Human cord blood stem cell-modulated regulatory T lymphocytes reverse the autoimmune-caused type 1 diabetes in nonobese diabetic (NOD) mice

Yong Zhao, Brian Lin, Robert Darflinger, Yongkang Zhang, Mark J Holterman, Randal A Skidgel, Yong Zhao, Brian Lin, Robert Darflinger, Yongkang Zhang, Mark J Holterman, Randal A Skidgel

Abstract

Background: The deficit of pancreatic islet beta cells caused by autoimmune destruction is a crucial issue in type 1 diabetes (T1D). It is essential to fundamentally control the autoimmunity for treatment of T1D. Regulatory T cells (Tregs) play a pivotal role in maintaining self-tolerance through their inhibitory impact on autoreactive effector T cells. An abnormality of Tregs is associated with initiation of progression of T1D.

Methodology/principal findings: Here, we report that treatment of established autoimmune-caused diabetes in NOD mice with purified autologous CD4(+)CD62L(+) Tregs co-cultured with human cord blood stem cells (CB-SC) can eliminate hyperglycemia, promote islet beta-cell regeneration to increase beta-cell mass and insulin production, and reconstitute islet architecture. Correspondingly, treatment with CB-SC-modulated CD4(+)CD62L(+) Tregs (mCD4CD62L Tregs) resulted in a marked reduction of insulitis, restored Th1/Th2 cytokine balance in blood, and induced apoptosis of infiltrated leukocytes in pancreatic islets.

