1
|
Mathew JM, Sanders JM, Cirocco R, Miller J, Leventhal JR. Differentiation of regulatory myeloid and T-cells from adult human hematopoietic stem cells after allogeneic stimulation. Front Immunol 2024; 15:1366972. [PMID: 38455047 PMCID: PMC10918006 DOI: 10.3389/fimmu.2024.1366972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction Donor hematopoietic stem cell (DHSC) infusions are increasingly being studied in transplant patients for tolerance induction. Methods To analyze the fate of infused DHSCs in patients, we developed an in vitro culture system utilizing CD34+DHSCs stimulated with irradiated allogeneic cells in cytokine supplemented medium long-term. Results Flow cytometric analyses revealed loss of the CD34 marker and an increase in CD33+ myeloid and CD3+ T-cell proportion by 10.4% and 72.7%, respectively, after 21 days in culture. T-cells primarily expressed TcR-αβ and were of both CD4+ and CD8+ subsets. Approximately 80% of CD3+ T cells lacked expression of the co-stimulatory receptor CD28. The CD4+ compartment was predominated by CD4+CD25+CD127-FOXP3+ Tregs (>50% CD4+CD127- compartment) with <1% of all leukocytes exhibiting a CD4+CD127+ phenotype. Molecular analyses for T-cell receptor excision circles showed recent and increased numbers of TcR rearrangements in generated T cells over time suggesting de novo differentiation from DHSCs. CD33+ myeloid cells mostly expressed HLA-DR, but lacked expression of co-stimulatory receptors CD80 and CD83. When studied as modulators in primary mixed lymphocyte reactions where the cells used to stimulate the DHSC were used as responders, the DHSC-lines and their purified CD8+, CD4+, CD33+ and linage negative subsets inhibited the responses in a dose-dependent and non-specific fashion. The CD8+ cell-mediated inhibition was due to direct lysis of responder cells. Discussion Extrapolation of these results into the clinical situation would suggest that DHSC infusions into transplant recipients may generate multiple subsets of donor "chimeric" cells and promote recipient Treg development that could regulate the anti-donor immune response in the periphery. These studies have also indicated that T cell maturation can occur in vitro in response to allogeneic stimulation without the pre-requisite of a thymic-like environment or NOTCH signaling stimulatory cell line.
Collapse
Affiliation(s)
- James M. Mathew
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jes M. Sanders
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Robert Cirocco
- HLA Laboratory, LeHigh Valley Health Network, Allentown, PA, United States
| | - Joshua Miller
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph R. Leventhal
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
2
|
Leventhal J, Mathew J, Salomon D, Kurian S, Friedewald J, Gallon L, Konieczna I, Tambur A, charette J, Levitsky J, Jie C, Kanwar YS, Abecassis MM, Miller J. Nonchimeric HLA-Identical Renal Transplant Tolerance: Regulatory Immunophenotypic/Genomic Biomarkers. Am J Transplant 2016; 16:221-34. [PMID: 26227106 PMCID: PMC4718825 DOI: 10.1111/ajt.13416] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/14/2015] [Accepted: 06/02/2015] [Indexed: 01/25/2023]
Abstract
We previously described early results of a nonchimeric operational tolerance protocol in human leukocyte antigen (HLA)-identical living donor renal transplants and now update these results. Recipients given alemtuzumab, tacrolimus/MPA with early sirolimus conversion were multiply infused with donor hematopoietic CD34(+) stem cells. Immunosuppression was withdrawn by 24 months. Twelve months later, operational tolerance was confirmed by rejection-free transplant biopsies. Five of the first eight enrollees were initially tolerant 1 year off immunosuppression. Biopsies of three others after total withdrawal showed Banff 1A acute cellular rejection without renal dysfunction. With longer follow-up including 5-year posttransplant biopsies, four of the five tolerant recipients remain without rejection while one developed Banff 1A without renal dysfunction. We now add seven new subjects (two operationally tolerant), and demonstrate time-dependent increases of circulating CD4(+) CD25(+++) CD127(-) FOXP3(+) Tregs versus losses of Tregs in nontolerant subjects (p < 0.001). Gene expression signatures, developed using global RNA expression profiling of sequential whole blood and protocol biopsy samples, were highly associative with operational tolerance as early as 1 year posttransplant. The blood signature was validated by an external Immune Tolerance Network data set. Our approach to nonchimeric operational HLA-identical tolerance reveals association with Treg immunophenotypes and serial gene expression profiles.
Collapse
Affiliation(s)
- J.R. Leventhal
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| | - J.M. Mathew
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A,Department of Microbiology-Immunology; Northwestern University, Chicago, Illinois, U.S.A
| | - D.R. Salomon
- Department of Molecular and Experimental Medicine; The Scripps Research Institute, La Jolla, California, U.S.A
| | - S.M. Kurian
- Department of Molecular and Experimental Medicine; The Scripps Research Institute, La Jolla, California, U.S.A
| | - J.J. Friedewald
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Medicine-Nephrology; Northwestern University, Chicago, Illinois, U.S.A
| | - L. Gallon
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Medicine-Nephrology; Northwestern University, Chicago, Illinois, U.S.A
| | - I. Konieczna
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A
| | - A.R. Tambur
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| | - j. charette
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| | - J. Levitsky
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Medicine-Hepatology; Northwestern University, Chicago, Illinois, U.S.A
| | - C. Jie
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A
| | - Y. S. Kanwar
- Department of Pathology; Northwestern University, Chicago, Illinois, U.S.A
| | - M. M. Abecassis
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A,Department of Microbiology-Immunology; Northwestern University, Chicago, Illinois, U.S.A
| | - J. Miller
- Comprehensive Transplant Center; Northwestern University, Chicago, Illinois, U.S.A,Department of Surgery- Transplantation; Northwestern University, Chicago, Illinois, U.S.A
| |
Collapse
|
3
|
Hivelin M, Klimczak A, Cwykiel J, Sonmez E, Nasir S, Gatherwright J, Siemionow M. Immunomodulatory Effects of Different Cellular Therapies of Bone Marrow Origin on Chimerism Induction and Maintenance Across MHC Barriers in a Face Allotransplantation Model. Arch Immunol Ther Exp (Warsz) 2015; 64:299-310. [PMID: 26708158 DOI: 10.1007/s00005-015-0380-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/05/2015] [Indexed: 01/31/2023]
Abstract
Many more patients would benefit from vascularized composite allotransplantation if less toxic and safer immunosuppressive protocols will become available. Tolerance induction protocols with donor cells co-transplantation are one of the promising pathways to reduce maintenance immunosupressive regimens. We investigated the role of donor bone marrow cells (BMC), mesenchymal stromal cells (MSC) and in vivo created chimeric cells (CC) used as supportive therapies in a fully MHC-mismatched rat face transplantation model. Twenty-four fully MHC-mismatched hemiface transplantations were performed between ACI (RT1(a)) donors and Lewis (RT1(l)) recipients under combined seven-day immunosuppressive regimen of anti-αβ-T-cell receptor (TCR) monoclonal antibody and cyclosporin A. We studied four experimental groups-group 1: no cellular therapy; group 2: supportive therapy with BMC; group 3: supportive therapy with MSC; group 4: supportive therapy with CC generated in a primary chimera. We evaluated clinical and histological rejection grades, transplanted cells migration, donor-specific chimerism in the peripheral blood and bone marrow compartments, and CD4(+)/CD25(+) T-cell levels. Face allograft rejection was observed at 26.8 ± 0.6 days post-transplant (PT) in the absence of cellular therapy, at 34.5 ± 1.1 days for group 2, 29.3 ± 0.8 days for group 3, and 30.3 ± 1.38 PT for group 4. The longest survival was observed in allografts supported by co-transplantation of BMC. All support in cellular therapies delayed face allograft rejection by chimerism induction and/or immunomodulatory properties of co-transplanted cells. Survival time was comparable between groups, however, further studies, with different cell dosages, delivery routes and delivery times are required.
