Acute response of peripheral blood cell to autologous hematopoietic stem cell transplantation in type 1 diabetic patient

Xiaofang Zhang, Lei Ye, Jiong Hu, Wei Tang, Ruixin Liu, Minglan Yang, Jie Hong, Weiqing Wang, Guang Ning, Weiqiong Gu, Xiaofang Zhang, Lei Ye, Jiong Hu, Wei Tang, Ruixin Liu, Minglan Yang, Jie Hong, Weiqing Wang, Guang Ning, Weiqiong Gu

Abstract

Objective: Autologous nonmyeloablative hematopoietic stem cell transplantation (AHST) was the first therapeutic approach that can improve β cell function in type 1 diabetic (T1D) patients. This study was designed to investigate the potential mechanisms involved.

Design and methods: We applied AHST to nine T1D patients diagnosed within six months and analyzed the acute responses in peripheral blood for lymphocyte subpopulation as well as for genomic expression profiling at the six-month follow-up.

Results: We found six patients obtained insulin free (IF group) and three remained insulin dependent (ID group); C-peptide production was significantly higher in IF group compared to ID group. The acute responses in lymphocytes at six-month follow-up include declined CD3(+)CD4(+), CD3(+)CD8(+) T cell population and recovered B cell, NK cell population in both groups but with no significant differences between the two groups; most immune-related genes and pathways were up-regulated in peripheral blood mononuclear cell (PBMC) of both groups while none of transcription factors for immune regulatory component were significantly changed; the IF group demonstrated more AHST-modified genetic events than the ID group and distinct pattern of top pathways, co-expression network as well as 'hub' genes (eg, TCF7 and GZMA) were associated with each group.

Conclusions: AHST could improve the islet function in newly diagnosed T1D patients and elimination of the islet specific autoreactive T cells might be one of the mechanisms involved; T1D patients responded differently to AHST possibly due to the distinct transcriptional events occurring in PBMC.

Trial registration: ClinicalTrials.gov NCT00807651.

Conflict of interest statement

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

Figures

Figure 1. Time course of fasting C-peptide…
Figure 1. Time course of fasting C-peptide (A), Cmax (B), AUCC (C), GADA (D) and HbA1c (E) in IF group and ID group respectively.
Black circles, insulin free. Black squares, insulin dependent. X axis represents the time course relative to HSCT. A p<0.05, pre-treatment vs all follow-up time in IF group; IF vs ID group at 1 month and 12 months post-treatment. B p<0.05, pre-treatment vs all follow-up times in IF group; IF vs ID group at one month and six months post-treatment. C p<0.05, pre-treatment vs all follow-up times in IF group; IF vs ID group at 12 months post-treatment. D p = 0.036, IF vs ID group at six months post-treatment.
Figure 2. Workflow of the genomic expression…
Figure 2. Workflow of the genomic expression profiling of the PMBC in patients with type 1 diabetes.
Differentially expressed genes (pre-treatment vs post-treatment) meet the criterion p

Figure 3. Sector plots of differential network…

Figure 3. Sector plots of differential network analysis in IF group (A) and ID group…

Figure 3. Sector plots of differential network analysis in IF group (A) and ID group (B).
The difference in connectivity (DiffK) is plotted on the X axis, and p values are plotted on the Y axis. Horizontal lines indicate a difference in connectivity of −0.2 and 0.2, whereas vertical lines depict a p value 0.05. Number indicates sector 1–6 and plots represent genes. The genes ploted in the sector 1 and 3 are considered as difference in connectivity.
Figure 3. Sector plots of differential network…
Figure 3. Sector plots of differential network analysis in IF group (A) and ID group (B).
The difference in connectivity (DiffK) is plotted on the X axis, and p values are plotted on the Y axis. Horizontal lines indicate a difference in connectivity of −0.2 and 0.2, whereas vertical lines depict a p value 0.05. Number indicates sector 1–6 and plots represent genes. The genes ploted in the sector 1 and 3 are considered as difference in connectivity.

