or vitreoretinal surgery (4),

only treat latestage disease and selectively target the microvascular component of the

disease, while therapies for the early stages are limited to preventive strategies, such as

lifestyle changes.

Chronic calorie restriction (CR) and intermittent fasting (IF) represent non

pharmacological strategies for prevention and treatment of diseases such as diabetes,

obesity, and aging. However, CR can lead to nutrient deficiencies and fatigue (5; 6),

low metabolic rate and increased risk for subsequent obesity (7), infertility (5),

suppression of immunity (8) and increased susceptibility to viral infections (9; 10). IF

prevents the adverse effects of CR as it restricts feeding to only a part of the day or to

certain days per week. Thus, IF may be a more beneficial strategy compared to chronic

CR as it combines the beneficial effects elicited by fasting physiology and a recovery

period during the feeding phase which facilitates regeneration and promotes tissue

repair (11).

Accumulating evidence indicates that the gut microbiota contributes to key

physiological functions such as energy metabolism, metabolic signaling, and regulation

of integrity of the gut barrier (1214). Individuals with chronic diseases such as diabetes

exhibit altered bacterial composition (15), which likely not only influences glucose

metabolism but also, we reasoned, may contribute to development of complications.

5 Diabetes Page 6 of 39

Gut microbes have a symbiotic relationship with the host: they digest exogenous,

indigestible foods generating secondary metabolites that can affect host physiology,

and they metabolize endogenous substrates in the absence of food during prolonged

fasting.

An example of endogenous metabolites being converted to beneficial

secondary metabolites by the microbiome is the generation of bile acids (BAs). Primary

BAs generated by the liver are highly hydrophobic and can cause colitis; however,

intestinal convert them to more hydrophilic, less toxic, secondary BAs that are

potent agonists for nuclear receptors and have physiological functions. Using the db/db

mouse and an IF (every other day fasting) regimen for seven months, we showed that

IF prevented development of DR and increased generation of a secondary BA and

neuroprotective agent tauroursodeoxycholate (TUDCA). We further demonstrated the

relevance of TUDCA by demonstrating localization of TGR5, the receptor that mediates

TUDCA’s effects in the retina. In addition, the pharmacological activation of TGR5

using INT 767 in a second diabetes model prevented development of DR.

Materials and Methods

Animals and intermittent fasting. Male B6.BKS(D)Leprdb/J (stock number:000697)

Homozygous Leprdb/db were diabetic and heterozygous Leprdb/m were used as controls

(denoted as db/db and db/m hereafter). All mice were obtained from The Jackson

Laboratory (Bar Harbor, ME) and housed in the institutional animal care facilities at the

University of Florida (IACUC # 201106420) with strict 12h:12h light: dark cycle; all studies were repeated at Indiana University (IACUC #10604 and #11167). Blood glucose levels were tested every two weeks in db/db mice. Animals were considered as diabetic and used in the IF experiment if the serum glucose level was above 250 mg/dL on two consecutive measurements. The animals were fed ad lib (AL) before the

6 Page 7 of 39 Diabetes

IF was initiated at 4 months of age. The db/m and db/db mice were each divided in two

subgroups, AL feeding (control) and IF, wherein the animals were fasted for 24 hours

every other day for 7 months, beginning at night (Fig. 1A). Glycated hemoglobin was

measured using the A1CNow+ kit (Bayer HealthCare, Sunnyvale, CA) on the day prior

to euthanasia.

Acellular capillaries. The eyes were fixed in 2% formalin and trypsin digest was

performed for analysis of acellular capillaries as previously published (16).

Microbiome analysis. Fecal samples (n=180) were obtained by collection every 4

hours for 48 hours. Genomic DNA was isolated from ~0.1 g using the PowerSoil® DNA

Isolation Kit (MO Bio, Carlsbad, CA), according to manufacturer’s protocol. DNA was

sent to Second Genome, Inc. (South San Francisco, CA) for 16S rRNA gene V4

sequencing with the MiSeq platform. The full report and statistical analysis from

Second Genome is available upon request.

Colon morphometric analysis. Colon tissue was collected at termination of the

experiment, fixed in 10% buffered formalin and paraffinembedded according to

standard procedures at the IUPUI Pathology Core. Ten m sections were cut and

stained by either hematoxylin and eosin or periodic acid Schiff (PAS) for mucin. Entire

sections were scanned with an Axio Scan Z1 (Zeiss) and a 20X objective.

Morphometric analysis of goblet cells, villi length and muscularis thickness was

undertaken using a Lumenera Infinity 12C camera and NIS Elements software (Nikon

Instruments, Melville, NY).

Peptidoglycan ELISA. Plasma samples were diluted 1:10 and analyzed for

7 Diabetes Page 8 of 39

peptidoglycan levels using the mouse peptidoglycan ELISA kit (MBS263268,

Mybioscource Inc, San Diego, CA) according to manufacturer’s instructions.

Bile acid analysis. At the termination of the experiment, whole blood was collected by cardiac puncture and transferred in EDTA coated tubes. Plasma was separated and frozen at 80oC. Samples were sent to Metabolon Inc (Morrisville, NC) and global metabolic analysis was performed with their Metabolon Platform using a Waters

ACQUITY ultraperformance liquid chromatography (UPLC) and a Thermo Scientific Q

Exactive high resolution/accurate mass spectrometer interfaced with a heated electrospray ionization (HESIII) source and Orbitrap mass analyzer operated at 35,000 mass resolution. The full report and statistical analysis from Metabolon is available upon request.

Quantitative real-time-PCR (qRT-PCR). Total RNA was extracted from mouse retinal tissue using RNeasy Mini kit (Qiagen, Germantown, MD) and reverse transcription was performed according previous published methods (17). Validated primers for Gpbar1

(TGR5), Tnf, and endogenous controls B2m and Ppia, were purchased from Qiagen and qRTPCR was carried out using SYBR Green (Qiagen).

INT 767 treatment of diabetic animals. 8week old male DBA2/J mice were obtained from The Jackson Laboratory and maintained on a 12h:12h light:dark cycle. Mice were injected with streptozotocin (STZ; SigmaAldrich, St. Louis, MO) intraperitoneal [40 mg/kg made freshly in 50 mM sodium citrate buffer, pH 4.5 (vehicle)] for five consecutive days, or with vehicle only. Blood glucose levels were measured 1 week after the last STZ injection and mice with glucose levels >250 mg/dL on two separate

8 Page 9 of 39 Diabetes

days were considered diabetic. DBA/2J mice were fed with a Western diet (WD, 21%

milk fat, 0.2 % cholesterol, TD88137) obtained from HarlanTeklad (Madison, WI) after

diabetes onset in the STZ groups and were treated for 8 weeks with either WD or WD

containing INT767 (30 mg/kg bw/day) (Intercept Pharmaceuticals, New York, NY)

(18).

Immunofluorescence staining. Enucleated eyes were fixed in 4% paraformaldehyde

overnight at 4oC. The next day, eyes were incubated with 30% sucrose for 48 hours and

then quickly frozen in optical cutting temperature (OCT) compound. Retinal cross

sections (12 m) were cut at 20oC. The following antibodies were used for

immunostaining: rabbit monoclonal to Iba (Wako, 01919741, 1:500), rat monoclonal to

CD45 (clone 30F11, R&D Systems, 1:50), rabbit polyclonal to GPCR TGR5 (ABCAM,

ab72608, 1:250), monoclonal murine anti NeuN, (clone A60, EMD Millipore,

MAB377MI, 1:250). Sections were preincubated with 5% goat serum (Invitrogen) in

PBS for 1h, followed by incubation with primary antibodies (in 1% normal goat serum)

overnight at 4oC. As secondary antibody, Alexa Fluor 488 was used for TGR5 and Iba1+

and Alexa Fluor 594 was used for NeuN and CD45 (Invitrogen). Positive cells were

counted from 35 sections at 100 m intervals for each eye, on at least four images per

section at 20X magnification. Retinal sections were imaged using a confocal scanning

laser microscope (ZEISS LSM 700 confocal microscope system with Axio Observer; Carl

Zeiss Mditec, Jena, Germany) and colocalization was analyzed by using Zen lite

software.