Collapse
Affiliation(s)
- Mikael Hivelin
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Aleksandra Klimczak
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA.,L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Joanna Cwykiel
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA.,Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA
| | - Erhan Sonmez
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Serdar Nasir
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | | | - Maria Siemionow
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA. .,Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Levitsky J, Mathew JM, Abecassis M, Tambur A, Leventhal J, Chandrasekaran D, Herrera N, Al-Saden P, Gallon L, Abdul-Nabi A, Yang GY, Kurian SM, Salomon DR, Miller J. Systemic immunoregulatory and proteogenomic effects of tacrolimus to sirolimus conversion in liver transplant recipients. Hepatology 2013; 57:239-48. [PMID: 22234876 PMCID: PMC3334454 DOI: 10.1002/hep.25579] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 12/21/2012] [Indexed: 12/12/2022]
Abstract
UNLABELLED Immunosuppression (IS) withdrawal from calcineurin inhibitors is only possible in ≈ 20% of liver transplant recipients. However, mammalian target of rapamycin inhibitors (e.g., sirolimus; SRL) appear to be more immunoregulatory and might promote a tolerant state for withdrawal. Our aim was to determine whether systemic (i.e., blood, marrow, and allograft) signatures of immunoregulation are promoted by conversion from tacrolimus (TAC) to SRL. We therefore performed the following serial assays before and after SRL conversion in liver transplant recipients to test for enhanced markers of immunoregulation: (1) flow-cytometry immunophenotyping of peripheral blood mononuclear cells (PBMCs) and bone marrow aspirates for regulatory T cells (Tregs) (e.g., CD4(+) CD25(+++) FOXP3(+) ) and regulatory dendritic cells (DCregs) (immunoglobulin-like transcript 3(+) /4(+) ); (2) liver biopsy immunohistochemical staining (e.g., FOXP3:CD3 and CD4:CD8 ratios) and immunophenotyping of biopsy-derived Tregs after growth in culture; (3) effects of pre- versus postconversion sera on Treg generation in mixed lymphocyte reactions; (4) peripheral blood nonspecific CD4 responses; and (5) peripheral blood gene transcripts and proteomic profiles. We successfully converted 20 nonimmune, nonviremic recipients (age, 57.2 ± 8.0; 3.5 ± 2.1 years post-liver transplantation) from TAC to SRL for renal dysfunction. Our results demonstrated significant increases in Tregs in PBMCs and marrow and DCregs in PBMCs (P < 0.01) after conversion. In biopsy staining, FOXP3:CD3 and CD4:CD8 ratios were significantly higher after conversion and a number of biopsy cultures developed new or higher FOXP3(+) cell growth. Nonspecific CD4 responses did not change. Both pre- and postconversion sera inhibited mixed lymphocyte reactions, although only TAC sera suppressed Treg generation. Finally, 289 novel genes and 22 proteins, several important in immunoregulatory pathways, were expressed after conversion. CONCLUSIONS TAC to SRL conversion increases systemic Tregs, DCregs, and immunoregulatory proteogenomic signatures in liver transplant recipients and may therefore facilitate IS minimization or withdrawal.
Collapse
Affiliation(s)
- Josh Levitsky
- Division of Gastroenterology & Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - James M. Mathew
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Michael Abecassis
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Anat Tambur
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joseph Leventhal
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Dhivya Chandrasekaran
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Nancy Herrera
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Patrice Al-Saden
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lorenzo Gallon
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Anmaar Abdul-Nabi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sunil M. Kurian
- The Scripps Research Institute, Department of Molecular and Experimental Medicine, La Jolla, CA
| | - Daniel R. Salomon
- The Scripps Research Institute, Department of Molecular and Experimental Medicine, La Jolla, CA
| | - Joshua Miller
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
5
|
Levitsky J, Leventhal JR, Miller J, Huang X, Chen L, Chandrasekaran D, Tambur AR, Mathew JM. Favorable effects of alemtuzumab on allospecific regulatory T-cell generation. Hum Immunol 2011; 73:141-9. [PMID: 22154724 DOI: 10.1016/j.humimm.2011.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 10/12/2011] [Accepted: 11/07/2011] [Indexed: 10/15/2022]
Abstract
We studied the effects of alemtuzumab on T-regulatory cells (Tregs) during alloactivation, first by differences in depletion of various naive versus alloactivated cell subsets in peripheral blood of healthy volunteers, then by adding serial concentrations to human leukocyte antigen (HLA)-DR-matched and -mismatched responding and stimulating cells in mixed lymphocyte reaction (MLR). Lymphoproliferation inhibition and the development of proliferating carboxyfluorescein succinimidyl ester (CFSE)-diluted CD4(+)CD25(high)CD127(-)FOXP3(+) (phenotypic) Tregs by flow cytometry were measured. Also, the ability of alemtuzumab-treated versus nontreated MLR generated CD4(+)CD127(-) cells to allospecifically inhibit MLRs and recruit additional responding Tregs was tested. We found a more pronounced refractoriness of alloactivated versus naive CD4(+)CD25(high) cells to alemtuzumab induced lymphodepletion. Alemtuzumab dose dependently inhibited lymphoproliferation while amplifying percentages of MLR-generated Tregs. This was somewhat augmented by human complement addition. CD127(-)CD4(+) cells immunoselected after 7 days from alemtuzumab-treated MLRs allospecifically inhibited lymphoproliferation and recruited additional Tregs in fresh MLR-responding cells, similar to modulators derived from MLRs without drug addition (media). Addition of tacrolimus and sirolimus to alemtuzumab further inhibited MLR proliferation. However, Treg percentages were markedly higher with sirolimus. These results support the notion that alemtuzumab induces immunoregulation in naïve T cells undergoing alloactivation absent presensitization, especially used in conjunction with maintenance SRL.
