Cutting Edge: Fasting-Induced Hypoleptinemia Expands Functional Regulatory T Cells in Systemic Erythematosus This information is current as of October 3, 2021. Yaoyang Liu, Yiyun Yu, Giuseppe Matarese and Antonio La Cava J Immunol published online 30 January 2012 http://www.jimmunol.org/content/early/2012/01/30/jimmun

ol.1102835 Downloaded from

Supplementary http://www.jimmunol.org/content/suppl/2012/01/30/jimmunol.110283 Material 5.DC1 http://www.jimmunol.org/

Why The JI? Submit online.

• 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 by guest on October 3, 2021

*average

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 © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published January 30, 2012, doi:10.4049/jimmunol.1102835

Cutting Edge: Fasting-Induced Hypoleptinemia Expands Functional Regulatory T Cells in Systemic Lupus Erythematosus Yaoyang Liu,*,† Yiyun Yu,* Giuseppe Matarese,‡ and Antonio La Cava* Fasting is beneficial in the prevention and amelioration During fasting, the levels of the circulating adipokine leptin of the clinical manifestations of autoimmune diseases in- are dramatically reduced (5). Leptin has all of the character- cluding systemic lupus erythematosus. The mechanisms istics of a proinflammatory (6) and links nutritional responsible for these effects are not well understood. status with neuroendocrine functions and immune responses. During fasting, there is a dramatic reduction of the levels Leptin promotes Th1 cell differentiation and the development of circulating leptin, an adipokine with proinflamma- of autoimmune responses in several animal disease models

+ + + (7), and it can act as a negative factor for the expansion of Downloaded from tory effects. Leptin also inhibits CD4 CD25 Foxp3 + regulatory T cells, which are known to contribute sig- CD4 regulatory T cells (TReg), a subset of T cells with an nificantly to the mechanisms of peripheral immune important role in the maintenance of peripheral immune tolerance. In this study, we show that fasting-induced tolerance to self-Ags (8). hypoleptinemia in (NZB 3 NZW)F lupus-prone mice In an attempt to connect these observations, we investigated 1 the effects of fasting in NZB/W mice and found that the induced an expansion of functional regulatory T cells reduction in circulating levels of leptin caused by starvation http://www.jimmunol.org/ that was reversed by leptin replacement. The specificity directly promoted the expansion of functional TReg. These of the findings was indicated by the lack of these effects findings can help to explain some of the beneficial effects of in leptin-deficient ob/ob mice and leptin receptor- fasting in SLE. deficient db/db mice. These observations help to ex- plain the beneficial effects of fasting in autoimmunity Materials and Methods and could be exploited for leptin-based immune inter- Mice vention in systemic lupus erythematosus. The Journal C57BL6/J (B6) wild-type (WT) mice, leptin-deficient B6ob/ob (ob/ob), leptin of Immunology, 2012, 188: 000–000. receptor-deficient B6db/db (db/db), and NZB/W mice were purchased from The Jackson Laboratory (Bar Harbor, ME) and maintained at the University by guest on October 3, 2021 of California Los Angeles. Mice were treated in accordance with institutional guidelines under approved protocols. All experiments were conducted in age- asting and/or caloric restriction are known to alleviate matched female mice that were divided into three groups. One control group the manifestations of autoimmune diseases through had ad libitum access to food and received i.p. injections of 0.2 ml PBS mechanisms that remain mostly elusive (1). For ex- at 9 AM and 6 PM daily for 2 d. The other two groups of mice were deprived F of food for 48 h and received i.p. injections of either 0.2 ml PBS or re- 3 ample, caloric restriction prolongs life in (NZB NZF)F1 combinant leptin (R&D Systems, Minneapolis, MN) dissolved in PBS at a (NZB/W) mice (2) that spontaneously develop systemic lupus dose of 1 mg/g body weight twice daily (also at 9 AM and 6 PM). All mice had erythematosus (SLE), an characterized by continuous access to water. the breakdown of tolerance to multiple nuclear Ags and an Cell isolation and staining uncontrolled activation of self-reactive lymphocytes that cause After blood drawing, erythrocytes were removed using red cell lysing buffer inflammation and tissue damage. Although it has been shown (Sigma-Aldrich, St. Louis, MO), and PBMC were used for flow cytometry. For that in NZB/W mice the reduction of caloric intake associates splenocytes, single-cell suspensions were prepared by passing cells through a cell with a decrease in lymphoproliferation, Ab production, and strainer before red cell lysis and resuspension in HL-1 medium (BioWhittaker, + + + 2 secretion of proinflammatory mediators (3), it remains un- Walkersville, MD). CD4 CD25 and CD4 CD25 T cells were isolated via magnetic bead separation with Miltenyi Biotec kits using an AutoMACS clear what drives the observed beneficial clinical, immuno- System (Miltenyi Biotec, Auburn, CA) and found .95% pure by flow logical, and biochemical effects (4). cytometry analysis.