Statistical analysis. Survival curve was plotted by KaplanMeier method and the

significance of the differences was determined by logrank test. The differences in gene

expression, metabolites and acellular capillaries were analyzed by twoway ANOVA

followed by Tukey’s honest significant difference post hoc test (GraphPad Prism, La

9 Diabetes Page 10 of 39

Jolla, CA). Statistical analysis methods for microbiome and metabolites are available

upon request.

Results

IF improved survival without impacting glycated hemoglobin levels.

Four monthold db/m and db/db mice were fed either an adlib diet (AL) or were

fasted every other 24hour interval (IF) for 7 months (Fig. 1A). Glycated hemoglobin

levels were not affected by the IF regimen and remained higher in both db/db cohorts

compared to controls (Fig. 1B). However, despite no change in glycated hemoglobin,

the survival rate increased significantly in db/db mice on the IF regimen (db/dbIF)

compared to db/db mice on AL feeding (db/dbAL) (Fig. 1C).

IF prevented development of acellular capillaries and infiltration of inflammatory

cells in the retina of db/db mice

We next examined the impact of IF on development of DR in db/db mice by

enumerating acellular capillaries (Fig. 2A-D)(19). As expected, db/dbAL showed

increased numbers of acellular capillaries (Fig. 2C) compared to agematched db/m

AL (Fig. 2A). However, db/dbIF mice (Fig. 2D, E) did not demonstrate an increase in

acellular capillaries. Since increases in retinal levels of inflammatory cells are another

feature of DR, cryosections were examined for changes in activated microglia using

Iba1 staining and differences in infiltrating hematopoietic cells using CD45 staining.

Db/dbAL mice showed a significant increase in Iba1+ cells compared to db/mAL mice and db/dbIF mice (Fig. 2F). CD45+ cell infiltration was significantly increased in the db/dbAL mice compared to the db/mAL mice and db/dbIF mice (Fig. 2G). Overall, IF protected db/db mice from developing histological features of DR.

10 Page 11 of 39 Diabetes

Long-term IF altered the gut microbiome composition

The composition and diversity of the microbiome is integrated with the pathophysiology

of diabetes. For example, certain bacteria are shown to induce diabetes (20), while

others alleviate it (21).Thus, we hypothesized that the beneficial effects of IF on DR

would, at least in part, be mediated through changes in microbiota composition. Fecal

samples were collected and genomic DNA sequencing was used to discover and

distinguish various bacterial taxa. A total of 1,363 filtered operational taxonomic units

(OTUs) were detected. Sample richness and Shannon diversity (Fig. S1A-F) were not

changed by the implementation of IF but beta diversity, a measure of the between

sample diversity, showed clear differences between db/dbAL and db/mAL mice (Fig.

3A), db/mIF vs. db/mAL (Fig. 3B), and db/dbIF vs. db/dbAL (Fig. 3C). Principal

component analysis of bacteria also demonstrated a clear separation of the four main

groups (db/mAL, db/dbAL, db/mIF and db/dbIF). The AL groups were closer to each

other while the IF groups diverged more (Fig. 3D). This evidence suggests that IF

restructured a distinct microbiome in the diabetic compared to control mice.

The most represented phyla were Bacteroidetes, Firmicutes, Verrucomicrobia,

Tenericutes, Actinobacteria, and Proteobacteria. The relative proportions of these varied

dramatically among the experimental groups but the majority of bacteria belonged to the

Bacteroidetes, Firmicutes and Verrucomicrobia phyla (Fig. 3E). The ratio of

Firmicutes/Bacteroidetes (F/B) has been widely used as an indicator of changes in the

microbiome with obesity (22). The F/B ratio was not altered in the db/mIF mice (Fig.

3E); however, it was dramatically increased in the db/dbIF mice due to a significant

increase of Firmicutes and a significant reduction of Bacteroidetes (Fig. 3E). Apart from

changes observed in the F/B ratio, Verrucomicrobia also changed significantly with IF

11 Diabetes Page 12 of 39

showing an increase in db/mIF and a reduction in db/dbIF (Fig. 3E). Overall, these data indicate that IF resulted in a more dramatic restructuring of the microbiota composition in the diabetic mice with a significant expansion of Firmicutes at the

expense of Bacteroidetes and Verrucomicrobia.

To understand how the microbiota composition was altered at the genus level,

the differential taxa in each group were assessed (Fig. 4A-C). Among the differential taxa between db/mAL and db/dbAL cohorts, the db/dbAL mice were enriched in

Lactobacillus, Bifidobacterium, Bacteroides and Akkermansia, and were deficit in species of Oscillospira, and Ruminococcus (Fig. 4A). The IF regimen in the db/db mice caused enrichment of species of the genus Lactobacillus, Oscillospira and

Ruminococcus and, reduction of species of the genus Akkermansia, Bacteroides and

Bifidobacterium (Fig. 4B-C). Interestingly, the difference in the microbiota between fed vs. fasting state in the IF cohorts was very small, limited to only 24 bacteria in db/dbIF mice and 69 bacteria in db/mIF mice (Fig. S2. A-B). Yet, we identified certain common species that were increased by fasting in both db/mIF and db/dbIF cohorts, mostly species in the and Ruminococcaceae families with representatives from Oscillospira and Ruminococcus genera. On the other hand, species of the genus

Bifidobacterium were commonly reduced by the IF diet. A list of the common and unique enriched bacterial species is shown in Fig. S2C and the common or uniquely reduced bacteria is shown in Fig. S2D.

IF is known to influence diurnal rhythms and selected microbes in the gut exhibit diurnal changes (23; 24). We investigated which taxa displayed diurnal patterns in healthy db/mAL mice and whether these taxa were affected by either the presence of diabetes or IF (25) . Surprisingly, we found only 34 OTUs showing diurnal cycling.

Among these were members of Lachnospiraceae and Ruminococcaceae families and

12 Page 13 of 39 Diabetes

specifically, species of Oscillospira and Ruminococcus. Examples of bacteria with

diurnal regulation is shown in Fig. S3.

Although there were few taxa differentially regulated by feeding / fasting or

diurnal cycling, the evolutionary pressure of fasting and refeeding in combination with

the host metabolic response to fasting led to the distinct restructuring of the microbiota in

the db/mIF and the db/dbIF mice. Members of the order Clostridiales (Fig. 4D) and

members of the Ruminococcaceae (Fig. 4E) and Lachnospiraceae families (Fig. 4F)

were uniquely equipped to adapt, expand and survive under IF conditions in db/db but

not in db/m mice. Lactobacillus (Fig. 4G) was enriched in both db/mIF and db/dbIF,

while the genus Bifidobacterium (Fig. 4H) and Clostridium (Fig. 4I) was reduced in both

groups. On the other hand, Oscillospira (Fig. 4J), Ruminococcus (Fig. 4K), and

Turicibacter (Fig. 4L) were enriched specifically in the db/dbIF, while Bacteroides (Fig.

4M), Akkermansia (Fig. 4N) and Allobaculum (Fig. 4O) were reduced only in the db/db

IF mice. We reasoned that these unique changes in the microflora of the db/dbIF mice

could promote the integrity of the gut barrier.

IF promoted restoration of diabetes-induced gut pathology

As disruption of the gut barrier integrity is an important mechanism of disease,

we examined whether IF beneficially impacted colon morphology as measured by goblet

cell number and villi length (Fig. 5). Db/dbAL had reduced numbers of goblet cells (Fig.

5 B, E) and reduced villi length (Fig. 5G, J). However, IF resulted in preservation of

colon morphology in the db/dbIF mice, as goblet cell number (Fig. 5D, E) and villi length

(Fig.5I, J) were similar to nondiabetic mice. No changes with diabetes or IF were

observed in the thickness of the muscularis (Fig. 5K-O).

13 Diabetes Page 14 of 39

Increases in bacterial products in plasma are indicative of intestinal barrier dysfunction or disruption. We measured peptidoglycan in plasma samples from our cohorts. As shown in Fig. 5P, db/dbAL demonstrated a significant increase in plasma peptidoglycan levels supportive of increased gut permeability. IF resulted in a decrease in gut permeability when db/dbIF mice were in the fasted state as there was a significant reduction of circulating peptidoglycan levels. However, when the db/dbIF mice were in the fed state this potentially beneficial change was lost.