Collapse
Affiliation(s)
- Josh Levitsky
- Division of Gastroenterology, Hepatology, Department of Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Infusions of bone marrow-derived cells together with 'space making' continue to be tested in clinical organ transplant tolerance protocols. These trials are based on the hypothesis that this might produce initial multilineage chimerism. There is some evidence that this in turn induces regulatory cells that control alloimmunity. Although a wealth of knowledge is available from animal models, this review deals with what we know or can speculate about donor bone marrow cells and chimerism in human organ transplantation. RECENT FINDINGS Calcineurin inhibitors are employed in most of these protocols to blunt the initial immune response. One protocol also has a stepwise regulatory cell generating treatment with sirolimus before total withdrawal. A number of donor chimeric lineages including stem cells, dendritic cells, myeloid precursors, and various lymphoid subpopulations have been described. Currently, it is recognized that the nature of cells that make up the chimerism could influence graft rejection versus acceptance. Tolerogenic donor chimeric cells may also generate regulatory subsets, thus controlling alloimmunity on two fronts. SUMMARY It might be speculated that prolonged and sustained regulation or possible anergy induced by chimerism may eventually lead to clonal deletion, thereby bringing about classical immunologic tolerance.
Collapse
|
7
|
Gorantla VS, Brandacher G, Schneeberger S, Zheng XX, Donnenberg AD, Losee JE, Lee WPA. Favoring the risk-benefit balance for upper extremity transplantation--the Pittsburgh Protocol. Hand Clin 2011; 27:511-20, ix-x. [PMID: 22051391 DOI: 10.1016/j.hcl.2011.08.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Upper extremity transplantation is an innovative reconstructive strategy with potential of immediate clinical application and the most near-term pay-off for select amputees, allowing reintegration into employment and society. Routine applicability and widespread impact of such strategies for the upper extremity amputees with devastating limb loss could be enabled by implementation of cellular therapies that integrate and unify the concepts of transplant tolerance induction with those of reconstructive transplantation. Such therapies offer the promise of minimizing the risks, maximizing the benefits and optimizing outcomes of these innovative procedures.
Collapse
Affiliation(s)
- Vijay S Gorantla
- Pittsburgh Reconstructive Transplantation Program, Division of Plastic Surgery, Department of Surgery, 3550 Terrace Street, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | |
Collapse
|
8
|
Mathew JM, Blomberg B, Ricordi C, Esquenazi V, Miller J. Evaluation of the tolerogenic effects of donor bone marrow cells using a severe combined immunodeficient mouse-human islet transplant model. Hum Immunol 2008; 69:605-13. [PMID: 18703102 DOI: 10.1016/j.humimm.2008.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 07/02/2008] [Accepted: 07/17/2008] [Indexed: 10/21/2022]
Abstract
The immunoregulatory role of human donor bone marrow cells (DBMC) has been studied extensively in our laboratory using in vitro and ex vivo assays. However, new experimental systems that can overcome the limitations of tissue culture assays but with more clinical relevance than purely animal experimentation, needed to be generated. Therefore we have developed a new human peripheral blood lymphocyte (PBL) severe combined immunodeficient (SCID) mouse islet transplantation model without the occurrence of graft-versus-host disease (GvHD) and have used it to evaluate the tolerogenic effects of DBMC. Nonobese diabetogenic (NOD)-SCID mice were transplanted with human deceased donor islets and were reconstituted with human PBL (allogeneic to islets; denoted as recipient) with or without DBMC from the islet donor. It was observed that the most cellularly economical dose was 3000 islets per animal and that injection into the portal vein was better than implantation under the kidney capsule. Even though maximal lymphoid reconstitution was observed with 40-million fresh and anti-CD3 activated recipient PBL (conventional method), the mice developed severe graft GvHD. However, with the new method of reconstitution where animals were injected with 20-million anti-CD3-activated plus 40-million anti-donor-activated recipient PBL, no discernible GvHD was observed. More importantly, this latter method was associated with islet transplant rejection, which in turn could be abrogated by co-injection of the animals with DBMC. These in vivo results confirmed our previous in vitro observations that human DBMC have regulatory activity.
Collapse
Affiliation(s)
- James M Mathew
- Department of Surgery, Transplant Laboratories, Northwestern University, Chicago, IL, USA.
| | | | | | | | | |
Collapse
|
9
|
Cirocco RE, Carreño MR, Mathew JM, Garcia-Morales RO, Fuller L, Esquenazi V, Ciancio G, Burke GW, Gaynor JJ, Blomberg BB, Rosen A, Kleiner G, Ricordi C, Miller J. FoxP3 mRNA transcripts and regulatory cells in renal transplant recipients 10 years after donor marrow infusion. Transplantation 2007; 83:1611-9. [PMID: 17589345 DOI: 10.1097/01.tp.0000266908.37446.02] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND We update more favorable 10-year deceased donor kidney transplant survival in 63 recipients infused perioperatively with donor vertebral body bone marrow (DBMC-i) vs. 219 noninfused controls having equivalent immunosuppression and demographics. We questioned if this was associated with putatively regulatory FoxP3 mRNA and cell phenotypes (CD4+CD25+high percentages and high DC2:DC1 ratios) in DBMC-i vs. noninfused controls. METHODS Baseline studies were performed on peripheral blood lymphocytes (PBLs) vs. marrow in normal laboratory volunteers of CD4+CD25+high percentages and DC2:DC1 by flow cytometry, and FoxP3 mRNA in CD3+ cells by real-time polymerase chain reaction. Similar studies were performed on PBL of the majority of the 10-year patients remaining with graft function: 21 (of the remaining 37) DBMC-i vs. 55 (of the remaining 105) controls. RESULTS In normal subjects, all parameters were significantly higher in marrow than in PBL, supporting our previous reports of ex vivo DBMC immunoregulation. At 9.8+/-.02 years posttransplant in DBMC-i vs. controls, death-censored percent graft failure was 17.5% vs. 32.9% (P=0.02) with 247.6+/-24 vs. 79.9+/-3.1 (mean+/-SE) FoxP3 copies/5,000 CD3+ cells (P=0.0001). PBL CD4+CD25+high percentages were lower, but DC2:DC1 values higher in both recipient groups than in end-stage renal disease patients who had lower FoxP3 levels (40.8+/-5.9, P<0.0001), consistent with non-CD4+CD25+high T regulatory cells generated long-term posttransplant. Individual higher FoxP3 values correlated with higher iliac crest chimerism in DBMC-i, but not in controls (with 50-fold lower chimerism). In chronically rejecting controls, FoxP3 was further decreased. CONCLUSIONS Peritransplant DBMC-i has higher 10-year renal transplant acceptance, chimerism, and FoxP3 mRNA in thus-far unclarified regulatory cell phenotypes.