*Division of Rheumatology, David Geffen School of Medicine, University of California Address correspondence and reprint requests to Prof. Antonio La Cava, Department Los Angeles, Los Angeles, CA 90095; †Department of Laboratory Diagnosis, Changhai of Medicine, University of California Los Angeles, 1000 Veteran Avenue 32-59, Los Hospital, Second Military Medical University, Shanghai 200433, China; and Angeles, CA 90095. E-mail address: [email protected] ‡Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, The online version of this article contains supplemental material. Consiglio Nazionale delle Ricerche, Federico II University of Napoli, Naples 80138, Italy Abbreviations used in this article: SLE, systemic lupus erythematosus; TEff, effector CD4+ T cells; T , regulatory CD4+ T cells; WT, wild-type. Received for publication October 3, 2011. Accepted for publication December 22, 2011. Reg This work was supported by National Institutes of Health Grants AR53239 and Copyright Ó 2012 by The American Association of Immunologists, Inc. 0022-1767/12/$16.00 AI95921 (to A.L.C.).

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1102835 2 CUTTING EDGE: FASTING EXPANDS TReg THROUGH HYPOLEPTINEMIA IN SLE

Flow cytometry mice starved for 48 h had hypoleptinemia when compared Phenotypic analyses were performed with combinations of fluorochrome- with ad libitum-fed animals, whereas no changes in leptin conjugated Ab using standard techniques. After Fc blocking, fluorochrome- levels were observed in starved ob/ob mice [which cannot conjugated anti-mouse Ab to CD4 and CD25 (eBioscience, San Diego, produce functional leptin because of a mutation in the leptin CA) or isotype control Ab were used for staining prior to acquisition on an FACSCalibur flow cytometer (BD Biosciences, San Jose, CA) and subsequent gene (9)] and db/db mice [which carry a mutation in the analysis using FlowJo software (Tree Star, Ashland, OR). For intracellular leptin receptor (10)] (Table I). The administration of leptin staining, cells were first stained for the expression of cell-surface markers and to starved mice restored the circulating levels of this adipokine then fixed, permeabilized, and stained using the Foxp3 staining kit (eBio- to normal concentrations in leptin-sufficient mice (Table I), science) according to the manufacturer’s instructions. and spleen cellularity returned to values similar to those seen Suppression assays in ad libitum-fed controls in starved mice that received leptin + 2 (Supplemental Table I). After cell isolation, CD4 CD25 effector T cells (TEff) were labeled with CFSE by incubation in 0.5 mM CFSE at 37˚C for 10 min. Samples were then Because we reported that leptin can constrain the expansion placed in HL-1/5% FCS and incubated in ice for 5 min, then washed two of TReg in vivo (8), we investigated whether the reduction in times. TReg/TEff (ratio 1:1 or 1:4) mixtures or control samples consisting of circulating levels of leptin induced by fasting could affect the TEff alone were plated in 96-well round-bottom plates (Corning Life Sciences, Lowell, MA). Dynabeads mouse anti-CD3/CD28 beads were added to a final TReg numbers. It was found that the frequency of TReg in ratio of 0.5 beads/cell. Samples were