IF altered bile acid metabolism

We next tested whether the microbial changes induced by IF could generate

beneficial metabolites. Bile acids are known to alter the gut microbiome composition

(26), and conversely, the microbiome is known to alter the bile acid pool (27). Thus, we

reasoned that the observed microbiome changes induced by IF would impact BA

metabolism in the db/db mice. Metabolic analysis of plasma from the experimental

cohorts identified the primary BA cholate but not chenodeoxycholate. A list of bile acids

found in the plasma is shown in Table S1.

Cholate was increased in the db/dbAL mice compared to db/mAL mice and was

reduced in the db/dbIF mice (Fig. 6A). Deoxycholate, the secondary BA that is

produced by the activities of 7αdehydroxylase, an enzyme found in intestinal bacteria,

was moderately increased with IF especially during the fasting period (Fig. 6B)

supporting intestinal transformation of BAs. However, the ratio of conjugated to

unconjugated BA was increased in the db/dbIF mice compared to db/mIF during

feeding, suggesting that there may be a reduction of bacteria that express bile salt

hydrolase (BSH) activity required to deconjugate primary and secondary bile acids in the

db/dbIF mice. A significant reduction of Bifidobacterium (Fig. 4H), Clostridium (Fig 4I)

14 Page 15 of 39 Diabetes

and Bacteroides (Fig. 4M) was observed and these bacteria are known to express BSH,

which may be responsible, in part, for these observed changes in levels of conjugated to

unconjugated secondary BAs.

An increase in BA products indicative of 7α and 7βhydroxysteroid

dehydrogenase (HSDH) enzymatic activity was also found. Taurochenodeoxycholate

(TCDCA) was identified and significantly increased in the db/dbIF mice (Fig. 6C).

TCDCA is converted to TUDCA by the actions of both 7α and 7βHSDH (28) and

TUDCA was significantly increased in the plasma of db/dbIF mice (Fig. 6D). As taurine

conjugated secondary bile acids are potent activators of TGR5 (29), we hypothesize that

the beneficial effects we observed following IF were due to an increase in TUDCA

synthesis in the gut.

TGR5 activation reduces acellular capillaries and inflammation in the diabetic

retina.

To determine whether TUDCA could mediate beneficial effects in the retina, we

examined TGR5 expression. TGR5 was localized primarily to the ganglion cell layer of

the retina (Fig. 7A). Neither diabetes nor the IF regimen changed the localization or

expression of TGR5 protein (Fig. 7B-F) as assessed by immunohistochemistry nor

changed TGR5 mRNA levels (Fig. 7G). However, TNFα, a downstream target of TGR5

was reduced significantly by IF in the diabetic cohort (Fig. 7H). To further support the

role of retinal TGR5, we asked if pharmacological activation of TGR5 would prevent DR.

Previously, we characterized the novel highly potent semisynthetic bile acid derivative

and dual TGR5/FXR agonist INT767 (18). INT767 was tested in a second diabetes

model, the DBA/2J mice treated with streptozotocin and placed on a high fat diet. This

model exhibits a more rapid progression of the vascular and inflammatory endpoints

15 Diabetes Page 16 of 39

associated with DR (30) and diabetic nephropathy (31), and replicates the natural history

and metabolic characteristics of human metabolic syndrome and Type 2 diabetes (31;

32). INT767 treatment of diabetic mice resulted in a marked reduction in the number of

acellular capillaries (Fig. 7C) and reduced numbers of CD11b+ macrophages (Fig. 7D),

CD45+ leukocytes (Fig. 7E) and activated Iba1+ microglia within the retina (Fig. 7F).

Discussion

The salient findings of this study suggest that restructuring of the microbiome by IF favors generation of secondary BAs, such as TUDCA, eliciting protective effects in the retina (Fig. 8). Development of acellular capillaries, activation of retinal microglia

and infiltration of peripheral immune cells into the retina were all reduced by IF in the

db/db mice. IF produced a reconfiguration of the microbiota in the db/db mice resulting

in an enrichment in Firmicutes and a reduction in Bacteroides. Interestingly, this was

not observed for db/m mice undergoing IF, indicating that the diabetic

microenvironment benefited Firmicutes expansion. High F/B ratio has been associated

with healthy obesity (33; 34) and increased capacity for harvesting energy (22). In

addition, a high F/B ratio has been implicated in regulating fat storage (35) and is

consistent with our findings wherein IF did not change the weight of the db/db mice but

promoted a healthier obese phenotype with increased longevity.

Another example of a healthy obese phenotype associated with expansion of

Firmicutes is the case of black bears. During the summer when active and feeding,

these bears experience a healthy weight gain and an expansion of Firmicutes (36). Our

study provides support for the clinical observation that some obese individuals are

metabolically healthy (ranging from 3 to 57%, depending on the study) (13) and may

have a gut microbiota structure that contributes to this beneficial outcome.

16 Page 17 of 39 Diabetes

BAs are significant modulators of the gut microbiome as they exhibit antimicrobial

activities and can inhibit bacteria outgrowth (37). Members of the Firmicutes family are

expanded in high BA conditions, while members of Bacteroides and Actinobacteria

(Bifidobacterium) are reduced (26). This finding is particularly relevant, as it appears that

the db/dbAL mice have increased cholate compared to db/mAL. The altered BA

metabolism observed in the diabetic mouse effectively drove the restructuring of the

diabetic microbiota towards the unique composition with high F/B ratio.

The observed changes in the gut microbiome of db/dbIF mice were deemed

advantageous for gut and intestinal barrier integrity. We utilized plasma peptidoglycan as

a surrogate for gut barrier function. Peptidoglycan levels were increased in diabetes and

the IF diet reduced the levels during fasting. This suggests that gut barrier integrity is

compromised by diabetes and that the IF regimen (during the fasting day) can improve

it. Our results are in agreement with a recent study demonstrated that hyperglycemia

drives the increased intestinal permeability observed in diabetes (38). In our model,

when diabetic mice are under IF, glucose remains elevated during the feeding day of the

IF regimen, but is reduced during the fasting phase of the IF regimen. Furthermore, the

composition of the leaked bacteria maybe important in driving the low grade systemic

inflammation in diabetes (39). Peptidoglycan is a component of grampositive bacteria

and endotoxin is mostly derived from gramnegative bacteria. Firmicutes are gram

positive, while Bacteroides are gram negative (40). Thus, our results suggest that the

reduction of peptidoglycan occurring in the db/db mouse on the IF regimen signals a

transient restoration/improvement in gut integrity and could also lead to a transient

reduction in endotoxemia. However, this remains to be tested.

In diabetes, cholesterol and BA metabolism is altered (41). Thus, we focused our

efforts in understanding how remodeling of the microbiota by IF affected

17 Diabetes Page 18 of 39

biotransformation of the BA pool. Primary BAs are synthesized from cholesterol in the liver and act as detergents for the emulsification of dietary fats and as endocrine hormones for the regulation of glucose and lipid metabolism (42; 43). Conjugated or unconjugated primary BAs enter the gallbladder and are released in the small intestine, where BA are structurally modified into various secondary BAs that are less toxic to the host and can exert multiple hormonal regulatory roles (44). αHSDH and 7βHSDH are two key enzymes that drive the efficient biosynthesis of secondary BAs such as TUDCA from taurochenodeoxycholate (TCDCA) (28). Thus, the change in microbiome composition affects the enzymatic activity of these pathways and alters the BA pool.

Of interest is the observation that bear bile has long been used in Chinese traditional medicine for the treatment of liver, gallbladder, heart and eye problems.

Bear bile is highly enriched in TUDCA (45). Recently the bear gut microbiome was used to isolated new, undiscovered 7βHSDH enzymes for more efficient production of

TUDCA (28). Little is known regarding the protective effects of TUDCA in the diabetic retina (4652). However, this hydrophilic BA can cross the blood brain barrier, and likely can cross the blood retinal barrier. In support of the ability of circulating TUDCA to reach the retina and act on retinal cells are the observations that TUDCA can inhibit photoreceptor apoptosis (47; 48; 51), improve retinal pigment epithelial function (49), inhibit proliferation of human retinal endothelial cells (53), promote the recruitment of endothelial progenitor cells (54) and protect against DR (53). At the molecular level,

TUDCA has been shown to inhibit apoptosis through reduction of endoplasmic reticulum stress (ER) (55) and modulation of the unfolded protein response (56).