Conclusions/significance: These data demonstrate that treatment with mCD4CD62L Tregs can reverse overt diabetes, providing a novel strategy for the treatment of type 1 diabetes as well as other autoimmune diseases.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Flow analysis of CD4 +…
Figure 1. Flow analysis of CD4+CD62L+ Tregs following in vitro co-culture with human cord blood stem cells CB-SC.
Mouse spleen lymphocytes were isolated from female NOD mice (aged 6–8 weeks) and then co-cultured with CB-SC for 2–4 days at different ratios (A) or a ratio 1∶10 of CB-SC∶lymphocytes (B–D). Subsequently, the total lymphocytes were harvested for following cell count or flow analyses. Lymphocytes cultured in absence of CB-SC served as control. (A) CB-SC have no significant effect on mouse lymphocyte proliferation during co-culture at different ratios of CB-SC∶lymphocytes. Data represent mean±s.d. of four experiments. (B) Percentage of CD4+CD25+ Treg, CD4+Foxp3+ Treg, and CD4+CD62L+ Treg after in vitro co-culture with CB-SC. (C) Flow analysis of CD25 and Foxp3 expressions in CD4+CD62L+ Tregs after in vitro co-culture with CB-SC. (D) Flow analysis on CD4+CD62L+ Tregs after intra-cellular cytokine staining. Isotype-matched IgG served as control. Data in B–C are representative of three to five experiments.
Figure 2. CB-SC-modulated CD4 + CD62L +…
Figure 2. CB-SC-modulated CD4+CD62L+ Tregs (mCD4CD62L Tregs) reverse hyperglycemia in overt diabetic NOD mice.
Mouse spleen lymphocytes isolated from female NOD mice (aged 6–8 weeks) were co-cultured with CB-SC for cell sorting as described in methods. These purified CD4+CD62L+ Tregs (mCD4CD62L Tregs) were used for treatment of spontaneously-developed autoimmune-caused diabetes in NOD mice. The purified CD4+CD62L+ Tregs from lymphocytes not co-cultured with CB-SC (control CD4CD62L Tregs) served as control for mCD4CD62L Tregs. Injection of vehicle PBS served as an additional control (n = 5). (A) The mCD4CD62L Tregs correct hyperglycemia in diabetic NOD mice. Overt diabetic NOD mice were treated with mCD4CD62L Tregs (total 5 million cells/mouse, i.p., red line; representative data are from 6 diabetic mice sensitive to mCD4CD62L Treg treatment with euglycemia, n = 8 mice). Purified control CD4CD62L Tregs served as control (total 5 million cells/mouse, i.p., blue line, n = 5 mice). PBS served as an additional control (black line, n = 5 mice). (B) Intraperitoneal glucose tolerance testing (IPGTT) 3 weeks following the 1st treatment with mCD4CD62L Tregs. Seven-week old NOD mice served as normal control. (C) Determination of blood insulin levels by ELISA. (D) Effect of treatment on mouse body weight. (E–H) Pancreatic histology analyses: Pancreata from mCD4CD62L Treg-treated diabetic mice and control mice were collected for immunohistochemistry after observation for 45 days. Representative sections are shown in each panel (G, H). (E) Morphometric analysis of pancreatic β-cell mass. Pancreatic β-cell mass was determined by point-counting morphometry on insulin-positive islet β cells followed by immunostaining with guinea pig anti-insulin Ab (Dako) and counter-staining with hematoxylin. (F) Quantification of Ki67-positive cells in pancreatic islets after double immunostaining with Ki67 and insulin Abs. Isotype-matched rabbit IgG served as control for rabbit anti-Ki67 mAb. (G) Confocal microscopy shows double-immunostaining for insulin (red) and a cell proliferation nuclear marker Ki67 (green) (scale bar, 50 µm), with a high magnification (two bottom rows, scale bar 10 µm). Control CD4CD62L Treg-treated diabetic mice (top panels) showed the Ki67-positive cells distributed in the infiltrated inflammatory cells (blue, with high density), not in β-cell area (dashed pink circle), with almost complete disappearance of β cells (red). (H) Double-immunostaining for β-cell marker insulin (red) and α-cell marker glucagon (green), followed by nuclear counter-staining with DAPI (blue). Seven-week old NOD mice served as non-diabetic control to show normal islet architecture. Scale bar, 50 µm.
Figure 3. Treatment with mCD4CD62L Tregs reverses…
Figure 3. Treatment with mCD4CD62L Tregs reverses insulitis and immune dysfunction in diabetic NOD mice.
Overt diabetic NOD mice treated with mCD4CD62L Tregs were sacrificed for pancreatic histological analysis and evaluation of blood cytokine levels after observation for 45 days (n = 8). The control CD4CD62L Treg-treated diabetic mice (n = 5) and PBS-treated diabetic mice (n = 5) served as controls. (A and B) Treatment with mCD4CD62L Tregs corrects insulitis in overt type 1 diabetic NOD mice. Representative data are from 6 diabetic mice (6/8 mice) sensitive to mCD4CD62L Treg treatment with euglycemia. (A) Scoring of insulitis. Pancreatic islets were scored for % mononuclear cell infiltration after immunostaining for insulin and counter-staining with hematoxylin as described in methods. (B) Representative images for different type of insulitis. Data were collected from mCD4CD62L Treg-treated diabetic NOD mice. Scale bar, 50 µm. (C) Determination of plasma IFN-γ level by ELISA. Non-diabetic NOD mice at age of 6 weeks served as normal control. (D) Measurement of plasma IL-4 level by ELISA. (E) Determination of plasma IL-10 level measured by ELISA. (F) Determination of plasma TGF-β1 level measured by ELISA. Data are shown as mean±s.d. of mouse plasma cytokine levels from three experiments.
Figure 4. Treatment with mCD4CD62L Tregs enhance…
Figure 4. Treatment with mCD4CD62L Tregs enhance expression of TGF-β1 in pancreatic islets.
Overt diabetic NOD mice treated with mCD4CD62L Tregs were sacrificed for pancreatic immunohistochemistry studies after observation for 45 days (n = 8). Representative data are from 6 diabetic mice (6/8 mice) sensitive to mCD4CD62L Treg treatment with euglycemia. The control CD4CD62L Treg-treated diabetic mice served as control (n = 5). (A) TGF-β1 staining surrounds a pancreatic islet of mCD4CD62L Treg-treated diabetic mice, as determined by double-immunostaining for TGF-β1 (yellow) and insulin (red). TGF-β1 positive cells (bright yellow) and released TGF-β1 in matrix (faint yellow) were distributed in the islet β cell area (red) and surrounded islet β cells (top panels, scale bar 50 µm). High magnification is shown in bottom panels, scale bar 10 µm. Isotype-matched mouse IgG1 served as a negative control for TGF-β1 immunostaining in a serial pancreatic section. Representative images were obtained from five experiments. (B) TUNEL assay. The proteinase K-pretreated pancreatic slides were initially immunostained with TGF-β1 (yellow) and insulin (red) Abs, followed by TUNEL assay (green), and nuclear counterstaining with DAPI (blue). Scale bar 20 µm. Representative images were obtained from four experiments. (C) Percentage of apoptotic cells in subtypes of infiltrated leukocytes in pancreatic islets. Cryosections (8 µm thickness) of frozen pancreata from mCD4CD62L Treg-treated diabetic mice (n = 4) and control mice (n = 4) were initially detected with In Situ Cell Death Detection Kit (Roche), followed by immunostaining with different monoclonal Abs and imaging with a Zeiss LSM 510 META confocal microscope. Cryosections incubated with label solution without the TUNEL reaction mixture and/or isotype-matched IgG served as negative controls. Data represent mean±s.d. of five experiments.