incubated in a 37˚C incubator for 72 h. starved WT and NZB/W lupus mice was significantly in- Proliferation of CFSE-labeled TEff was evaluated by flow cytometry and creased as compared with ad libitum-fed mice (Fig. 1). Of expressed as FlowJo software-calculated division index (the average number of interest, leptin administration to ob/ob mice, which had no cell divisions). Supernatants were collected for ELISA detection of IFN-g in Downloaded from the cocultures. Percent suppression was calculated using proliferation meas- change of leptin levels after starvation (Table I), associated 2 urements and the following formula: percent suppression = (1 TEff in TReg: with a reduction in the number of T (Fig. 1C, 1D), suggest- 3 Reg TEff/TEff alone) 100. ing that leptin per se has inhibitory effects on the expansion ELISA of TReg in vivo. Also, db/db mice (which are hyperleptinemic but unresponsive to leptin) showed no change in TReg frequ- Concentration of leptin and IFN-g were determined by commercial ELISA kits (R&D Systems) according to the manufacturer’s instructions. ency after starvation, and leptin replacement did not change http://www.jimmunol.org/ this finding (Fig. 1E, 1F). The TReg that had expanded Statistical analyses after starvation had similar CD25 expression (which can be Analyses were performed using Prism 4 software (GraphPad, San Diego, CA). a marker of immune cell activation) in the different groups Nonparametric testing among three groups was performed by Kruskal-Wallis of animals (Supplemental Table I), and the frequency of ANOVA. Comparisons among three groups were performed by Dunn’s + 2 CD4 CD25 cells (TEff) in PBMC was reduced by fasting multiple comparison test. The p values ,0.05 were considered statistically significant. and restored by leptin replacement in WT, ob/ob, and NZB/ W mice (Supplemental Table I). Taken together, these results indicate that fasting associates Results and Discussion by guest on October 3, 2021 The effects of fasting were compared among WT mice, leptin- with an increase in TReg frequency when the leptin pathway is deficient ob/ob mice, leptin receptor-deficient db/db mice, and intact (in WT and NZB/W mice), and the TReg frequency lupus prone NZB/W mice. Control animals were fed ad remains unaltered when fasting cannot alter leptinemia due libitum or starved 48 h (treated or not with leptin). As ex- to an impairment of the leptin/leptin receptor axis (such as in pected, starvation led to a reduction in body weight in all ob/ob and in db/db mice). These events depend on leptin groups of mice when compared with ad libitum-fed animals because in WT and NZB/W mice the TReg frequency re- (Table I). Starvation also associated with a significant reduc- turned to numbers comparable to those found in ad libitum- tion in the number of splenocytes and TEff in starved WT, ob/ fed mice following leptin replacement (Fig. 1). ob, and NZB/W mice (Supplemental Table I). To also address whether the observed changes in TReg fre- It is well established that fasting associates with the reduction quency associated or not with a normal function of the sup- of circulating leptin levels (5). Accordingly, WT and NZB/W pressor cells, in vitro suppression assays were performed. The