TUDCA also inhibits mitochondrial membrane depolarization (57). Receptor activation of TGR5 by TUDCA may also decrease the production of proinflammatory cytokines and induce antiinflammatory responses (58). Although TGR5 protein expression did not change with IF treatment, our studies would support that TGR5 activation is central

18 Page 19 of 39 Diabetes

to the retinal protection observed, possible through eliciting antiinflammatory effects,

as INT767 effectively prevented DR and reduced infiltration of inflammatory cells in

the retina of STZ/WD mice. While INT 767 is known as a dual agonist of TGR5 and

FXR, the absence of FXR in the retina (59) allowed us to utilize this highly potent agent

as a selective TGR5 agonist in the retina. However, additional studies will be required

to establish a direct link between TGR5 activation and IF.

Our study has certain limitations that must be acknowledged. First, fecal rather

than mucosal bacterial samples were utilized. As such, our analysis may not be

representative of the entire microbiome. However, fecal sampling is typically used in

human studies. Second, we used plasma samples rather than cecum samples for the

analysis of metabolites. Our intention was to identify microbial metabolites that could be

found in peripheral circulation. Specifically, those that could target the retina. Third, we

used the db/db model as a model of type 2 diabetes to study the IF effects on

microbiota. The STZ/WD model of diabetes was used to establish the link between

TGR5 activation and prevention of DR. The STZ/WD represents an accelerated model of

DR that histologically replicates the DR seen in other diabetes models (30) and also has

the natural history and metabolic characteristics of human metabolic syndrome and Type

2 diabetes (31; 32). Finally, although we show an association between increases in

TUDCA and prevention of retinopathy, additional studies using antibiotics or fecal

transfers will be required to demonstrate this conclusively.

In sum, our study shows a novel relationship between changes in the microbiota,

IF and increases in TUDCA. IF likely provides a way to change BA metabolism in favor

of increasing the endogenous generation of TUDCA (Fig. 8). This work highlights

several findings with potential clinical significance. Diet interventions, such as IF, in

diabetes may result in a unique and highly beneficial microbiome response. IF

19 Diabetes Page 20 of 39

represents a physiological way to enhance production of the neuroprotective TUDCA.

Finally, we identify a role for TGR5 and bile acid signaling in the retina that, to our

knowledge, was not previously known and may represent a new therapeutic target for

DR.

Acknowledgements: Studies were supported by the NIH (EY07739, EY12601,

EY025383, EY023629 and HL110170) to MBG, DK093954 to CEM and by the JDRF (3

APF2017396AN) to EB. We would like to thank Tatiana E. Salazar, Dung V. Nguyen,

James M. Dominguez II, Matthew Richardson, Shakir Hasan Hindi, Harkeerat Dhami,

Jared Andre Smith, Joshua Hayes Thomas, Emily Francesca Hutchinson for their technical help with the animal experiments.

Author Contributions: EB, YY, JVB and MBG designed experiments. EB, YY, LM,

CPV, YD and XXW performed experiments. EB, YY, LM, LC, CNR, DM, EGM and FC analyzed the data. EB, YY, LM, FAW, HD, GYO, RGM, QL, ML, MEB, JVB, and MBG discussed the results and interpreted the data. EB, YY, LM, GYO, LA, CEM, FAW,

MEB, MCY, ML and MBG wrote and revised the paper. Dr. Maria B. Grant is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

LC, RNC, and DM are associated with Second Genome Inc. LA is associated with

Intercept Pharmaceuticals. No conflict of interest is declared by the rest of the authors.

20 Page 21 of 39 Diabetes

References

1. Zhang X, Saaddine JB, Chou CF, Cotch MF, Cheng YJ, Geiss LS, Gregg EW, Albright AL, Klein BE, Klein R: Prevalence of diabetic retinopathy in the United States, 20052008. Jama 2010;304:649656

2. Antonetti DA, Klein R, Gardner TW: Diabetic retinopathy. The New England journal of medicine 2012;366:12271239

3. Gardner TW, Davila JR: The neurovascular unit and the pathophysiologic basis of diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2017;255:16

4. Stewart MW: Treatment of diabetic retinopathy: Recent advances and unresolved challenges. World J Diabetes 2016;7:333341

5. Fairburn CG, Harrison PJ: Eating disorders. Lancet 2003;361:407416

6. White AM, Johnston CS, Swan PD, Tjonn SL, Sears B: Blood ketones are directly related to fatigue and perceived effort during exercise in overweight adults adhering to lowcarbohydrate diets for weight loss: a pilot study. J Am Diet Assoc 2007;107:1792 1796

7. Fothergill E, Guo J, Howard L, Kerns JC, Knuth ND, Brychta R, Chen KY, Skarulis MC, Walter M, Walter PJ, Hall KD: Persistent metabolic adaptation 6 years after "The Biggest Loser" competition. Obesity (Silver Spring) 2016;24:16121619

8. Clinthorne JF, Beli E, Duriancik DM, Gardner EM: NK cell maturation and function in C57BL/6 mice are altered by caloric restriction. Journal of immunology 2013;190:712 722

9. Gardner EM, Beli E, Clinthorne JF, Duriancik DM: Energy intake and response to infection with influenza. Annu Rev Nutr 2011;31:353367

10. Ritz BW, Aktan I, Nogusa S, Gardner EM: Energy restriction impairs natural killer cell function and increases the severity of influenza infection in young adult male C57BL/6 mice. J Nutr 2008;138:22692275

11. Cheng CW, Villani V, Buono R, Wei M, Kumar S, Yilmaz OH, Cohen P, Sneddon JB, Perin L, Longo VD: FastingMimicking Diet Promotes Ngn3Driven betaCell Regeneration to Reverse Diabetes. Cell 2017;168:775788 e712

12. Clavel T, Desmarchelier C, Haller D, Gerard P, Rohn S, Lepage P, Daniel H: Intestinal microbiota in metabolic diseases: from bacterial community structure and functions to species of pathophysiological relevance. Gut Microbes 2014;5:544551

13. Rial SA, Karelis AD, Bergeron KF, Mounier C: Gut Microbiota and Metabolic Health: The Potential Beneficial Effects of a Medium Chain Triglyceride Diet in Obese Individuals. Nutrients 2016;8

14. Levy M, Thaiss CA, Zeevi D, Dohnalova L, ZilbermanSchapira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y, PevsnerFischer M, Shapiro H, Christ A, Harmelin A,

21 Diabetes Page 22 of 39

Halpern Z, Latz E, Flavell RA, Amit I, Segal E, Elinav E: MicrobiotaModulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome Signaling. Cell 2015;163:14281443

15. Tilg H, Moschen AR: Microbiota and diabetes: an evolving relationship. Gut 2014;63:15131521

16. Bhatwadekar AD, Yan Y, Qi X, Thinschmidt JS, Neu MB, Li Calzi S, Shaw LC, Dominiguez JM, Busik JV, Lee C, Boulton ME, Grant MB: Per2 mutation recapitulates the vascular phenotype of diabetes in the retina and bone marrow. Diabetes 2013;62:273282

17. Kono T, Ahn G, Moss DR, Gann L, ZarainHerzberg A, Nishiki Y, Fueger PT, Ogihara T, EvansMolina C: PPARgamma activation restores pancreatic islet SERCA2 levels and prevents betacell dysfunction under conditions of hyperglycemic and cytokine stress. Mol Endocrinol 2012;26:257271

18. Rizzo G, Passeri D, De Franco F, Ciaccioli G, Donadio L, Rizzo G, Orlandi S, Sadeghpour B, Wang XX, Jiang T, Levi M, Pruzanski M, Adorini L: Functional characterization of the semisynthetic bile acid derivative INT767, a dual farnesoid X receptor and TGR5 agonist. Mol Pharmacol 2010;78:617630

