Soluble factors secreted by T cells promote β-cell proliferation

Ercument Dirice, Sevim Kahraman, Wenyu Jiang, Abdelfattah El Ouaamari, Dario F De Jesus, Adrian K K Teo, Jiang Hu, Dan Kawamori, Jason L Gaglia, Diane Mathis, Rohit N Kulkarni, Ercument Dirice, Sevim Kahraman, Wenyu Jiang, Abdelfattah El Ouaamari, Dario F De Jesus, Adrian K K Teo, Jiang Hu, Dan Kawamori, Jason L Gaglia, Diane Mathis, Rohit N Kulkarni

Abstract

Type 1 diabetes is characterized by infiltration of pancreatic islets with immune cells, leading to insulin deficiency. Although infiltrating immune cells are traditionally considered to negatively impact β-cells by promoting their death, their contribution to proliferation is not fully understood. Here we report that islets exhibiting insulitis also manifested proliferation of β-cells that positively correlated with the extent of lymphocyte infiltration. Adoptive transfer of diabetogenic CD4(+) and CD8(+) T cells, but not B cells, selectively promoted β-cell proliferation in vivo independent from the effects of blood glucose or circulating insulin or by modulating apoptosis. Complementary to our in vivo approach, coculture of diabetogenic CD4(+) and CD8(+) T cells with NOD.RAG1(-/-) islets in an in vitro transwell system led to a dose-dependent secretion of candidate cytokines/chemokines (interleukin-2 [IL-2], IL-6, IL-10, MIP-1α, and RANTES) that together enhanced β-cell proliferation. These data suggest that soluble factors secreted from T cells are potential therapeutic candidates to enhance β-cell proliferation in efforts to prevent and/or delay the onset of type 1 diabetes.