Table I. Body weight and leptin measurements in mice fed ad libitum or fasted

Mice Ad Libitum Fast + PBS Fast + Leptin WT Initial body weight (g) 20.59 6 0.72 20.97 6 0.79 20.59 6 0.74 Body weight after 48 h (g) 20.77 6 0.68 17.20 6 0.70* 17.8 6 0.78* Plasma leptin (ng/ml) 7.22 6 0.67 4.06 6 0.64** 6.78 6 0.55*** ob/ob Initial body weight (g) 58.36 6 0.61 58.42 6 1.39 58.66 6 1.38 Body weight after 48 h (g) 58.46 6 0.60 54.82 6 1.47 54.68 6 1.36 Plasma leptin (ng/ml) 0.19 6 0.01 0.23 6 0.01 6.35 6 0.73**** db/db Initial body weight (g) 59.5 6 0.29 57.1 6 1.88 60.77 6 1.36 Body weight after 48 h (g) 59.6 6 0.25 52.9 6 2.89 56.13 6 1.72 Plasma leptin (ng/ml) 9.43 6 1.13 8.01 6 0.29 11.46 6 0.65*** NZB/W Initial body weight (g) 23.82 6 0.45 23.63 6 0.35 22.92 6 0.38 Body weight after 48 h (g) 23.87 6 0.42 19.92 6 0.58***** 19.30 6 0.43***** Plasma leptin (ng/ml) 9.79 6 0.40 5.75 6 0.48****** 8.70 6 0.58*** Mice were fed ad libitum or fasted 48 h. Values represent mean 6 SEM. *p , 0.05 versus initial body weight; **p , 0.05 versus ad libitum; ***p , 0.05 versus fast + PBS; ****p , 0.01 versus ad libitum and fast + PBS; *****p , 0.001 versus initial body weight; ******p , 0.01 versus ad libitum. The Journal of Immunology 3 Downloaded from http://www.jimmunol.org/ by guest on October 3, 2021

+ + + FIGURE 1. Fasting expands TReg in NZB/W lupus mice. CD4 CD25 Foxp3 T cells from WT, ob/ob, db/db, and NZB/W mice fed ad libitum or starved 48 h, without or with leptin treatment. Representative (A, C, E, G) and cumulative flow cytometry data (B, D, F, H)(n = 4–7/group) on gated CD4+ T cells from PBMC in one of three independent experiments. *p , 0.05, **p , 0.01.

results showed that the changes in TReg frequency under the The link between nutritional status and immune regulation different conditions (ad libitum, fast, and fast + leptin) did suggested by recent reports that showed that the mammalian not associate with differences in the suppressive capacity of target of rapamycin pathway could influence the activity of TReg on TEff (Fig. 2, Supplemental Table II), indicating that TReg through leptin (16) identifies in the current study a new TReg expanded by fasting were functionally comparable possibility to modulate TReg activity in SLE via leptin-based among groups. intervention [also considering that leptin is elevated in SLE Altogether, these results widen earlier observations that patients (17)]. In particular, the frequency of TReg could be showed that caloric restriction ameliorated SLE manifestations tuned for therapeutic purposes in SLE not only through (2, 11, 12). The observation that functional TReg could ex- fasting but also, more specifically, using neutralizing Abs to pand during fasting in lupus mice is relevant to the disease leptin to promote the expansion in vivo of functional TReg in pathogenesis because the frequency of TReg is lower in NZB/ the disease. W mice as compared with WT mice (13, 14), and this aspect has been linked to an inability of the dysfunctional immune Disclosures system in SLE to control disease course and progression (15). The authors have no financial conflicts of interest. 4 CUTTING EDGE: FASTING EXPANDS TReg THROUGH HYPOLEPTINEMIA IN SLE

FIGURE 2. Downloaded from In vitro suppression of TEff proliferation by TReg (1:1 ratio) in ad libitum-fed versus 48 h-starved mice without or with leptin replacement. (A)WTmice; (B) ob/ob;(C) db/db;and(D) NZB/W mice. The figure shows proliferation in flow cytometry of anti-CD3/CD28 Ab-stimulated, splenocyte-derived, CFSE-labeled TEff cul- tured alone (gray line) or with (black line) TReg. http://www.jimmunol.org/ by guest on October 3, 2021