19. Hammes HP, Lin J, Renner O, Shani M, Lundqvist A, Betsholtz C, Brownlee M, Deutsch U: Pericytes and the pathogenesis of diabetic retinopathy. Diabetes 2002;51:31073112

20. Manfredo Vieira S, Hiltensperger M, Kumar V, ZegarraRuiz D, Dehner C, Khan N, Costa FRC, Tiniakou E, Greiling T, Ruff W, Barbieri A, Kriegel C, Mehta SS, Knight JR, Jain D, Goodman AL, Kriegel MA: Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science 2018;359:11561161

21. Zhao L, Zhang F, Ding X, Wu G, Lam YY, Wang X, Fu H, Xue X, Lu C, Ma J, Yu L, Xu C, Ren Z, Xu Y, Xu S, Shen H, Zhu X, Shi Y, Shen Q, Dong W, Liu R, Ling Y, Zeng Y, Wang X, Zhang Q, Wang J, Wang L, Wu Y, Zeng B, Wei H, Zhang M, Peng Y, Zhang C: Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018;359:11511156

22. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI: An obesity associated gut microbiome with increased capacity for energy harvest. Nature 2006;444:10271031

23. Thaiss CA, Levy M, Korem T, Dohnalova L, Shapiro H, Jaitin DA, David E, Winter DR, GuryBenAri M, Tatirovsky E, Tuganbaev T, Federici S, Zmora N, Zeevi D, Dori Bachash M, PevsnerFischer M, Kartvelishvily E, Brandis A, Harmelin A, Shibolet O, Halpern Z, Honda K, Amit I, Segal E, Elinav E: Microbiota Diurnal Rhythmicity Programs Host Transcriptome Oscillations. Cell 2016;167:14951510 e1412

24. Thaiss CA, Zeevi D, Levy M, ZilbermanSchapira G, Suez J, Tengeler AC, Abramson L, Katz MN, Korem T, Zmora N, Kuperman Y, Biton I, Gilad S, Harmelin A, Shapiro H, Halpern Z, Segal E, Elinav E: Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell 2014;159:514529

22 Page 23 of 39 Diabetes

25. Hughes ME, Hogenesch JB, Kornacker K: JTK_CYCLE: an efficient nonparametric algorithm for detecting rhythmic components in genomescale data sets. J Biol Rhythms 2010;25:372380

26. Islam KB, Fukiya S, Hagio M, Fujii N, Ishizuka S, Ooka T, Ogura Y, Hayashi T, Yokota A: Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology 2011;141:17731781

27. Gerard P: Metabolism of cholesterol and bile acids by the gut microbiota. Pathogens 2013;3:1424

28. Song C, Wang B, Tan J, Zhu L, Lou D: Discovery of tauroursodeoxycholic acid biotransformation enzymes from the gut microbiome of black bears using metagenomics. Scientific reports 2017;7:45495

29. Sato H, Macchiarulo A, Thomas C, Gioiello A, Une M, Hofmann AF, Saladin R, Schoonjans K, Pellicciari R, Auwerx J: Novel potent and selective bile acid derivatives as TGR5 agonists: biological screening, structureactivity relationships, and molecular modeling studies. J Med Chem 2008;51:18311841

30. Hazra S, Rasheed A, Bhatwadekar A, Wang X, Shaw LC, Patel M, Caballero S, Magomedova L, Solis N, Yan Y, Wang W, Thinschmidt JS, Verma A, Li Q, Levi M, Cummins CL, Grant MB: Liver X receptor modulates diabetic retinopathy outcome in a mouse model of streptozotocininduced diabetes. Diabetes 2012;61:32703279

31. Wang XX, Jiang T, Shen Y, Caldas Y, MiyazakiAnzai S, Santamaria H, Urbanek C, Solis N, Scherzer P, Lewis L, Gonzalez FJ, Adorini L, Pruzanski M, Kopp JB, Verlander JW, Levi M: Diabetic nephropathy is accelerated by farnesoid X receptor deficiency and inhibited by farnesoid X receptor activation in a type 1 diabetes model. Diabetes 2010;59:29162927

32. Ghelani H, RazmovskiNaumovski V, Nammi S: Chronic treatment of (R)alphalipoic acid reduces blood glucose and lipid levels in highfat diet and lowdose streptozotocin induced metabolic syndrome and type 2 diabetes in SpragueDawley rats. Pharmacol Res Perspect 2017;5:e00306

33. Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI: Obesity alters gut microbial ecology. Proceedings of the National Academy of Sciences of the United States of America 2005;102:1107011075

34. Ley RE, Turnbaugh PJ, Klein S, Gordon JI: Microbial ecology: human gut microbes associated with obesity. Nature 2006;444:10221023

35. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI: The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences of the United States of America 2004;101:1571815723

36. Sommer F, Stahlman M, Ilkayeva O, Arnemo JM, Kindberg J, Josefsson J, Newgard CB, Frobert O, Backhed F: The Gut Microbiota Modulates Energy Metabolism in the Hibernating Brown Bear Ursus arctos. Cell reports 2016;14:16551661

23 Diabetes Page 24 of 39

37. Kurdi P, Kawanishi K, Mizutani K, Yokota A: Mechanism of growth inhibition by free bile acids in lactobacilli and bifidobacteria. J Bacteriol 2006;188:19791986

38. Thaiss CA, Levy M, Grosheva I, Zheng D, Soffer E, Blacher E, Braverman S, Tengeler AC, Barak O, Elazar M, BenZeev R, LehaviRegev D, Katz MN, Pevsner Fischer M, Gertler A, Halpern Z, Harmelin A, Aamar S, Serradas P, Grosfeld A, Shapiro H, Geiger B, Elinav E: Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science 2018;359:13761383

39. Pussinen PJ, Havulinna AS, Lehto M, Sundvall J, Salomaa V: Endotoxemia is associated with an increased risk of incident diabetes. Diabetes Care 2011;34:392397

40. Wexler HM: Bacteroides: the good, the bad, and the nittygritty. Clin Microbiol Rev 2007;20:593621

41. Prawitt J, Caron S, Staels B: Bile acid metabolism and the pathogenesis of type 2 diabetes. Curr Diab Rep 2011;11:160166

42. Chiang JY: Bile acid metabolism and signaling. Compr Physiol 2013;3:11911212

43. Li T, Chiang JY: Bile acids as metabolic regulators. Curr Opin Gastroenterol 2015;31:159165

44. Ridlon JM, Kang DJ, Hylemon PB: Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006;47:241259

45. Jones JD, Zollman PE: Black bear (Ursus americanus) bile composition: seasonal changes. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 1997;118:387390

46. FernandezSanchez L, BravoOsuna I, Lax P, ArranzRomera A, Maneu V, Esteban Perez S, Pinilla I, PueblaGonzalez MDM, HerreroVanrell R, Cuenca N: Controlled delivery of tauroursodeoxycholic acid from biodegradable microspheres slows retinal degeneration and vision loss in P23H rats. PloS one 2017;12:e0177998

47. Lawson EC, Bhatia SK, Han MK, Aung MH, Ciavatta V, Boatright JH, Pardue MT: Tauroursodeoxycholic Acid Protects Retinal Function and Structure in rd1 Mice. Adv Exp Med Biol 2016;854:431436

48. Mantopoulos D, Murakami Y, Comander J, Thanos A, Roh M, Miller JW, Vavvas DG: Tauroursodeoxycholic acid (TUDCA) protects photoreceptors from cell death after experimental retinal detachment. PloS one 2011;6:e24245

49. Murase H, Tsuruma K, Shimazawa M, Hara H: TUDCA Promotes Phagocytosis by Retinal Pigment Epithelium via MerTK Activation. Investigative ophthalmology & visual science 2015;56:25112518

50. Oveson BC, Iwase T, Hackett SF, Lee SY, Usui S, Sedlak TW, Snyder SH, Campochiaro PA, Sung JU: Constituents of bile, bilirubin and TUDCA, protect against oxidative stressinduced retinal degeneration. J Neurochem 2011;116:144153

24 Page 25 of 39 Diabetes

51. Phillips MJ, Walker TA, Choi HY, Faulkner AE, Kim MK, Sidney SS, Boyd AP, Nickerson JM, Boatright JH, Pardue MT: Tauroursodeoxycholic acid preservation of photoreceptor structure and function in the rd10 mouse through postnatal day 30. Investigative ophthalmology & visual science 2008;49:21482155