References

    1. Trucco M. Regeneration of the pancreatic beta cell. J Clin Invest. 2005;115:5–12.
    1. Zhao Y, Glesne D, Huberman E. A human peripheral blood monocyte-derived subset acts as pluripotent stem cells. Proc Natl Acad Sci U S A. 2003;100:2426–2431.
    1. Zhao Y, Wang H, Mazzone T. Identification of stem cells from human umbilical cord blood with embryonic and hematopoietic characteristics. Exp Cell Res. 2006;312:2454–2464.
    1. Ricordi C, Hering BJ, Shapiro AM. Beta-cell transplantation for diabetes therapy. Lancet. 2008;372:27–28.
    1. Ablamunits V, Sherry NA, Kushner JA, Herold KC. Autoimmunity and beta cell regeneration in mouse and human type 1 diabetes: the peace is not enough. Ann N Y Acad Sci. 2007;1103:19–32.
    1. Bonner-Weir S, Weir GC. New sources of pancreatic beta-cells. Nat Biotechnol. 2005;23:857–861.
    1. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 2008
    1. Bluestone JA, Tang Q, Sedwick CE. T Regulatory Cells in Autoimmune Diabetes: Past Challenges, Future Prospects. J Clin Immunol 2008
    1. Bresson D, Togher L, Rodrigo E, Chen Y, Bluestone JA, et al. Anti-CD3 and nasal proinsulin combination therapy enhances remission from recent-onset autoimmune diabetes by inducing Tregs. J Clin Invest. 2006;116:1371–1381.
    1. Chatenoud L, Bach JF. Resetting the functional capacity of regulatory T cells: a novel immunotherapeutic strategy to promote immune tolerance. Expert Opin Biol Ther. 2005;5(Suppl 1):S73–S81.
    1. Chatenoud L, Bach JF. Regulatory T cells in the control of autoimmune diabetes: the case of the NOD mouse. Int Rev Immunol. 2005;24:247–267.
    1. Roncarolo MG, Battaglia M. Regulatory T-cell immunotherapy for tolerance to self antigens and alloantigens in humans. Nat Rev Immunol. 2007;7:585–598.
    1. Tarbell KV, Petit L, Zuo X, Toy P, Luo X, et al. Dendritic cell-expanded, islet-specific CD4+CD25+CD62L+ regulatory T cells restore normoglycemia in diabetic NOD mice. J Exp Med. 2007;204:191–201.
    1. You S, Slehoffer G, Barriot S, Bach JF, Chatenoud L. Unique role of CD4+CD62L+ regulatory T cells in the control of autoimmune diabetes in T cell receptor transgenic mice. Proc Natl Acad Sci U S A. 2004;101(Suppl 2):14580–14585.
    1. Li Q, Xu B, Michie SA, Rubins KH, Schreriber RD, et al. Interferon-alpha initiates type 1 diabetes in nonobese diabetic mice. Proc Natl Acad Sci U S A. 2008;105:12439–12444.
    1. Schneider A, Rieck M, Sanda S, Pihoker C, Greenbaum C, et al. The effector T cells of diabetic subjects are resistant to regulation via CD4+FOXP3+ regulatory T cells. J Immunol. 2008;181:7350–7355.
    1. Tang Q, Adams JY, Penaranda C, Melli K, Piaggio E, et al. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity. 2008;28:687–697.
    1. Zehn D, Bevan MJ. T cells with low avidity for a tissue-restricted antigen routinely evade central and peripheral tolerance and cause autoimmunity. Immunity. 2006;25:261–270.
    1. Allen JS, Pang K, Skowera A, Ellis R, Rackham C, et al. Plasmacytoid dendritic cells are proportionally expanded at diagnosis of Type 1 diabetes and enhance islet autoantigen presentation to T cells through immune complex capture. Diabetes 2008
    1. Huang Y, Fugier-Vivier IJ, Miller T, Elliott MJ, Xu H, et al. Plasmacytoid precursor dendritic cells from NOD mice exhibit impaired function: are they a component of diabetes pathogenesis? Diabetes. 2008;57:2360–2370.
    1. Jin Y, Chen X, Podolsky R, Hopkins D, Makala LH, et al. APC dysfunction is correlated with defective suppression of T cell proliferation in human type 1 diabetes. Clin Immunol 2008
    1. Marleau AM, Summers KL, Singh B. Differential contributions of APC subsets to T cell activation in nonobese diabetic mice. J Immunol. 2008;180:5235–5249.