Collapse
Affiliation(s)
- Robert E Cirocco
- Department of Surgery, The Lillian Jean Kaplan Renal Transplant Center of the Division of Transplantation, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33101, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wang Z, Davies JD. CD8 blockade promotes the expansion of antigen-specific CD4+ FOXP3+ regulatory T cells in vivo. Int Immunopharmacol 2007; 7:249-65. [PMID: 17178393 PMCID: PMC1797098 DOI: 10.1016/j.intimp.2006.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2006] [Revised: 10/24/2006] [Accepted: 10/26/2006] [Indexed: 11/28/2022]
Abstract
Treatment with a cocktail of CD4 and CD8-specific monoclonal antibodies (mAb) induces long-term transplantation tolerance and regulatory CD4(+) T cells that induce tolerance in non-tolerant T cells. In contrast, treatment with a CD4-specific mAb alone fails to induce long-term tolerance. The current study was designed to test the hypothesis that CD8 blockade plays a role in promoting the development of CD4(+) regulatory T cells. Using the DO11.10 CD4(+) TCR transgenic mouse model we show that treatment with a CD4/CD8-specific mAb cocktail induces antigen-specific tolerance to OVA, measured by a significant decrease in OVA-specific IgG, on challenge with antigen. Although treatment with OVA and the CD4-specific mAb alone also induces a significant decrease in OVA-specific antibody, the number of DO11.10 cells is significantly greater in mice treated with the CD4/CD8-specific mAb cocktail, and this is associated with a significant increase in proliferation of DO11.10 cells in response to specific antigen. DO11.10 cells sorted from mice made tolerant to OVA with the CD4/CD8-specific mAb cocktail promote an OVA-specific IgG1 (Th2-type) response but not an OVA-specific IgG3 (Th1-type) response on transfer into new syngeneic recipients, suggesting their ability to regulate the type of antigen-specific immune response that ensues after priming with antigen. In addition, DO11.10 cells from tolerant mice express markers that are characteristic of CD4(+) regulatory cells, including FOXP3, GITR and CTLA4, but not CD25. Taken as a whole, these data suggest that CD8 blockade promotes CD4(+) FOXP3(+) regulatory CD4(+) T cells by promoting their proliferation in tolerant mice.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens/immunology
- Antigens, CD/immunology
- Antigens, Differentiation/immunology
- CD4 Antigens/immunology
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Female
- Forkhead Transcription Factors/immunology
- Glucocorticoid-Induced TNFR-Related Protein
- Immunoglobulin G/blood
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Ovalbumin/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Nerve Growth Factor/immunology
- Receptors, Tumor Necrosis Factor/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Z Wang
- Torrey Pines Institute for Molecular Studies, San Diego, CA 92121, USA
| | | |
Collapse
|
11
|
Li S, Salgar SK, Thanikachalam M, Murdock AD, Gammie JS, Demetris AJ, Zeevi A, Pham SM. Modeling CTLA4-linked autoimmunity with RNA interference in mice. Proc Natl Acad Sci U S A 2006; 136:238-46. [PMID: 17046021 DOI: 10.1016/j.jss.2006.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 05/02/2006] [Accepted: 05/25/2006] [Indexed: 11/27/2022] Open
Abstract
The CTLA4 gene is important for T lymphocyte-mediated immunoregulation and has been associated with several autoimmune diseases, in particular, type 1 diabetes. To model the impact of natural genetic variants of CTLA4, we constructed RNA interference (RNAi) "knockdown" mice through lentiviral transgenesis. Variegation of expression was observed in founders but proved surmountable because it reflected parental imprinting, with derepression by transmission from male lentigenics. Unlike the indiscriminate multiorgan autoimmune phenotype of the corresponding knockout mice, Ctla4 knockdown animals had a disease primarily focused on the pancreas, with rapid progression to diabetes. As with the human disease, the knockdown phenotype was tempered by genetic-modifier loci. RNAi should be more pertinent than gene ablation in modeling disease pathogenesis linked to a gene-dosage variation.
Collapse
Affiliation(s)
- Sen Li
- Division of Cardiothoracic Surgery, University of Miami School of Medicine, Miami, Florida 33156, USA
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Reich-Zeliger S, Bachar-Lustig E, Gan J, Reisner Y. Tolerance induction by veto CTLs in the TCR transgenic 2C mouse model. I. Relative reactivity of different veto cells. THE JOURNAL OF IMMUNOLOGY 2005; 173:6654-9. [PMID: 15557156 DOI: 10.4049/jimmunol.173.11.6654] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several bone marrow cells and lymphocyte subpopulations, known as veto cells, were shown to induce transplantation tolerance across major histocompatibility Ags. Due to the low frequency of the effector T cells against which the veto cells inhibitory activity is aimed, the fate of the effector cells was traditionally followed indirectly by functional limiting dilution assays, which are cumbersome and depend on numerous parameters. In the present study the fate of the effector T cells was monitored directly by FACS, using TCR transgenic mouse CD8(+) T cells in which the transgene is directed against H-2(d) (the 2C model). This assay is validated by demonstrating the potency, selectivity, radiation sensitivity, and contact dependency of anti-third-party CTLs previously demonstrated by the limiting dilution assay. In contrast to veto CTLs, nonactivated CD8(+) T cells lack veto activity. Comparison by FACS in the 2C model revealed a hierarchy of veto cells, in the order of veto CTLs activated NK cells, activated CD4(+) T cells, and activated B cells. The latter cells as well as nonactivated CD4(+) or NK cells were shown to be completely devoid of veto activity.