52. Xia H, Nan Y, Huang X, Gao J, Pu M: Effects of Tauroursodeoxycholic Acid and AlphaLipoicAcid on the Visual Response Properties of Cat Retinal Ganglion Cells: An In Vitro Study. Investigative ophthalmology & visual science 2015;56:66386645

53. Wang CF, Yuan JR, Qin D, Gu JF, Zhao BJ, Zhang L, Zhao D, Chen J, Hou XF, Yang N, Bu WQ, Wang J, Li C, Tian G, Dong ZB, Feng L, Jia XB: Protection of tauroursodeoxycholic acid on high glucoseinduced human retinal microvascular endothelial cells dysfunction and streptozotocininduced diabetic retinopathy rats. J Ethnopharmacol 2016;185:162170

54. Cho JG, Lee JH, Hong SH, Lee HN, Kim CM, Kim SY, Yoon KJ, Oh BJ, Kim JH, Jung SY, Asahara T, Kwon SM, Park SG: Tauroursodeoxycholic acid, a bile acid, promotes blood vessel repair by recruiting vasculogenic progenitor cells. Stem cells 2015;33:792805

55. Yoon YM, Lee JH, Yun SP, Han YS, Yun CW, Lee HJ, Noh H, Lee SJ, Han HJ, Lee SH: Tauroursodeoxycholic acid reduces ER stress by regulating of Aktdependent cellular prion protein. Scientific reports 2016;6:39838

56. Engin F, Yermalovich A, Nguyen T, Hummasti S, Fu W, Eizirik DL, Mathis D, Hotamisligil GS: Restoration of the unfolded protein response in pancreatic beta cells protects mice against type 1 diabetes. Science translational medicine 2013;5:211ra156

57. Rodrigues CM, Ma X, LinehanStieers C, Fan G, Kren BT, Steer CJ: Ursodeoxycholic acid prevents cytochrome c release in apoptosis by inhibiting mitochondrial membrane depolarization and channel formation. Cell Death Differ 1999;6:842854

58. Brestoff JR, Artis D: Commensal bacteria at the interface of host metabolism and the immune system. Nat Immunol 2013;14:676684

59. Dwyer MA, Kazmin D, Hu P, McDonnell DP, Malek G: Research resource: nuclear receptor atlas of human retinal pigment epithelial cells: potential relevance to agerelated macular degeneration. Mol Endocrinol 2011;25:360372

25 Diabetes Page 26 of 39

Fig. 1. IF feeding prolonged survival of db/db mice without improving glycemic control. (A) Depiction of the experimental protocol with the treatment groups. Mice were put on IF diet at 4 months of age and were held of IF for 7 months. ZT represents zeitgeber time, i.e. time when the lights went on. (B) Glycated hemoglobin levels in db/db-AL and db/m- IF mice were higher compared to db/m mice, however, IF did not correct glycated hemoglobin levels. Data represent means ± SEM (n=6-39); *p < 0.05. (C) Db/db-AL had reduced survival compared to db/m mice. IF improved survival rate of db/db-IF mice to that of normal controls; *p < 0.05.

126x138mm (300 x 300 DPI)

Page 27 of 39 Diabetes

163x135mm (300 x 300 DPI)

Diabetes Page 28 of 39

Fig. 3. IF resulted in distinct changes in the microbiota of db/db mice. (A-C). Beta diversity (Presence/Absence Ordination): Dimensional reduction of the Jaccard distance on presence/absence OTU table, using the PCoA ordination method. (A) db/m-AL (green circles) vs. db/db-AL(blue triangles); samples separated along Axis 2 according to sample type. (B) db/m-AL (green triangles) vs. db/m IF (feeding: red triangles; fasting: red circles); samples separated along axis 2 according to type, which indicated that samples from IF and AL treatments have different incidences of OTUs. (C) db/db-AL (blue triangles) vs. db/db-IF (feeding: purple triangles; fasting: purple circles); samples separated along axis 1 according to type. Samples from db/d-AL had a smaller degree of variation in microbiome beta diversity than db/db-IF samples. p=0.001 in all comparisons; n=5 mice/group. (D) Principal component analysis of microbiota. Relative abundance of bacteria present in at least 10% of the samples (n = 180) was used. Samples grouped by both disease (spheres: db/m; cubes: db/db) and treatment (blue: ad lib; red: IF), but less clearly by feeding/fasting within the IF groups. Note that the AL cohorts are closer to each other, while the IF cohorts were much further apart, suggesting bigger differences in the microbiomes of db/m-IF and db/db- IF. (E) Relative proportions of the 6 most abundant phyla (the aggregate relative abundance for each of the phyla not represented here is less than 0.1). F/B: ratio is Firmicutes: Bacteroidetes. a p < 0.01 vs db/m-AL; b p < 0.001 vs db/m-AL; c p < 0.05 vs. db/db-AL; d p < 0.001 vs. db/db-AL; e p < 0.05 vs. db/m IF (feeding); f p < 0.001 vs. db/m IF (feeding); g p < 0.05 vs. db/db IF (fasting). All OTUs (detected in at least 10% of the samples) from all samples per each group were used, irrespective of ZT.

194x143mm (300 x 300 DPI)

Page 29 of 39 Diabetes

Fig. 4. IF effects on microbiome composition at the genus level. (A) Genera that show statistically significant changes in db/dbAL vs db/mAL mice. (B) Genera that were statistically different in db/dbIF vs db/mIF in the feeding phase. (C) Genera that were statistically different in db/dbIF vs db/mIF in the fasting phase. Points are OTUs belonging to each respective genus. Features were considered significant if FDRcorrected pvalue ≤ 0.05, and the absolute value of Log2 Fold Change ≥ 1. (DO). Time course of relative bacterial abundances of selected taxa during the 48hr cycle of IF regimen are shown. ZT represents zeitgeber time, i.e. time when the lights went on. Diurnal oscillations in selected taxa and IF effects: (D) Order Clostridiales; (E) Family Ruminococcus; (F) Family Lachnospiraceae; (G) Genus Lactobacillus; (H) Genus Bifidobacterium; (I) Genus Clostridium; (J) Genus Oscillospira; (K) Genus Ruminoccocus; (L) Genus Turicibacter; (M) Genus Bacteroides; (N) Genus Akkermansia; (O) Genus Allobaculum. Data are sums of relative abundance of all OTUs within each genus ± SEM.

180x208mm (300 x 300 DPI) Diabetes Page 30 of 39

Page 31 of 39 Diabetes

Fig. 5. IF treatment affected gut morphology. (AD) PAS staining showing mucin positive goblet cells. Diabetes reduced goblet cell number, but IF prevented this reduction. (E) Quantification of goblet positive cells. Data represent number of goblet cells/villus. *p < 0.05, n=1330 villi group. (FI) Hematoxylin and eosin staining of colon. The length of villi was reduced significantly with diabetes but IF restored levels to normal. (J) Quantification of the villi length, in pixels. *p < 0.05, n=1726 villi/group. (KN) Hematoxylin and eosin staining of the muscularis layer. (O) Quantification of the muscularis width. 20 measurements distributed evenly were done on each section, and data represent averages per section in pixels, ± SD; n=47 sections/group. In all cases, one section per mouse was analyzed. Magnifications bars: 50 µm. (P) Peptidoglycan, a component of bacterial cell wall, was measured in plasma from the cohorts using ELISA. Data represent means ± SD (n=35). p<0.05. Two Way ANOVA.

215x279mm (300 x 300 DPI) Diabetes Page 32 of 39

Page 33 of 39 Diabetes

Fig. 6. IF enhanced bile acid metabolism with increases in the neuroprotective bile acid TUCDA. Analysis of plasma samples for bile acid metabolism (A) cholate (B) deoxycholate (C) Taurochenodeoxycholate (TCDCA) was identified and significantly increased with IF diet in the db/dbIF mice. TCDCA is converted to Tauroursodeoxycholate (TUDCA) by the actions of both 7α and 7β HSDH. (D) TUDCA was significantly increased in the plasma of db/dbIF mice. Data represent means ± SD (n=46). p<0.05. Two Way ANOVA.