Figures

Figure 1
Figure 1
Adoptive transfer of diabetes stimulates β-cell proliferation in NOD.RAG1−/− recipients. A: Experimental strategy showing total splenocyte (DM or pre-DM) or depleted splenocyte (diabetic mice) transfer (1 × 107 cells) into NOD.RAG1−/− mice. BrdU (100 mg/kg/BW) was injected 3 weeks post‐transfer, and 6 h later, the pancreases were harvested for immunohistochemical analyses. B: Paraffin-embedded sections of pancreas from mice receiving DM or pre-DM splenocytes, costained with proliferation markers BrdU, pHH3, or Ki67 with insulin and DAPI. Scale bar, 50 µm. Arrows indicate proliferating β-cells (BrdU+/insulin+). Insets show magnified view of representative proliferating β-cells. Quantification of data shown in B for BrdU (C), pHH3 (D), and Ki67 (E) (n = 4–16 mice in each group). *P < 0.05; **P < 0.01; ***P < 0.001 (Student t test). F: TUNEL staining of paraffin-embedded sections of pancreatic tissues obtained from recipient mice receiving DM or pre-DM splenocytes for apoptosis detection. Scale bar, 100 µm. Arrows indicate TUNEL+/β-cell+ cells undergoing apoptosis. Inset shows a magnified representative image of TUNEL+ β-cell. Lower image represents positive control of TUNEL staining in rat tumor tissue. G: Quantification of data in F. n = 4–6 mice in each group. Data are expressed as means ± SEM. FACS, fluorescence-activated cell sorting; Spl, splenocyte.
Figure 2
Figure 2
Confirmation of proliferating β-cells. Two-dimensional (A) and three-dimensional (B) confocal microscopy view of pancreatic section derived from total diabetic NOD mouse splenocyte–injected animal. Dotted line in A represents the border between islet and immune cells. Magnified area highlighted from B shows three-dimensional imaging of insulin (red) (C), Ki67 (green) (D), and DAPI (blue) (E). Scale bar, 100 µm. Arrows indicate proliferating β-cells. F: Orthographic image of the same pancreatic section in A shows the horizontal and vertical view of a proliferating β-cell in a circle (ZEN-2009). G: Mouse spleen section stained as positive control for F4/80 (green), common macrophage marker, and Ki67 (red). H: Total diabetic splenocyte–injected NOD.RAG1−/− pancreatic section stained for F4/80 (green), insulin (blue), and Ki67 (red). Scale bar, 100 µm. Arrow indicates proliferating β-cells. Magnified view of proliferating (I) and nonproliferating (J) macrophages from H. K: Mouse spleen section stained as positive control for CD3 (red), T-cell marker, and Ki67 (green). L: Total diabetic splenocyte–injected NOD.RAG1−/− pancreatic section stained for CD3 (red), insulin (blue), and Ki67 (green). Scale bar, 100 µm. Magnified view of proliferating T cell (M) and β-cell (N) from L. O: Sorting of β- and non–β-cells from dispersed islets from mice after adoptive transfer of pre-DM or DM splenocytes by flow cytometry based on size (FS Lin) and autofluorescence (FL1). P: Sorted β-cells stained for CD45 and BrdU. Dot plot showing gated-out CD45+ cells (Q) and BrdU+ β-cells from pre-DM (R) or DM (S) splenocyte-transferred mice. T: Quantification of data in R and S. n = 3 each group. *Proliferating macrophages or T cells. **P < 0.01. Experiment was performed in triplicate. Data are expressed as means ± SEM. FSC-A, forward scatter detector A; SSC-A, side scatter detector A.
Figure 3
Figure 3
T-cell subsets play a major role in β-cell proliferation. A: Paraffin-embedded sections of pancreatic tissues derived from recipient mice receiving total diabetic or B-cell–depleted diabetic NOD mouse splenocytes, costained for the proliferation marker BrdU (green) with insulin (red) and DAPI (blue). Scale bar, 50 µm. Arrows indicate proliferating β-cells (BrdU+/insulin+). Insets show a magnified representative image of a proliferating β-cell. B: Quantification of proliferating β-cells in pancreatic sections obtained from mice receiving total (DM or pre-DM) or B‐cell–, CD4+-, CD8+-, and CD4+/CD8+–double-depleted diabetic NOD splenocytes. n = 4–6 mice each group. *P < 0.05; ***P < 0.001 (Student t test). Data are expressed as means ± SEM. Spl, splenocyte. (A high-quality color representation of this figure is available in the online issue.)
Figure 4
Figure 4
Pancreatic islet infiltration positively correlates with β-cell proliferation. A: Immunofluorescence (IF) and hematoxylin and eosin (H&E) staining of consecutive pancreatic sections harvested from NOD.RAG1−/− mice 3 weeks after receiving total (DM or pre-DM) or B-cell–, CD4+-, CD8+-, and CD4+/CD8+–double-depleted diabetic NOD splenocytes. Scale bar, 400 µm. Pancreatic islets are outlined with dotted lines for ease of comprehension. B: Pancreatic islets showing insulitis expressed as a percentage in the treated groups in A. C: Linear regression of islet infiltration and BrdU+ β-cells in pancreas sections harvested from NOD.RAG1−/− mice transferred with total diabetic splenocytes. Each square represents a mouse (n = 9) scored for insulitis in at least 20 islets (n = 9). r = 0.713; P = 0.004. D: Pancreatic sections harvested from STZ-induced diabetic NOD mice at day 3 and day 7, costained for the proliferation marker BrdU (green), with insulin (red) and DAPI (blue). Scale bar, 50 µm. Arrows indicate proliferating β-cells (BrdU+/insulin+). Insets show a magnified view of a representative proliferating β-cell. E: Quantification of β-cell proliferation (bars) and insulitis scores (red dots) in the pancreatic islets in STZ-injected NOD mice at 1–7 days post-STZ administration (n = 3 mice for each time point). Data are expressed as means ± SEM. dep, depleted.