Collapse
MESH Headings
- Animals
- Cell Communication/genetics
- Cell Communication/immunology
- Cell Communication/radiation effects
- Cells, Cultured
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic/genetics
- Cytotoxicity, Immunologic/radiation effects
- Female
- Flow Cytometry
- Gamma Rays
- Immune Tolerance/genetics
- Lymphocyte Activation/genetics
- Lymphocyte Activation/radiation effects
- Lymphocyte Culture Test, Mixed
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Transgenic
- Models, Animal
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/radiation effects
Collapse
|
13
|
Jin Y, Fuller L, Ciancio G, Burke GW, Tzakis AG, Ricordi C, Miller J, Esquenzai V. Antigen presentation and immune regulatory capacity of immature and mature-enriched antigen presenting (dendritic) cells derived from human bone marrow. Hum Immunol 2004; 65:93-103. [PMID: 14969764 DOI: 10.1016/j.humimm.2003.11.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2003] [Accepted: 11/20/2003] [Indexed: 11/17/2022]
Abstract
Several reports including those from this laboratory have demonstrated that bone marrow cells (BMC) downregulate in vitro both mixed leukocyte reaction and cytotoxic T lymphocyte reactions. We consequently hypothesized that a general property of immature cells of hematopoietic organs is their ability to suppress immune reactivity. As one of these suppressive activities, the lack of costimulatory molecules was proposed as a mechanism by which immature antigen presenting cells of the bone marrow might be involved. In the present report, we used two culture environments, each of which would regulate a different maturation pattern of human bone marrow-derived enriched dendritic antigen presenting cells (DC or APC) to determine the respective effects on in vitro immune regulatory function. Human BMC depleted of CD3+ cells were cultured with either: interleukin-4 (IL-4) and granulocyte macrophage-colony stimulating factor (GM-CSF), to maintain DC-enriched populations in an immature state (iAPC); or an interferon-gamma (IFNgamma), tumor necrosis factor alpha (TNF-alpha), GM-CSF, LPS, and IL-6 cocktail to promote the maturation of DC-enriched APC (mAPC). These iAPC and mAPC were, respectively, phenotypically characterized and also tested in vitro for the following: (1) both direct and indirect-antigen presentation functions; (2) immune regulatory functions on the response of autologous and allogeneic peripheral blood lymphocytes (PBL); and (3) Western blot analysis determining the levels of both major histocompatibility complex (MHC) class I related cytoplasmic transporter molecules associated with antigen processing (TAP1) and as well as proteasome activator molecules (PA28alpha). The iAPC population expressed fewer dendritic cell markers (CD83 and DCsign), and costimulator molecules (CD86 and CD40) than the mAPC, such that there was an approximate threefold increase in expression of CD83, 2.5-fold increase in DCsign, and a threefold increase in CD40 and CD86 on mAPC than on iAPC (p=0.005 for CD83; p=0.001 for DCsign; p=0.001 for CD86; and p=0.001 for CD40). In lymphoproliferative assays, indirect and direct alloantigen presentation by iAPC was weaker than by mAPC (p=0.05 and 0.04). In addition, iAPC were able to downregulate allogeneic CTL responses. Also, after pulsing with Epstein-Barr virus (EBV) protein antigens, the iAPC were less efficient in their presentation to autologous EBV-specific T-cell lines, and caused an inhibition of EBV-CTL generation. The expression of TAP1 and PA28alpha was reduced in iAPC in comparison to mAPC. These findings support the notion that a maturation state of BMC-derived APC correlates with their capacity to present antigen. The observed in vitro deficiency of this function by immature bone marrow cells may therefore contribute to the immune downregulatory capacity seen in the BMC compartment.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2
- ATP-Binding Cassette Transporters/analysis
- ATP-Binding Cassette Transporters/metabolism
- Antigen Presentation/immunology
- Antigens, CD/metabolism
- Antigens, Viral/immunology
- Antigens, Viral/pharmacology
- B-Lymphocytes/immunology
- B-Lymphocytes/virology
- B7-2 Antigen
- Blotting, Western
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/immunology
- CD3 Complex/analysis
- CD3 Complex/immunology
- CD40 Antigens/analysis
- CD40 Antigens/metabolism
- Capsid Proteins/immunology
- Capsid Proteins/pharmacology
- Cell Adhesion Molecules/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Clonal Anergy/immunology
- Cytokines/pharmacology
- Cytotoxicity, Immunologic/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/physiology
- Epstein-Barr Virus Nuclear Antigens/immunology
- Epstein-Barr Virus Nuclear Antigens/pharmacology
- Flow Cytometry
- Humans
- Immunoglobulins/metabolism
- Interleukins/pharmacology
- Isoantigens/immunology
- Isoantigens/metabolism
- Lectins, C-Type/metabolism
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/radiation effects
- Lipopolysaccharides/pharmacology
- Lymphocyte Activation/immunology
- Lymphocyte Culture Test, Mixed
- Membrane Glycoproteins/metabolism
- Mitomycin/pharmacology
- Muscle Proteins/analysis
- Muscle Proteins/metabolism
- Proteasome Endopeptidase Complex
- Receptors, Cell Surface/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- CD83 Antigen
Collapse
Affiliation(s)
- Yide Jin
- Department of Surgery, University of Miami School of Medicine, Miami, FL 33101, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Mathew JM, Garcia-Morales RO, Carreno M, Jin Y, Fuller L, Blomberg B, Cirocco R, Burke GW, Ciancio G, Ricordi C, Esquenazi V, Tzakis AG, Miller J. Immune responses and their regulation by donor bone marrow cells in clinical organ transplantation. Transpl Immunol 2003; 11:307-21. [PMID: 12967784 DOI: 10.1016/s0966-3274(03)00056-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Infusions of donor bone marrow derived cells (DBMC) continue to be tested in clinical protocols intended to induce specific immunologic tolerance of solid organ transplants based on the observations that donor-specific tolerance is induced this way in animal models. We studied the immunological effects of human DBMC infusions in renal transplantation using modifications in lymphoproliferation (MLR) and cytotoxicity (CML) assays. The salient observations and tentative conclusions are summarized in this review. Among many types of organs transplanted using DBMC at this center, it was found that the cadaver renal recipients (CAD) had significantly decreased chronic rejection and higher graft survival when compared to equivalent non-infused controls. DBMC infusion was also associated with a marginal and non-specific immune depression. It was also observed that the number of chimeric donor cells gradually increased in the iliac crest bone marrow compartment with a concomitant decrease in the peripheral blood and that the increase was more rapid in living-related donor (LRD)-kidney/DBMC recipients in spite of a lower number of DBMC infused (<25%) than in the CAD-kidney/DBMC group. In the LRD recipients with residual anti-donor responses, purified chimeric cells of either donor or recipient inhibited recipient immune responses to the donor significantly more strongly than the freshly obtained bone marrow from the specific donor or volunteer suggesting an active regulatory role for chimeric cells. A number of (non-chimeric) subpopulations of bone marrow cells including CD34(+) stem cells and the CD34(-) early progeny like CD38(+), CD2(+), CD5(+) and CD1(+) lymphoid cells as well as CD33(+) (but CD15(-)) myeloid cells down-regulated the MLR and CML responses of allogeneic PBMC stimulated with (autologous) donor spleen cells. These regulatory effects appeared to be refractory to the action of commonly used immunosuppressive drugs and occurred during the early phase of the immune response through cell-cell interactions. Most of these DBMC sub-populations had stimulatory capabilities, albeit markedly lower than donor spleen cells, but only through the indirect antigen presentation pathway. When co-cultured with allogeneic stimulators, purified CD34(+) cells were found to give rise both to CD3(-) TCRalphabeta(+), as well as CD3(+) TCRalphabeta(+) cells and, thereby, responded in MLR to allogeneic stimulation (but did not generate cytotoxic effector cells). Also, a number of DBMC subpopulations inhibited the CML and to a lesser extent the MLR, of autologous post-thymic responding T cells stimulated with allogeneic irradiated cells, mediated through soluble factors. Finally, non-chimeric DBMC also inhibited the proliferative and cytotoxic responses of autologous T cells to EBV antigens, inducing T suppressor cells, which in turn could inhibit autologous anti-EBV CTL generation and B cell anti-CMV antibody production. These studies all suggested a strong inhibitory property of a number of DBMC sub-populations in vitro and in vivo with the notion that they promote unresponsiveness.