130x129mm (300 x 300 DPI)

Diabetes Page 34 of 39

177x207mm (300 x 300 DPI)

Page 35 of 39 Diabetes

Fig. 8. A model for IF-induced changes in the microbiome and their potential impact on development of DR. In diabetic mice, IF resulted in significant expansion of bacteria of the Firmicutes phylum that metabolize primary bile acids to secondary bile acids, such as TUDCA. TUDCA then, enters the circulation and crosses the blood retina barrier, targets its receptor, TGR5 in the ganglion cell layer and protects against retinal neurodegeneration and inflammation.

165x185mm (300 x 300 DPI)

Diabetes Page 36 of 39 Page 37 of 39 Diabetes

A Control-IF B Diabetes-IF Feedin Fastin Feedin Fastin

94otu43063 Phylum g g g g Bacteroidetes 94otu1346 Firmicutes

94otu15334

94otu6138

94otu9254

u

n 94otu15375

u

e n 94otu31487

G

s

e

G s 94otu43360

94otu13105

94otu4147

Allobaculum

94otu17070

unclassified

−5.0 −2.5 0.0 2.5 log 2 fold change of D-IF fasting vs.D-IF feeding

Fig. S2. Microbiome analysis. (A) Genera that show statistically significant changes in db/m-IF fasting vs db/m-IF feeding mice. (B) Genera that show statistically significant changes in db/db-IF fasting vs db/db-IF feeding mice. Diabetes Page 38 of 39

Enriched OTUs class order family genus (#OTUs) C Bacteroidia Bacteroidales S24-7 unclassified (2) Coprococcus (2) db/m IF fasting db/db IF feeding Lachnospiraceae unclassified (2) Clostridiales Oscillospira (5) Ruminococcaceae Ruminococcus(2) unclassified unclassified(4) Erysipelotrichi Erysipelotrichales Erysipelotrichaceae unclassified(3) Mollicutes RF39 unclassified unclassified (1)

class order family genus (#OTUs) Bacteroidia Bacteroidales S24-7 unclassified (9) Dorea (1) Lachnospiraceae unclassified (14) Oscillospira (7) Clostridia Clostridiales db/m IF fasting db/db IF fasting Ruminococcaceae Ruminococcus (6) unclassified (11) unclassified unclassified (52) class order family genus (#OTUs) Erysipelotrichi Erysipelotrichales Erysipelotrichaceae unclassified (1) Dehalobacteriaceae Dehalobacterium (1) Mollicutes RF39 unclassified unclassified (1) Lachnospiraceae unclassified (6) unclassified (3) class order family genus (#OTUs) Clostridia Clostridiales Ruminococcaceae Oscillospira (7) Lachnospiraceae unclassified (3) Ruminococcus (1) Clostridia Clostridiales Ruminococcaceae unclassified (4) unclassified unclassified (14) unclassified(17) unclassified(17) Erysipelotrichi Erysipelotrichales Erysipelotrichaceae Coprobacillus (1) Mollicutes RF39 unclassified unclassified (1) Mollicutes RF39 unclassified unclassified (1)

Continuing Fig. S2. Microbiome analysis. (C). Venn diagram for the comparison of lists of bacterial species that were significantly increased from their perspective AL diets in order to identify commonly or uniquely increased OTUs among the four data sets of db/m-IF feeding, db/m- IF fasting, db/db-IF feeding, and db/db-IF fasting. Page 39 of 39 Diabetes D class order family genus (#OTUs) Decreased OTUs Actinomycetales Micrococcaceae unclassified (1) Actinobacteria Bifidobacteriales Bifidobacteriaceae Bifidobacterium (1) db/m IF fasting db/db IF feeding Alphaproteobacteria Caulobacterales Caulobacteraceae unclassified (1) Bacteroidia Bacteroidales S24-7 unclassified (3) unclassified (1) Clostridia Clostridiales Ruminococcaceae unclassified (2) unclassified unclassified (11) Enterobacteriales Enterobacteriaceae unclassified (1) Gammaproteobacteria Pseudomonadales Moraxellaceae Acinetobacter (1) Mollicutes RF39 unclassified unclassified (1) class order family genus (#OTUs) Actinomycetales Corynebacteriaceae Corynebacterium (1) Actinobacteria Bifidobacteriales Bifidobacteriaceae Bifidobacterium (2) Bacilli Turicibacterales Turicibacteraceae Turicibacter (1) Bacteroidaceae Bacteroides (15) Bacteroidia Bacteroidales S24-7 unclassified (35) Clostridiaceae unclassified (2) db/m IF fasting db/db IF fasting Lachnospiraceae unclassified (6) Peptostreptococcaceae unclassified (1) Clostridia Clostridiales Oscillospira (3) Ruminococcaceae Ruminococcus (3) class order family genus (#OTUs) unclassified (1) Actinobacteria Bifidobacteriales Bifidobacteriaceae Bifidobacterium (6) unclassified unclassified (25) Adlercreutzia (3) Bacteroidia Bacteroidales S24-7 unclassified (1) Coriobacteriia Coriobacteriales Coriobacteriaceae Clostridiaceae unclassified (3) unclassified (1) Erysipelotrichi Erysipelotrichales Erysipelotrichaceae Allobaculum (7) Dorea (1) Lachnospiraceae Gammaproteobacteria Enterobacteriales Enterobacteriaceae unclassified (2) unclassified (4) Mollicutes RF39 unclassified unclassified (4) Clostridia Clostridiales Oscillospira Verrucomicrobiae Verrucomicrobiales Verrucomicrobiaceae Akkermansia (3) Ruminococcaceae Ruminococcus (3) unclassified (2) class order family genus (#OTUs) Prevotellaceae Prevotella (1) unclassified unclassified (11) Bacteroidia Bacteroidales Mollicutes RF39 unclassified unclassified (1) S24-7 unclassified (3) Christensenellaceae unclassified (1) Dorea (1) Lachnospiraceae Clostridia Clostridiales unclassified (1) Ruminococcaceae unclassified (5) unclassified unclassified (3) Coriobacteriia Coriobacteriales Coriobacteriaceae unclassified (1) Verrucomicrobiae Verrucomicrobiales Verrucomicrobiaceae Akkermansia (4)

Continuing Fig. S2. Microbiome analysis. (D) Venn diagram for the comparison of lists of bacteria that were significantly decreased from their perspective AL diets in order to identify commonly or uniquely decreased OTUs . Diabetes Page 40 of 39 G.Bifidobacterium OTU 1259 G.Sutterella OTU 378 A Intermittent Fasting B Intermittent Fasting Ad-lib Feeding Fasting Ad-lib Feeding Fasting 3.00E-05 1.60E-05

2.50E-05 1.40E-05 ) ) % ( %

( 1.20E-05 e

c 2.00E-05 e c n n a a

d 1.00E-05 d u u b b a

1.50E-05 a

e 8.00E-06 e v i v t i t a l a l e 6.00E-06

r 1.00E-05 e r 4.00E-06 5.00E-06 2.00E-06

0.00E+00 0.00E+00 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 ZT time ZT time

db/m-AL db/db-AL db/m-IF db/db-IF db/m-AL db/db-AL db/m-IF db/db-IF

C D G.Coprococcus OTU 182 G.Oscillospira OTU 13 Intermittent Fasting Intermittent Fasting Ad-lib Feeding Fasting Ad-lib Feeding Fasting

5.00E-04 3.50E-02 4.50E-04 3.00E-02

) 4.00E-04 ) % (

% (

e 3.50E-04

2.50E-02 c e n c

a 3.00E-04 n d

a 2.00E-02 u d b 2.50E-04 a u

e b v

a 1.50E-02

i 2.00E-04

t e a l v e 1.50E-04 i r t 1.00E-02 a l

1.00E-04 e r 5.00E-05 5.00E-03 0.00E+00 0.00E+00 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 121620 0 4 8 121620 0 4 8 121620 0 4 8 121620 0 4 8 12 ZT time ZT time db/m-AL db/db-AL db/m-IF db/db-IF db/m-AL db/db-AL db/m-IF db/db-IF