Figure 5
Figure 5
β-cell proliferation is stimulated by infiltrating lymphocytes via soluble factors. A: Experimental strategy showing total splenocytes (DM or pre-DM) or depleted splenocytes (diabetic mice), cocultured with 5–6-week-old NOD.RAG1−/− mouse islets (150 islets/condition) for 48 h in 5 mmol/L glucose in the presence or absence of transwell inserts. B: Linear regression of insulitis score and BrdU+ β-cells in single pancreatic islets harvested from NOD.RAG1−/− mice transferred with total diabetic splenocytes. Each square represents a single islet out of 120 analyzed islets. r = 0.80; P < 0.001. C: Representing pancreatic sections derived from total splenocyte– or STZ-injected mice used for determining the ratio between infiltrating immune cells vs. insulin+ β-cells. Islets are indicated by dotted lines in the right panel. The area of infiltration is shown around the islet in the left panels. β-cell proliferation in agar-embedded NOD.RAG1−/− islets cocultured with total splenocytes from DM or pre-DM at a ratio of 1:1, 1:2, 1:5, or 1:10 or without splenocytes in the presence (D) or absence (E) of transwell inserts (n = 3–4). *P < 0.05; **P < 0.01 (Student t test). F: Quantification of proliferating β-cells in pancreatic islets cocultured with total (DM or pre-DM), negatively, or positively selected DM splenocytes at 1:1 or 1:10 ratio with transwell conditions (n = 3–6 for each condition). *P < 0.05; **P < 0.01; ***P < 0.001 (Student t test). Data are expressed as means ± SEM. ND, not detected; O/N, overnight; Spl, splenocyte.
Figure 6
Figure 6
Lymphocyte-secreted soluble factors that drive β-cell proliferation. Luminex assay results from culture medium obtained 48 h after coculturing NOD.RAG1−/− islets with NOD splenocytes (DM or pre-DM) at a ratio of 1:1, 1:2, 1:5, or 1:10 or only splenocytes at 10× in the presence or absence of transwell for IL-2 (A), IL-6 (B), IL-10 (C), MIP-1α (D), and RANTES (E) (n = 3–4 for each condition). *, §P < 0.05; **, §§P < 0.01; ***, §§§P < 0.001 (Student t test). *, diabetic vs. diabetic; §, diabetic vs. prediabetic. Data are expressed as means ± SEM.
Figure 7
Figure 7
Effect of soluble factors on β-cell proliferation is reversed by inhibitory/neutralizing antibody treatment in vitro. A: Detection of the cytokine/chemokine receptor subunit mRNAs by real-time PCR from sorted β-cells and splenocytes harvested from C57BL/6 mice. Tata-box-binding protein (TBP) was used as reference. BF: Agar-embedded pancreatic islets from C57BL/BJ mouse treated in the absence (control) or presence of low-dose recombinant proteins with or without inhibitory/neutralizing molecules (as described in research design and methods) for 48 h (150 islets/condition, three to four replicates). Representative sections are shown. Islets were costained for the proliferation marker Ki67 (green) with insulin (red) and DAPI (blue). Arrows indicate proliferating β-cells (Ki67+/insulin+). Scale bar, 200 µm. Insets show a magnified image of a representative proliferating β-cell. G: Quantification of data in BF (n = 3–4 in each group). *, #P < 0.05; **, ##P < 0.01; ***, ###P < 0.001 (Student t test). *, untreated vs. cytokine/chemokine or inhibitory/neutralizing antibody treated; #, cytokine/chemokine treated vs. inhibitory/neutralizing antibody treated. Data are expressed as means ± SEM. CCR1, C-C chemokine receptor type 1; GP130, glycoprotein 130; IL-2R, IL-2 receptor.
Figure 8
Figure 8
Lymphocyte-secreted soluble factors enhance β-cell proliferation in vitro. One hundred fifty agar-embedded islets harvested from C57BL/6 mice cocultured without (untreated control) (A) or with total DM splenocytes in the absence (treated control) (B) or presence of inhibitory/neutralizing molecule Ro 26-4550 (C), anti–IL-6 (D), anti–IL-10 (E), maraviroc 3.3 nmol/L (F), or maraviroc 5.2 nmol/L (G). Islets were costained for the proliferation marker Ki67 (green), insulin (red), and DAPI (blue). Arrows indicate proliferating β-cells (Ki67+/insulin+). Scale bar, 200 µm. Insets show magnified image of a representative proliferating β-cell. H: Quantification of proliferating β-cells in AG (n = 3–4 in each group). *P < 0.05; **, ##P < 0.01; ***, ###P < 0.001 (Student t test). *, untreated vs. DM splenocyte treated; #, DM splenocyte treated vs. inhibitory/neutralizing antibody treated. Data are expressed as means ± SEM. Ins, insulin; Spl, splenocyte.