Collapse
Affiliation(s)
- James M Mathew
- Department of Surgery, Division of Transplantation (R-440), University of Miami School of Medicine, 1600 N.W. 10th Ave., Miami, FL 33136, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Reisner Y, Gur H, Reich-Zeliger S, Martelli MF, Bachar-Lustig E. Hematopoietic stem cell transplantation across major genetic barriers: tolerance induction by megadose CD34 cells and other veto cells. Ann N Y Acad Sci 2003; 996:72-9. [PMID: 12799285 DOI: 10.1111/j.1749-6632.2003.tb03235.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Studies in mice and humans demonstrate that transplantation of hematopoietic progenitors in numbers larger than commonly used ("megadose" transplants) overcomes major genetic barriers. In vitro studies suggest that veto cells, within the population of hematopoietic progenitors, facilitate this favorable outcome. Thus, when purified CD34(+) cells were added to bulk mixed-lymphocyte reactions (MLRs) they suppressed CTLs against the donor's stimulators, but not against stimulators from a third party. This tolerizing activity depends on cell contact and can be blocked by the caspase inhibitor BD-FMK, suggesting that the effector host T cells are deleted by apoptosis upon interaction with the CD34(+) cells. Early myeloid CD33(+) cells generated by short-term ex vivo expansion of CD34(+) cells also exhibit veto activity, and these cells can be grown in large numbers. Tolerance induction can be further enhanced by other veto cells. Perhaps the most potent veto cell is the CD8+ CTL. However, this cell is also associated with marked GVHD (graft-versus-host disease. GVHD can be separated from the veto activity by generating anti-third party CTLs under IL2 deprivation. Under such selective pressure only the stimulated clones which make IL2 can survive, while anti-host clones die. In vivo studies show that such anti-third party veto CTLs can be used safely for tolerance induction without GVHD.
Collapse
Affiliation(s)
- Yair Reisner
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | | | | | | | | |
Collapse
|
16
|
Mathew JM, Blomberg B, Fuller L, Burke GW, Ciancio G, Kenyon N, Ricordi C, Tzakis AG, Esquenazi V, Miller J. A novel micro-cell-mediated lympholytic assay for the evaluation of regulatory cells in human alloreactive CTL responses. J Immunol Methods 2003; 272:67-80. [PMID: 12505713 DOI: 10.1016/s0022-1759(02)00432-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Since cell-mediated lympholysis (CML), the most commonly used in vitro experimental cytotoxic method for the evaluation of regulatory cells, requires large numbers of cells that are often the limiting factor, we have developed a new micro-cell-mediated lympholytic (m-CML) assay. Various numbers of responding cells were stimulated with equivalent numbers of allogeneic irradiated stimulator cells in the presence of (fivefold) serial dilutions of regulatory cells. On the 8th day, 4-h 51Cr-release assays were performed by adding 5000 labeled target cells from the corresponding stimulators to the cultures. Even though results that were comparable to the macro- (bulk) CML and MLR modulation experiments were obtained with all the cell combinations tested in the m-CML, the combinations with 50,000 responder cells and stimulating cells and dilutions of 25,000 to 40 modulator (regulatory) cells were found to be the most reproducible for assaying regulatory cell potency in vitro. Similarly, expression of the results as percentage inhibition using percent specific lysis values was the simplest method of calculation. This assay was standardized for the evaluation of the inhibitory activity of a variety of regulatory cells, including long-term cultures of cadaver-donor vertebral body bone marrow cells (vDBMC-L), in vitro generated CD8 positive and CD28 negative suppressor T cells and donor chimeric cells isolated from renal transplant recipients who had been perioperatively infused with donor bone marrow cells (DBMC). The results indicate that the m-CML assay is a sensitive and reliable micromethod with at least 10-fold fewer responders, stimulator and modulator cell numbers needed than macro-CML assays for the evaluation of regulatory cells obtained from a variety of immune systems in vitro.
Collapse
Affiliation(s)
- James M Mathew
- Division of Transplantation, Department of Surgery, University of Miami School of Medicine, R-440, 1600 N.W. 10th Ave., FL 33136, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Carreno MR, Fuller L, Mathew JM, Ciancio G, Burke GW, Esquenazi V, Ricordi C, Tzakis AG, Miller J. Human donor bone marrow cells induce in vitro "suppressor T cells" that functionally suppress autologous B cells. Hum Immunol 2003; 64:21-30. [PMID: 12507811 DOI: 10.1016/s0198-8859(02)00774-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have reported a beneficial effect of donor vertebral body bone marrow cells (DBMC) infusions in cadaver renal allograft recipients in a 6-year follow-up, but with a transient increase in early (6 month) postoperative CMV infections and concomitant suppressed immunoglobulins (Ig) production. We also found that although there was no difference between the DBMC-infused and non-infused (control) groups in the development of donor-specific antibody, we now describe an additional difference seen in the percent reactive antibody (PRA) reactivity against a panel of HLA antigens that developed postoperatively. We hypothesize that (allogeneic) antigen presenting cells in the DBMC, systemically infused, caused the generation of recipient T suppressor (T4-suppressor) cells, thereby "inducing" a negative influence on B cell Ig production. We tested this notion in vitro by incubating PBL from CMV IgG positive laboratory volunteers with either (allogeneic) T-cell depleted DBMC or donor spleen cells (DSPC) from (the same) cadaver donors. After 7 days, the (responding) T cells were collected using magnetic beads and placed in culture with purified B cells freshly obtained from the same (autologous) CMV positive volunteer. To these cultures were added either media or 40 ng of CMV antigen. After 3, 5, 7, and 9 days, the expression of surface anti-CMV Ig was measured by flow cytometry using a panel of fluorescent markers double-labeled for activated B cells (CD20, CD19, and HLA DRw) and CMV-FITC. We also determined the phenotype of the cultured T cells using anti-CD3, CD4, and CD62L specific monoclonal antibodies. B cells that had been in contact with autologous T cells derived from DBMC cultures (TBM) were less likely to express anti-CMV surface Ig than those cultured with DSPC (TSP). The flow cytometry analysis revealed an increase in the number of T4 suppressor cells (CD3+, CD4+, CD62L+) in the TBM group, whereas the T4 helper phenotype (CD3+, CD4+, CD62L-) predominated in the TSP group. These in vitro findings support the notion that (allogeneic) DBMC infusions can induce a T4 suppressor (regulatory) influence and thereby indirectly affect B-cell function.