    1. Summers KL, Marleau AM, Mahon JL, McManus R, Hramiak I, et al. Reduced IFN-alpha secretion by blood dendritic cells in human diabetes. Clin Immunol. 2006;121:81–89.
    1. Kukreja A, Cost G, Marker J, Zhang C, Sun Z, et al. Multiple immuno-regulatory defects in type-1 diabetes. J Clin Invest. 2002;109:131–140.
    1. Pop SM, Wong CP, Culton DA, Clarke SH, Tisch R. Single cell analysis shows decreasing FoxP3 and TGFbeta1 coexpressing CD4+CD25+ regulatory T cells during autoimmune diabetes. J Exp Med. 2005;201:1333–1346.
    1. You S, Belghith M, Cobbold S, Alyanakian MA, Gouarin C, et al. Autoimmune diabetes onset results from qualitative rather than quantitative age-dependent changes in pathogenic T-cells. Diabetes. 2005;54:1415–1422.
    1. Bayry J, Lacroix-Desmazes S, Dasgupta S, Kazatchkine MD, Kaveri SV. Efficacy of regulatory T-cell immunotherapy: are inflammatory cytokines key determinants? Nat Rev Immunol. 2008;8:1p.
    1. Brusko T, Wasserfall C, McGrail K, Schatz R, Viener HL, et al. No alterations in the frequency of FOXP3+ regulatory T-cells in type 1 diabetes. Diabetes. 2007;56:604–612.
    1. Brusko TM, Wasserfall CH, Clare-Salzler MJ, Schatz DA, Atkinson MA. Functional defects and the influence of age on the frequency of CD4+CD25+ T-cells in type 1 diabetes. Diabetes. 2005;54:1407–1414.
    1. Gombert JM, Herbelin A, Tancrede-Bohin E, Dy M, Chatenoud L, et al. Early defect of immunoregulatory T cells in autoimmune diabetes. C R Acad Sci III. 1996;319:125–129.
    1. Lindley S, Dayan CM, Bishop A, Roep BO, Peakman M, et al. Defective suppressor function in CD4(+)CD25(+) T-cells from patients with type 1 diabetes. Diabetes. 2005;54:92–99.
    1. Tritt M, Sgouroudis E, d'Hennezel E, Albanese A, Piccirillo CA. Functional waning of naturally occurring CD4+ regulatory T-cells contributes to the onset of autoimmune diabetes. Diabetes. 2008;57:113–123.
    1. Zhao Y, Huang Z, Qi M, Lazzarini P, Mazzone T. Immune regulation of T lymphocyte by a newly characterized human umbilical cord blood stem cell. Immunol Lett. 2007;108:78–87.
    1. Abdi R, Fiorina P, Adra CN, Atkinson M, Sayegh MH. Immunomodulation by mesenchymal stem cells: a potential therapeutic strategy for type 1 diabetes. Diabetes. 2008;57:1759–1767.
    1. Meier JJ, Lin JC, Butler AE, Galasso R, Martinez DS, et al. Direct evidence of attempted beta cell regeneration in an 89-year-old patient with recent-onset type 1 diabetes. Diabetologia. 2006;49:1838–1844.
    1. Li MO, Flavell RA. TGF-beta: a master of all T cell trades. Cell. 2008;134:392–404.
    1. Roncarolo MG, Battaglia M, Gregori S. The role of interleukin 10 in the control of autoimmunity. J Autoimmun. 2003;20:269–272.
    1. Wan YY, Flavell RA. The roles for cytokines in the generation and maintenance of regulatory T cells. Immunol Rev. 2006;212:114–130.
    1. Perruche S, Zhang P, Liu Y, Saas P, Bluestone JA, et al. CD3-specific antibody-induced immune tolerance involves transforming growth factor-beta from phagocytes digesting apoptotic T cells. Nat Med. 2008;14:528–535.
    1. Staeva-Vieira T, Peakman M, von HM. Translational mini-review series on type 1 diabetes: Immune-based therapeutic approaches for type 1 diabetes. Clin Exp Immunol. 2007;148:17–31.
    1. Nauta AJ, Fibbe WE. Immunomodulatory properties of mesenchymal stromal cells. Blood. 2007;110:3499–3506.
    1. Bour-Jordan H, Salomon BL, Thompson HL, Santos R, Abbas AK, et al. Constitutive expression of B7-1 on B cells uncovers autoimmunity toward the B cell compartment in the nonobese diabetic mouse. J Immunol. 2007;179:1004–1012.
    1. Goudy KS, Burkhardt BR, Wasserfall C, Song S, Campbell-Thompson ML, et al. Systemic overexpression of IL-10 induces CD4+CD25+ cell populations in vivo and ameliorates type 1 diabetes in nonobese diabetic mice in a dose-dependent fashion. J Immunol. 2003;171:2270–2278.

Source: PubMed

3
Abonner