References

    1. Mathis D, Vence L, Benoist C. beta-Cell death during progression to diabetes. Nature 2001;414:792–798
    1. Redondo MJ, Fain PR, Eisenbarth GS. Genetics of type 1A diabetes. Recent Prog Horm Res 2001;56:69–89
    1. Turley S, Poirot L, Hattori M, Benoist C, Mathis D. Physiological beta cell death triggers priming of self-reactive T cells by dendritic cells in a type-1 diabetes model. J Exp Med 2003;198:1527–1537
    1. Sreenan S, Pick AJ, Levisetti M, Baldwin AC, Pugh W, Polonsky KS. Increased beta-cell proliferation and reduced mass before diabetes onset in the nonobese diabetic mouse. Diabetes 1999;48:989–996
    1. von Herrath MG, Wolfe T, Möhrle U, Coon B, Hughes A. Protection from type 1 diabetes in the face of high levels of activated autoaggressive lymphocytes in a viral transgenic mouse model crossed to the SV129 strain. Diabetes 2001;50:2700–2708
    1. Sherry NA, Kushner JA, Glandt M, Kitamura T, Brillantes AM, Herold KC. Effects of autoimmunity and immune therapy on beta-cell turnover in type 1 diabetes. Diabetes 2006;55:3238–3245
    1. Nir T, Melton DA, Dor Y. Recovery from diabetes in mice by beta cell regeneration. J Clin Invest 2007;117:2553–2561
    1. Keenan HA, Sun JK, Levine J, et al. Residual insulin production and pancreatic β-cell turnover after 50 years of diabetes: Joslin Medalist Study. Diabetes 2010;59:2846–2853
    1. Meier JJ, Bhushan A, Butler AE, Rizza RA, Butler PC. Sustained beta cell apoptosis in patients with long-standing type 1 diabetes: indirect evidence for islet regeneration? Diabetologia 2005;48:2221–2228
    1. Lee YC, Nielsen JH. Regulation of beta cell replication. Mol Cell Endocrinol 2009;297:18–27
    1. Meier JJ, Butler AE, Saisho Y, et al. Beta-cell replication is the primary mechanism subserving the postnatal expansion of beta-cell mass in humans. Diabetes 2008;57:1584–1594
    1. Finegood DT, Scaglia L, Bonner-Weir S. Dynamics of beta-cell mass in the growing rat pancreas. Estimation with a simple mathematical model. Diabetes 1995;44:249–256
    1. Karnik SK, Chen H, McLean GW, et al. Menin controls growth of pancreatic beta-cells in pregnant mice and promotes gestational diabetes mellitus. Science 2007;318:806–809
    1. Michael MD, Kulkarni RN, Postic C, et al. Loss of insulin signaling in hepatocytes leads to severe insulin resistance and progressive hepatic dysfunction. Mol Cell 2000;6:87–97
    1. Peshavaria M, Larmie BL, Lausier J, et al. Regulation of pancreatic beta-cell regeneration in the normoglycemic 60% partial-pancreatectomy mouse. Diabetes 2006;55:3289–3298
    1. Dirice E, Kulkarni RN. Pathways underlying β-cell regeneration in type 1, type 2 and gestational diabetes. In Islet Cell Growth Factors. Kulkarni RN, Ed. Texas, Landies Bioscience, 2011, p. 1–15
    1. Maedler K, Schumann DM, Sauter N, et al. Low concentration of interleukin-1beta induces FLICE-inhibitory protein-mediated beta-cell proliferation in human pancreatic islets. Diabetes 2006;55:2713–2722
    1. Ellingsgaard H, Ehses JA, Hammar EB, et al. Interleukin-6 regulates pancreatic alpha-cell mass expansion. Proc Natl Acad Sci USA 2008;105:13163–13168
    1. Cameron MJ, Strathdee CA, Holmes KD, Arreaza GA, Dekaban GA, Delovitch TL. Biolistic-mediated interleukin 4 gene transfer prevents the onset of type 1 diabetes. Hum Gene Ther 2000;11:1647–1656
    1. Pennline KJ, Roque-Gaffney E, Monahan M. Recombinant human IL-10 prevents the onset of diabetes in the nonobese diabetic mouse. Clin Immunol Immunopathol 1994;71:169–175
    1. Flodström-Tullberg M, Yadav D, Hägerkvist R, et al. Target cell expression of suppressor of cytokine signaling-1 prevents diabetes in the NOD mouse. Diabetes 2003;52:2696–2700
    1. Kulkarni RN, Winnay JN, Daniels M, et al. Altered function of insulin receptor substrate-1-deficient mouse islets and cultured beta-cell lines. J Clin Invest 1999;104:R69–R75
    1. Haxhinasto S, Mathis D, Benoist C. The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells. J Exp Med 2008;205:565–574
    1. King AJ, Fernandes JR, Hollister-Lock J, Nienaber CE, Bonner-Weir S, Weir GC. Normal relationship of beta- and non-beta-cells not needed for successful islet transplantation. Diabetes 2007;56:2312–2318