Fig. S3. Examples of OTUs that show diurnal variations in the AL regimen. The JTK cycle was used to examine whether a circadian rhythm exists for relative abundance of bacteria in db/m and db/db for each OTU. The comparison of the pattern in relative abundance among (db/m IF, db/db-IF), (db/m-AL, db/db-AL), (db/m-IF, db/m-AL), and (db/db-IF, db/db-AL) was conducted using Kolmogorov–Smirnov test, a nonparametric test of equality of probability distribution. (A) c_Actinobacteria, o_Bifidobacteriales, f_Bifidobacteriaceae, g_Bifidobacterium, s_unclassified_otu1259. (B) c_Betaproteobacteria, o_Burkholderiales, f_Alcaligenaceae, g_Sutterella, s_97otu21533_otu378. (B) c_Clostridia, o_Clostridiales, f_ Lachnospiraceae, g_ Coprococcus, s_unclassified_otu182. (C) c_Clostridia, o_Clostridiales, f_ Ruminococcaceae, g_Oscillospira, s_97otu13166_otu13. Green lines: db/m-AL; blue lines: db/db-AL; red lines: db/m-IF; purple lines: db/db-IF. c_(class), o_(order), f_(family), g_(genus), s_(species). Page 41 of 39 Diabetes

T a ble 1S . Analysis of bile acid content in plasma interm itte nt fa sting Ad-libitum fee ding fa sting bile a cids

db/m (n=4) db/db (n= 4) db/m (n= 5 ) db/db (n= 4-5 ) db/m (n= 5) db/db (n=4 -6) cholate (CA) 0.166 ± 0.04 11.986 ± 6.3 2.840 ± 1.69 3.923 ± 1.71 4.001 ± 1.96 4.851 ± 1.53 alpha-muricholate (α-MCA) 0.192 ± 0.00 1.173 ± 0.62 1.025 ± 0.55 0.920 ± 0.37 3.374 ± 1.90 1.396 ± 0.48 unconjug ated beta-muricholate (β-MCA) 0.560 ± 0.13 2.120 ± 1.55 2.275 ± 0.96 1.656 ± 0.67 7.648 ± 3.51 4.304 ± 2.23 a ll prim a ry unconjug a ted bile a cids: 0.91 7 ± 0.1 6 15 .279 ± 7 .73 6.1 40 ± 3.17 6.49 9 ± 2.6 9 15 .023 ± 7 .35 10.551 ± 4.03 glycocholate (GCA) 0.257 ± 0.10 2.376 ± 1.16 0.438 ± 0.18 2.146 ± 0.60 0.355 ± 0.13 0.941 ± 0.38 Prim a ry bile a cids tauro-beta-muricholate (Tβ-MCA) 0.809 ± 0.20 2.243 ± 0.73 0.418 ± 0.15 5.927 ± 2.47 0.576 ± 0.15 3.405 ± 1.17 conjug a ted taurochenodeoxycholate (TCDCA) 0.697 ± 0.12 1.776 ± 0.52 0.556 ± 0.19 8.961 ± 4.54 0.482 ± 0.11 3.092 ± 1.19 taurocholate (TCA) 0.382 ± 0.16 4.868 ± 1.79 0.661 ± 0.27 4.545 ± 1.41 0.426 ± 0.14 2.076 ± 0.60 a ll prim a ry conjug a ted bile acids: 2.14 4 ± 0.5 6 11 .264 ± 3 .70 2.0 72 ± 0.75 1 9.93 6 ± 8.3 0 1 .839 ± 0 .49 9.5 15 ± 2.86 a ll prim a ry bile a cids (conjug a ted a nd unconjug a ted): 3.06 2 ± 0.5 5 26 .543 ± 1 1.1 3 8.2 12 ± 3.74 2 6.43 5 ± 7.9 1 16 .862 ± 7 .78 20.0 66 ± 6.45 12-dehydrocholate (12-DCA) 0.053 ± 0.00 0.237 ± 0.11 2.663 ± 1.50 0.686 ± 0.44 9.607 ± 4.09 0.435 ± 0.23 3-dehydrocholate (3-DCA) 0.097 ± 0.00 1.103 ± 0.59 0.892 ± 0.55 0.523 ± 0.27 1.711 ± 0.80 0.456 ± 0.18 7-ketodeoxycholate (7KDCA) 0.563 ± 0.15 0.974 ± 0.54 5.309 ± 2.89 0.794 ± 0.43 14.628 ± 6.52 2.096 ± 1.25 unconjug atedd eoxycholate (DCA) 0.209 ± 0.03 0.876 ± 0.27 0.906 ± 0.37 1.524 ± 0.22 2.374 ± 0.96 2.387 ± 0.72 ursocholate (UCA) 0.141 ± 0.02 1.228 ± 0.64 1.852 ± 0.92 0.582 ± 0.25 3.773 ± 1.46 0.862 ± 0.28 ursodeoxycholate (UDCA) 0.544 ± 0.14 0.631 ± 0.28 3.104 ± 1.26 0.928 ± 0.41 6.770 ± 3.40 2.196 ± 0.97 S econda ry bile a cid a ll se conda ry unconjug a te d bile a cids : 1.60 6 ± 0.2 7 5 .048 ± 2 .32 14.7 27 ± 7.38 5.03 8 ± 1.8 3 38 .863 ± 1 6.9 3 8.4 31 ± 3.13 taurodeoxycholate (TDCA) 0.265 ± 0.09 1.737 ± 0.65 0.415 ± 0.20 1.946 ± 0.39 0.765 ± 0.22 2.280 ± 0.59 taurohyodeoxycholic acid (THDCA) 0.638 ± 0.19 2.990 ± 1.25 0.465 ± 0.15 1.717 ± 0.46 0.687 ± 0.27 2.805 ± 1.09 conjug a ted tauroursodeoxycholate (TUDCA) 1.024 ± 0.16 0.913 ± 0.26 0.645 ± 0.10 2.668 ± 1.02 0.664 ± 0.18 2.063 ± 0.28 a ll se conda ry conjug a ted bile a cids: 1.92 7 ± 0.3 7 5 .641 ± 1 .80 1.5 25 ± 0.44 5.97 3 ± 1.6 5 2 .116 ± 0 .61 6.6 30 ± 1.78 a ll seconda ry bile a cids : 3.53 4 ± 0.4 4 10 .689 ± 4 .06 16.2 52 ± 7.82 1 1.01 1 ± 2.6 2 40 .980 ± 1 7.0 2 15.0 61 ± 4.62 a ll bile a cids (prim a ry a nd se conda ry ) 6.59 5 ± 0.8 9 43 .490 ± 1 9.5 6 24.4 64 ± 11 .54 3 7.44 6 ± 10.13 57 .842 ± 2 4.6 8 35.1 27 ± 11 .05 a ll conjug a te d bile a cids (prim ary a nd se conda ry ) 4.07 2 ± 0.8 8 16 .905 ± 5 .49 3.5 97 ± 1.11 2 5.90 9 ± 9.9 0 3 .956 ± 0 .97 16.1 45 ± 4.59 a ll unconjug a ted bile a cids(prim a ry a nd seconda ry ) 2.52 4 ± 0.4 0 20 .327 ± 1 0.0 4 20.8 67 ± 10 .54 1 1.53 7 ± 4.4 8 53 .886 ± 2 4.1 6 18.9 82 ± 7.07 ra tio of conjug a ted/ unconjug a ted 1.88 9 ± 0.5 1 1 .663 ± 0 .61 0.2 56 ± 0.05 5.02 8 ± 3.2 2 0 .211 ± 0 .13 0.9 20 ± 0.18 ra tio of D CA /CA (indica tion of 7 -α dehy drox y la tion) 1.57 9 ± 0.3 6 0 .213 ± 0 .09 0.4 99 ± 0.09 1.87 7 ± 1.3 7 1 .202 ± 0 .41 0.6 53 ± 0.22 ra tio of conjug a ted /unconjug a ted seconda ry bile a cids (indica tion of B S H ) 1.45 3 ± 0.4 3 1 .720 ± 0 .51 0.1 77 ± 0.05 1.85 5 ± 0.7 1 0 .169 ± 0 .11 0.9 18 ± 0.21