References

    1. American Diabetes Association. Diagnosis and classification of diabetes. Diabetes Care. 2004;27(suppl I):S5–S10.
    1. Lehuen Agnès, Diana Julien, Zaccone Paola, Cooke Anne. Immune cell crosstalk in type 1 diabetes. Nature Reviews Immunology. 2010;10:501–5133.
    1. Notkins AL, Lernmark A. Autoimmune type 1 diabetes: resolved and unresolved issues. J Clin Invest. 2001;108:1247–1252.
    1. The Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long term complications in insulin-dependent diabetes mellitus. N Engl J Med. 1993;329:977–98.
    1. Chatenoud L. CD3-specific antibody-induced active tolerance: From bench to bedside. Nat. Rev. Immunol. 2003;3:123–132.
    1. Voltarelli JC, Couri CE, Stracieri AB, Oliveira MC, Moraes DA, et al. Autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA. 2007;297:1568–1576.
    1. Couri CE, Oliveira MC, Stracieri AB, Moraes DA, Pieroni F, et al. C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA. 2009;301:1573–9.
    1. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2007;30(Suppl 1):S42–S474.
    1. Fuller TF, Ghazalpour A, Aten JE, Drake TA, Lusis AJ, et al. Weighted gene coexpression network analysis strategies applied to mouse weigh. Mamm Genome. 2007;18(6–7):463–72.
    1. Wright GW, Simon RM. A random variance model for detection of differential gene expression in small microarray experiments. Bioinformatics. 2003;19(18):2448–2455.
    1. Marmont AM, van Lint MT, Gualandi F, Bacigalupo A. Autologous marrow mstem cell transplantation for systemic lupus erythematosus of long duration. Lupus. 1997;6:545–548.
    1. Beard ME, Willis JA, Scott RS, Nesbit JW. Is type 1 diabetes transmissible by bone marrow allograft? Diabetes Care. 2002;25(4):799–800.
    1. Atkinson MA, Leiter EH. The NOD mouse model of type 1 diabetes: as good as it gets? Nature Med. 1999;6:601–604.
    1. Wen Y, Ouyang J, Yang R, Chen J, Liu Y, et al. Reversal of new onset type 1 diabetes in mice by syngeneic bone marrow transplantation. Biochem. Biophys Res Commun. 2008;374:282–287.
    1. Voltarelli JC, Couri CE, Stracieri AB, Oliveira MC, Moraes DA, et al. Autologous hematopoietic stem cell transplantation for type 1 diabetes. Ann N Y Acad Sci. 2008;2008 1150:220–9.
    1. Palmer JP, Fleming GA, Greenbaum CJ, Herold KC, Jansa LD, et al. C-peptide is the appropriate outcome measure for type 1 diabetes clinical trials to preserve beta-cell function. Diabetes. 2004;53:250–264.
    1. Muraro PA, Douek DC, Packer A, Chung K, Guenaga FJ, et al. Thymic output generates a new and diverse TCR repertoire after autologous stem cell transplantation in multiple sclerosis patients. J Exp Med. 2005;201:805–816.
    1. Alexander T, Thiel A, Rosen O, Massenkeil G, Sattler A, et al. Depletion of autoreactive immunologic memory followed by autologous hematopoietic stem cell transplantation in patients with refractory SLE induces long-term remission through de novo generation of a juvenile and tolerant immune system. Blood. 2009;113:214–223.
    1. Reynier F, Pachot A, Paye M, Xu Q, Turrel-Davin F, et al. Specific gene expression signature associated with development of autoimmune type-I diabetes using whole-blood microarray analysis. Genes Immun. 2010;11:269–78.
    1. Tsukamoto H, Nagafuji K, Horiuchi T, Mitoma H, Niiro H, et al. Analysis of immune reconstitution after autologous CD34+ stem/progenitor cell transplantation for systemic sclerosis: predominant reconstitution of Th1 CD4+ T cells. Rheumatology. 2011;50(5):944–52.
    1. Suk K, Kim S, Kim YH, Kim KA, Chang I, et al. IFN-gamma/TNF-alpha synergism as the final effector in autoimmune diabetes: a key role for STAT1/IFN regulatory factor-1 pathway in pancreatic beta cell death. J Immunol. 2001;166:4481–4489.
    1. Han JD, Bertin N, Hao T, Goldberg DS, Berriz GF, et al. Evidence for dynamically organized modularity in the yeast protein-protein interaction network. Nature. 2004;430:88–93.
    1. Jeong H, Mason SP, Barabasi AL, Oltvai ZN. Lethality and centrality in protein networks. Nature. 2001;411:41–42.
    1. Erlich HA, Valdes AM, Julier C, Mirel D, Noble JA, et al. Evidence for association of the TCF7 locus with type I diabetes. Genes Immun Suppl. 2009;1:S54–59.
    1. Zhou X, Yu S, Zhao DM, Harty JT, Badovinac VP, Xue HH. Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity. 2010;33:229–240.
    1. de Kleer I, Vastert B, Klein M, Teklenburg G, Arkesteijn G, et al. Autologous stem cell transplantation for autoimmunity induces immunologic self-tolerance by reprogramming autoreactive T cells and restoring the CD4+CD25+ immune regulatory network. Blood. 2006;107:1696–1702.
    1. Metkar SS, Menaa C, Pardo J, Wang B, Wallich R, et al. Human and mouse granzyme A induce a proinflammatory cytokine response. Immunity. 2008;29:720–733.
    1. Martinvalet D, Dykxhoorn DM, Ferrini R, Lieberman J. Granzyme A cleaves a mitochondrial complex I protein to initiate caspase-independent cell death. Cell. 2008;133:681–692.
    1. Held W, MacDonald HR, Weissman IL, Hess MW, Mueller C. Immunology Genes encoding tumor necrosis factor a and granzyme A are expressed during development of autoimmune diabetes. Proc Natl Acad Sci. 1990;87:2239–2243.

Source: PubMed

3
Předplatit