Collapse
Affiliation(s)
- Manuel R Carreno
- Departments of Surgery, Division of Transplantation, University of Miami School of Medicine, Miami, FL 33101, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Mathew JM, Carreno M, Fuller L, Burke GW, Ciancio G, Ricordi C, Tzakis AG, Esquenazi V, Miller J. Regulation of alloimmune responses (GvH reactions) in vitro by autologous donor bone marrow cell preparation used in clinical organ transplantation. Transplantation 2002; 74:846-55. [PMID: 12364866 DOI: 10.1097/00007890-200209270-00019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cadaver donor bone marrow cells (DBMC) are capable of a low-grade response to allogeneic stimulation in vitro, indicating their potential ability to cause graft versus host disease (GvHD). However, at this center, we have observed a lack of GvHD in kidney transplant recipients who received DBMC perioperatively. Therefore, we questioned whether an intrinsic immunoregulatory function of (subpopulations of) DBMC might play a role in this observation. METHODS In in vitro assays, DBMC was added to autologous splenic responder cells taking part in allogeneic mixed lymphocyte reaction (MLR) and cell-mediated lympholysis (CML) reactions. RESULTS When compared with autologous donor irradiated spleen cells as control modulators, DBMC significantly inhibited the CML, but to a much lesser extent than MLR, of autologous responding cells stimulated with allogeneic irradiated cells in a dose dependent manner. The down-regulation of CML responses was observed even in the presence of pharmacological concentrations FK506, mycophenolic acid, or cyclosporine-A. The inhibition could not be overcome by the addition of exogenous helper factors. Moreover, in contrast with findings previously reported from this laboratory of the in vitro inhibitory effect of DBMC on allogeneic responding cells in MLR and CML reactions to stimulating cells of the DBMC donor (allogeneic host versus graft inhibition), the following unique observations were made using DBMC inhibiting autologous reactions (GvH): (1) optimal restimulation of autologous responder cells in secondary cultures could not abrogate this inhibition; even activated responder cells could be inhibited by autologous DBMC, and (2) soluble factors were at least partially operative in this autologous (GvH) inhibition as indicated by experiments in transwells and by the CML inhibition caused by 50% supernatants from DBMC cultures. CONCLUSION These in vitro results indicate that DBMC treatment brings about a marked down-regulation of autologous immunocompetent cells in cytotoxicity reactions, partially at least through secreted soluble factor(s), (and in presence of immunosuppressive drugs in vivo) thereby preventing overt clinical GvHD.
Collapse
Affiliation(s)
- James M Mathew
- Department of Surgery, University of Miami School of Medicine, Florida 33136, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mathes DW, Randolph MA, Bourget JL, Nielsen GP, Ferrera VR, Arn JS, Sachs DH, Lee WPA. Recipient bone marrow engraftment in donor tissue after long-term tolerance to a composite tissue allograft. Transplantation 2002; 73:1880-5. [PMID: 12131681 DOI: 10.1097/00007890-200206270-00005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND An important component of a composite tissue limb allograft (CTA) is the vascularized bone marrow and bone marrow stroma, which when transplanted could create immediate marrow space and engraftment. We have previously demonstrated that tolerance to musculoskeletal allografts can be achieved with a 12-day course of cyclosporine without the presence of long-term peripheral donor cell chimerism. The objective of this study was to determine the fate of the donor bone marrow after transplantation of a limb allograft in a miniature swine model. METHODS CTAs from donor swine were heterotopically transplanted into six MHC-matched, minor-antigen-mismatched recipients, and a 12-day course of cyclosporine was given. Previous animals transplanted without cyclosporine rejected their grafts in less than 42 days. A non-MHC-linked marker, pig allelic antigen (PAA), was used to distinguish host and donor cells. Three PAA- animals received PAA+ CTAs, and three PAA+ animals received PAA- CTAs. Bone marrow was harvested from the donor limb grafts and the recipient and analyzed by flow cytometry and histology. Thymus, spleen, and mesenteric lymph nodes were also harvested from the recipient swine and evaluated for the presence of donor cells by flow cytometry. RESULTS All animals receiving cyclosporine demonstrated permanent tolerance to their allografts. Donor bone marrow cells were present in all grafts at the time of transplantation and during the immediate postoperative period. By 48 weeks, donor cells were no longer detectable within the marrow space of the allograft. In long-term animals host bone marrow cells replaced donor cells in the graft marrow space. No evidence of donor cell engraftment was found in recipient animals. CONCLUSION This study demonstrates that in long-term tolerant recipients of musculoskeletal allografts there is no evidence of persistent donor bone marrow cells in the hematopoietic tissues of the graft or the host. Rather, the recipient's bone marrow cells and lymphocytes repopulate the donor marrow space of the graft.
Collapse
Affiliation(s)
- David W Mathes
- Divisions of Plastic Surgery, Transplantation Biology Research Center, Harvard Medical School and the Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Exley MA, Tahir SM, Cheng O, Shaulov A, Joyce R, Avigan D, Sackstein R, Balk SP. A major fraction of human bone marrow lymphocytes are Th2-like CD1d-reactive T cells that can suppress mixed lymphocyte responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5531-4. [PMID: 11698421 DOI: 10.4049/jimmunol.167.10.5531] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Murine bone marrow (BM) NK T cells can suppress graft-vs-host disease, transplant rejection, and MLRs. Human BM contains T cells with similar potential. Human BM was enriched for NK T cells, approximately 50% of which recognized the nonpolymorphic CD1d molecule. In contrast to the well-characterized blood-derived CD1d-reactive invariant NK T cells, the majority of human BM CD1d-reactive T cells used diverse TCR. Healthy donor invariant NK T cells rapidly produce large amounts of IL-4 and IFN-gamma and can influence Th1/Th2 decision-making. Healthy donor BM CD1d-reactive T cells were Th2-biased and suppressed MLR and, unlike the former, responded preferentially to CD1d(+) lymphoid cells. These results identify a novel population of human T cells which may contribute to B cell development and/or maintain Th2 bias against autoimmune T cell responses against new B cell Ag receptors. Distinct CD1d-reactive T cell populations have the potential to suppress graft-vs-host disease and stimulate antitumor responses.