10. Chen, H., O. Charlat, L. A. Tartaglia, E. A. Woolf, X. Weng, S. J. Ellis, References N. D. Lakey, J. Culpepper, K. J. Moore, R. E. Breitbart, et al. 1996. Evidence that 1. Fernandes, G. 2008. Progress in nutritional immunology. Immunol. Res. 40: 244–261. the diabetes gene encodes the leptin receptor: identification of a mutation in the 2. Fernandes, G., P. Friend, E. J. Yunis, and R. A. Good. 1978. Influence of dietary leptin receptor gene in db/db mice. Cell 84: 491–495. restriction on immunologic function and renal disease in (NZB x NZW) F1 mice. 11. Johnson, B. C., A. Gajjar, C. Kubo, and R. A. Good. 1986. Calories versus protein in Proc. Natl. Acad. Sci. USA 75: 1500–1504. onset of renal disease in NZB x NZW mice. Proc. Natl. Acad. Sci. USA 83: 5659–5662. 3. Leiba, A., H. Amital, M. E. Gershwin, and Y. Shoenfeld. 2001. Diet and lupus. 12. Sun, D., A. Krishnan, J. Su, R. Lawrence, K. Zaman, and G. Fernandes. 2004. Lupus 10: 246–248. Regulation of immune function by calorie restriction and cyclophosphamide 4. Hsieh, C. C., and B. F. Lin. 2011. Dietary factors regulate in murine treatment in lupus-prone NZB/NZW F1 mice. Cell. Immunol. 228: 54–65. models of systemic lupus erythematosus. Autoimmun. Rev. 11: 22–27. 13. La Cava, A., F. M. Ebling, and B. H. Hahn. 2004. Ig-reactive CD4+CD25+ T cells 5. Maffei, M., J. Halaas, E. Ravussin, R. E. Pratley, G. H. Lee, Y. Zhang, H. Fei, from tolerized (New Zealand Black x New Zealand White)F1 mice suppress in vitro S. Kim, R. Lallone, S. Ranganathan, et al. 1995. Leptin levels in human and rodent: production of antibodies to DNA. J. Immunol. 173: 3542–3548. measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. 14. Scalapino, K. J., Q. Tang, J. A. Bluestone, M. L. Bonyhadi, and D. I. Daikh. 2006. Nat. Med. 1: 1155–1161. Suppression of disease in New Zealand Black/New Zealand White lupus-prone mice by 6. Lord, G. M., G. Matarese, J. K. Howard, R. J. Baker, S. R. Bloom, and adoptive transfer of ex vivo expanded regulatory T cells. J. Immunol. 177: 1451–1459. R. I. Lechler. 1998. Leptin modulates the T-cell immune response and reverses 15. La Cava, A. 2008. T-regulatory cells in systemic lupus erythematosus. Lupus 17: starvation-induced immunosuppression. Nature 394: 897–901. 421–425. 7. La Cava, A., and G. Matarese. 2004. The weight of leptin in immunity. Nat. Rev. 16. Procaccini, C., V. De Rosa, M. Galgani, L. Abanni, G. Calı`, A. Porcellini, Immunol. 4: 371–379. F. Carbone, S. Fontana, T. L. Horvath, A. La Cava, and G. Matarese. 2010. An 8. De Rosa, V., C. Procaccini, G. Calı`, G. Pirozzi, S. Fontana, S. Zappacosta, A. La oscillatory switch in mTOR kinase activity sets responsiveness. Cava, and G. Matarese. 2007. A key role of leptin in the control of regulatory T cell Immunity 33: 929–941. proliferation. Immunity 26: 241–255. 17. Garcia-Gonzalez, A., L. Gonzalez-Lopez, I. C. Valera-Gonzalez, E. G. Cardona- 9. Zhang, Y., R. Proenca, M. Maffei, M. Barone, L. Leopold, and J. M. Friedman. Mun˜oz, M. Salazar-Paramo, M. Gonza´lez-Ortiz, E. Martı´nez-Abundis, and 1994. Positional cloning of the mouse obese gene and its human homologue. Nature J. I. Gamez-Nava. 2002. Serum leptin levels in women with systemic lupus 372: 425–432. erythematosus. Rheumatol. Int. 22: 138–141.