    1. Gonzalez A, Andre-Schmutz I, Carnaud C, Mathis D, Benoist C. Damage control, rather than unresponsiveness, effected by protective DX5+ T cells in autoimmune diabetes. Nat Immunol 2001;2:1117–1125
    1. Fuchtenbusch M, Larger E, Thebault K, Boitard C: Transfer of diabetes from prediabetic NOD mice to NOD-SCID/SCID mice: association with pancreatic insulin content. Horm Metab Res 2005;37:63–67
    1. Lehuen A, Diana J, Zaccone P, Cooke A. Immune cell crosstalk in type 1 diabetes. Nat Rev Immunol 2010;10:501–513
    1. Adorini L, Gregori S, Harrison LC. Understanding autoimmune diabetes: insights from mouse models. Trends Mol Med 2002;8:31–38
    1. Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature 2010;464:1293–1300
    1. van Belle TL, Coppieters KT, von Herrath MG. Type 1 diabetes: etiology, immunology, and therapeutic strategies. Physiol Rev 2011;91:79–118
    1. Crawley JB, Rawlinson L, Lali FV, Page TH, Saklatvala J, Foxwell BM. T cell proliferation in response to interleukins 2 and 7 requires p38MAP kinase activation. J Biol Chem 1997;272:15023–15027
    1. Hanifi-Moghaddam P, Kappler S, Seissler J, et al. Altered chemokine levels in individuals at risk of type 1 diabetes mellitus. Diabet Med 2006;23:156–163
    1. Eizirik DL, Colli ML, Ortis F. The role of inflammation in insulitis and beta-cell loss in type 1 diabetes. Nat Rev Endocrinol 2009;5:219–226
    1. Phillips JM, Parish NM, Raine T, et al. Type 1 diabetes development requires both CD4+ and CD8+ T cells and can be reversed by non-depleting antibodies targeting both T cell populations. Rev Diabet Stud 2009;6:97–103
    1. Scaglia L, Cahill CJ, Finegood DT, Bonner-Weir S. Apoptosis participates in the remodeling of the endocrine pancreas in the neonatal rat. Endocrinology 1997;138:1736–1741
    1. Hu CY, Rodriguez-Pinto D, Du W, et al. Treatment with CD20-specific antibody prevents and reverses autoimmune diabetes in mice. J Clin Invest 2007;117:3857–3867
    1. Jun HS, Yoon CS, Zbytnuik L, van Rooijen N, Yoon JW. The role of macrophages in T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Exp Med 1999;189:347–358
    1. Dahlén E, Dawe K, Ohlsson L, Hedlund G. Dendritic cells and macrophages are the first and major producers of TNF-alpha in pancreatic islets in the nonobese diabetic mouse. J Immunol 1998;160:3585–3593
    1. Khajah M, Millen B, Cara DC, Waterhouse C, McCafferty DM. Granulocyte-macrophage colony-stimulating factor (GM-CSF): a chemoattractive agent for murine leukocytes in vivo. J Leukoc Biol 2011;89:945–953
    1. Codarri L, Gyülvészi G, Tosevski V, et al. RORγt drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol 2011;12:560–567
    1. Huffnagle GB, Lipscomb MF, Lovchik JA, Hoag KA, Street NE. The role of CD4+ and CD8+ T cells in the protective inflammatory response to a pulmonary cryptococcal infection. J Leukoc Biol 1994;55:35–42
    1. Rolfe BE, Campbell JH, Smith NJ, Cheong MW, Campbell GR. T lymphocytes affect smooth muscle cell phenotype and proliferation. Arterioscler Thromb Vasc Biol 1995;15:1204–1210
    1. Feldon SE, Park DJ, O’Loughlin CW, et al. Autologous T-lymphocytes stimulate proliferation of orbital fibroblasts derived from patients with Graves’ ophthalmopathy. Invest Ophthalmol Vis Sci 2005;46:3913–3921
    1. Liou HC, Smith KA. The roles of c-rel and interleukin-2 in tolerance: a molecular explanation of self-nonself discrimination. Immunol Cell Biol 2011;89:27–32
    1. Diehl S, Rincón M. The two faces of IL-6 on Th1/Th2 differentiation. Mol Immunol 2002;39:531–536
    1. Maghazachi AA, Al-Aoukaty A, Schall TJ. CC chemokines induce the generation of killer cells from CD56+ cells. Eur J Immunol 1996;26:315–319
    1. Maedler K, Sergeev P, Ris F, et al. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest 2002;110:851–860
    1. Darville MI, Eizirik DL. Notch signaling: a mediator of beta-cell de-differentiation in diabetes? Biochem Biophys Res Commun 2006;339:1063–1068
    1. Russ HA, Bar Y, Ravassard P, Efrat S. In vitro proliferation of cells derived from adult human beta-cells revealed by cell-lineage tracing. Diabetes 2008;57:1575–1583
    1. Kay JE, Kromwel L, Doe SE, Denyer M. Inhibition of T and B lymphocyte proliferation by rapamycin. Immunology 1991;72:544–549

Source: PubMed

Подписаться