Collapse
Affiliation(s)
- M A Exley
- Cancer Biology Program, Hematology/Oncology, Beth Israel-Deaconess Medical Center, Bone Marrow Transplant Program, Harvard Skin Disease Research Center, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Garcia-Morales RO, Ciancio G, Mathew J, Jin Y, Rosen A, Ricordi C, Burke GW, Blomberg B, Fuller L, Tzakis AG, Esquenazi V, Miller J. Perioperative donor bone marrow infusion in cadaver kidney transplant recipients. Transplant Proc 2001; 33:3840-3. [PMID: 11750636 DOI: 10.1016/s0041-1345(01)02626-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- R O Garcia-Morales
- Department of Surgery, Division of Transplantation, the Diabetes Research Institute, University of Miami School of Medicine and Miami Veterans Affairs Medical Center, Miami, Florida 33101, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mathew JM, Miller J. Immunoregulatory role of chimerism in clinical organ transplantation. Bone Marrow Transplant 2001; 28:115-9. [PMID: 11509928 DOI: 10.1038/sj.bmt.1703110] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
23
|
Ciancio G, Miller J, Garcia-Morales RO, Carreno M, Burke GW, Roth D, Kupin W, Tzakis AG, Ricordi C, Rosen A, Fuller L, Esquenazi V. Six-year clinical effect of donor bone marrow infusions in renal transplant patients. Transplantation 2001; 71:827-35. [PMID: 11349712 DOI: 10.1097/00007890-200104150-00002] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND To date, several single- and multicenter clinical trials have attempted to induce specific immunological unresponsiveness using donor bone marrow cell infusions to augment solid organ transplantation, but the outcomes have not been definitive. METHODS Between September 1994 and May 1998, 63 cadaver (CAD) renal transplant recipients of either one or two postoperative donor bone marrow cell (DBMC) infusions were prospectively compared with 219 non-infused controls given equivalent immunosuppression. There was at least a 1 HLA DR antigen match present between donors and recipients. The immunosuppressive regimen included a 10-day course of OKT3 induction, and tacrolimus, mycophenolate mofetil, and methylprednisolone maintenance. A total 7.01x10(8)+/-1.9x10(8) (SD) DBMC/kg was infused into the CAD recipients on either days 4 and 11 (n=42) or one half of that dose on day 4 (n=21) postoperatively. Clinical follow-up has ranged from 2.9 to 6.3 years (mean, 4.7 years). Studies were also performed of humoral immunity and quantitative cellular chimerism. RESULTS There is clear-cut equivalence in immunosuppressive dosaging and in the other major demographic variables in both groups. However, only 2/63 DBMC recipients had biopsy-proven chronic rejection, whereas 41/219 showed chronic rejection in the controls (P = <0.01). In both groups, mortality was not associated with rejection. The actuarial graft survival at 6.3 years in the CAD DBMC group was 84.3% compared with 72.2% in the control group (not statistically significant). However, if death with a functioning graft was excluded, graft survival was 94.1% in the DBMC group and 79.8% in the controls (P=0.039). Forty patients in the control group continue to have deteriorating renal function (increasing serum creatinine concentrations to 2 mg/dl and higher), compared with 2 patients in the DBMC group (P=0.04). In the DBMC group, chimerism in iliac crest marrow aspirates has increased 3-fold in yearly sequential measurements between 1 and 4 years postoperatively averaging 1.3+/-0.36% (SE) most recently. This has not occurred in the controls. CONCLUSIONS There now appears to be more solid long-term evidence, in kidney transplant recipients prospectively receiving DBMC infusions, of an improvement in long-term graft survival, and of the degree of chimerism positively correlating with the absence of graft loss.
Collapse
Affiliation(s)
- G Ciancio
- Department of Surgery, Diabetes Research Institute, University of Miami School of Medicine, Florida 33101, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mathew JM, Garcia-Morales R, Fuller L, Rosen A, Ciancio G, Burke GW, Carreno M, Temple D, Tzakis AG, Ricordi C, Miller J, Esquenazi V. Donor bone marrow-derived chimeric cells present in renal transplant recipients infused with donor marrow. I. Potent regulators of recipient antidonor immune responses. Transplantation 2000; 70:1675-82. [PMID: 11152096 DOI: 10.1097/00007890-200012270-00003] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Even though a number of transplant centers have adopted donor-specific bone marrow cell (DBMC) infusions to enhance donor cell chimerism, to date there has been no direct evidence linking chimerism with tolerance induction in human organ transplant recipients. METHODS Cells of donor phenotype were isolated 1 year postoperatively from the peripheral blood lymphocytes and iliac crest bone marrow of 11 living-related-donor (LRD) renal transplant recipients, who had received perioperative donor bone marrow cell infusions. These recipient-derived donor (RdD) cells were characterized phenotypically by flow cytometric analysis and functionally as modulators in mixed lymphocyte reaction (MLR) and cell-mediated lympholysis (CML) assays. RESULTS The yield of RdD cells ranged from 0.1 to O.9% of the starting material with the majority being TcRalphabeta, CD3 positive T cells, a substantial percentage of which coexpressed CD28. At 1 year posttransplant almost 50% of the LRD-kidney/DBMC recipients tested so far exhibited donor-specific unresponsiveness in MLR (7/17) and CML (6/13) reactions and this trend was further enhanced at 23 years. In the recipients with residual positive antidonor immune responses, the RdD cells inhibited recipient antidonor MLR and CML responses significantly more strongly than freshly isolated and similarly treated iliac crest bone marrow cells from the donor. RdD cells also inhibited the MLR of the recipient to third party allogeneic stimulator cells; however, this nonspecific effect was significantly weaker than specific inhibition. We also established long-term bone marrow cultures stimulated every 2 weeks with irradiated alogeneic feeder cells, that had similar functional properties thus possibly providing us with an in vitro correlate the RdD cells. CONCLUSIONS These results clearly support the notion that the infused donor cells play a positive role in the induction and/or maintenance of transplant tolerance.
Collapse
Affiliation(s)
- J M Mathew
- Department of Surgery, Research Institute, University of Miami School of Medicine, and the Miami Veterans Affairs Medical Center, FL 33136, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|