scholarly journals CC Chemokine Receptor 2 Expression in Donor Cells Serves an Essential Role in Graft-versus-Host-Disease

2003 ◽  
Vol 171 (9) ◽  
pp. 4875-4885 ◽  
Author(s):  
Arun R. Rao ◽  
Marlon P. Quinones ◽  
Edgar Garavito ◽  
Yogeshwar Kalkonde ◽  
Fabio Jimenez ◽  
...  
2019 ◽  
Vol 19 (7) ◽  
pp. 1941-1954 ◽  
Author(s):  
Kenneth A. Fowler ◽  
Viktoria Vasilieva ◽  
Ekaterina Ivanova ◽  
Olga Rimkevich ◽  
Andrey Sokolov ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (5) ◽  
pp. 825-836 ◽  
Author(s):  
James M. Coghill ◽  
Kenneth A. Fowler ◽  
Michelle L. West ◽  
LeShara M. Fulton ◽  
Hendrik van Deventer ◽  
...  

Key Points Extended donor Treg survival is required for protection from GVHD; donor Treg longevity depends on Treg CCR8 expression. Donor CD11c+ APCs promote Treg longevity in vivo; host CD11c+ APCs do not appear to contribute to donor Treg reconstitution.


Blood ◽  
2010 ◽  
Vol 115 (23) ◽  
pp. 4914-4922 ◽  
Author(s):  
James M. Coghill ◽  
Michael J. Carlson ◽  
Angela Panoskaltsis-Mortari ◽  
Michelle L. West ◽  
Joseph E. Burgents ◽  
...  

Abstract CC-chemokine receptor 7 (CCR7) is expressed on the surface of naive T cells, and plays a critical role in their movement into secondary lymphoid tissue. Here, we show that murine T cells lacking CCR7 (CCR7−/−) generate attenuated graft-versus-host disease (GVHD) responses compared with wild-type (WT) cells, with the difference varying inversely with the degree of major histocompatibility complex (MHC) disparity between the donor and recipient. CCR7−/− T cells exhibited an impaired ability to traffic to recipient lymph nodes, with an increased capacity to home to the spleen. CCR7−/− T cells, however, demonstrated a reduced ability to undergo in vivo expansion in the spleen due to impaired interactions with splenic antigen-presenting cells. On a cellular level, CCR7−/− T cells were functionally competent, demonstrating a normal in vitro proliferative capacity and a preserved ability to produce inflammatory cytokines. Importantly, CCR7−/− T cells were capable of generating robust graft-versus-leukemia (GVL) responses in vivo, as well as complete donor T-cell reconstitution. CCR7−/− regulatory T cells were able to protect against lethal GVHD when administered before WT conventional T cells. Our data suggest that CCR7 inhibition in the early posttransplantation period may represent a feasible new therapeutic approach for acute GVHD attenuation without compromising GVL responses.


2015 ◽  
Vol 21 (12) ◽  
pp. 2069-2078 ◽  
Author(s):  
Alina Schreder ◽  
Georgios Leandros Moschovakis ◽  
Stephan Halle ◽  
Jerome Schlue ◽  
Chun-Wei Lee ◽  
...  

2010 ◽  
Vol 16 (3) ◽  
pp. 311-319 ◽  
Author(s):  
Lisa A. Palmer ◽  
George E. Sale ◽  
John I. Balogun ◽  
Dan Li ◽  
Dan Jones ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1337-1337
Author(s):  
Michael J Carlson ◽  
James M. Coghill ◽  
Michelle L. West ◽  
Angela Panoskaltsis-Mortari ◽  
Bruce R. Blazar ◽  
...  

Abstract Abstract 1337 Poster Board I-359 INTRODUCTION Graft-versus-host disease (GVHD) is a major complication following allogeneic bone marrow transplantation (BMT). Despite advances in understanding the etiology of GVHD it remains a formidable obstacle to the widespread application of BMT. A number of studies have demonstrated that T regulatory (Treg) cells represent a potential therapy for GVHD as Tregs have been shown to inhibit GVHD while preserving the beneficial graft-versus-leukemia (GVL) effect. Numerous groups, including our own, have demonstrated the importance of T cell migration in the pathology of GVHD. Following conditioning, donor T cells migrate to secondary lymphoid tissues. Once activated in the lymphatics, T cells migrate to GVHD target organs including; the skin, liver, lung and the gastrointestinal (GI) tract in response to the local production of chemokines. Disruption of chemokine-chemokine receptor interactions has been demonstrated to affect the pathology of GVHD. Previously, we have shown that Tregs lacking the chemokine receptor CCR5, which binds CCL3, CCL4, and CCL5, do not protect animals from lethal GVHD as well as WT Tregs, due to their impaired migration to the liver and lung. Thus, a greater understanding of the function of chemokine receptors on Tregs is important in deciphering how Tregs function and whether targeting these cells to lymphoid tissue or GVHD target organs would be preferable for treating patients in clinical trials. METHODS We utilized a parent into F1 haploidentical model to assess the role of CCR1 in Treg-mediated protection from GVHD. Here we demonstrate Tregs lacking CCR1, another receptor for CCL3 and CCL5, were unable to protect animals against lethal acute GVHD. While 67% of B6D2 recipients given 1×106 WT Tregs supplemented with 5×106 WT T cells and 3×106 B6 T cell-depleted BM cells survived, only 15% of the recipients given CCR1−/− Tregs survived (p < 0.03; Fisher's exact test). B6D2 recipient mice given WT Tregs had significantly reduced clinical scores for GVHD compared to B6D2 recipients of CCR1−/− Tregs (p <0.05) with elevated GVHD scores starting on day 28 post-transplant. Histopathology revealed significantly worse pathology in the liver (p < 0.03) and colon (p < 0.05) of CCR1−/− Treg recipients vs. WT Treg recipients. In vitro analysis demonstrated that CCR1−/− Tregs were capable of suppressing T cell responses to allo-antigen equally as well as WT Tregs, and CCR1−/− Tregs attained a normal activation phenotype. Interestingly, preliminary experiments suggested that CCR1−/− Tregs migrated to and/or expanded in GVHD target organs to a similar extent as WT Tregs. CONCLUSIONS Treg expression of CCR1 is required for the inhibition of GVHD. Tregs lacking CCR1 led to significantly more tissue destruction in the liver and colon, two predominant sites of GVHD pathology. Of interest, the migration of CCR1−/− Tregs to GVHD target organs and secondary lymphoid tissues did not appear to be compromised suggesting that CCR1 may be required for the function of Tregsin vivo. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3736-3736
Author(s):  
Sam C Nalle ◽  
Peter A Savage ◽  
Jerrold R Turner

Abstract Abstract 3736 Background Graft-versus-host disease (GVHD) is a potentially fatal complication following allogeneic bone marrow transplantation (BMT). GVHD is characterized by three phases: (1) recipient tissue injury mediated by the conditioning regimen of irradiation and/or chemotherapy; (2) donor cell priming and activation; and (3) effector destruction of target tissue such as the intestine, liver, skin, and lung. There is some evidence to suggest that the second and third phases are dependent on the first, however, this has not been rigorously tested. Therefore, we developed major and minor mismatch models of GVHD and used immunodeficient recipients that readily accept allografts to analyze the requirement of irradiation conditioning on GVHD initiation and development. Experimental Design Wild-type (WT) B6 (H-2b) recipients received major mismatch (Balb/c, H-2d), minor mismatch (129, H-2b), or syngeneic (B6) BMTs. Recipients were lethally irradiated, and 24 hours later received an intravenous infusion of 5 × 106 bone marrow cells and 30 × 106 splenocytes, as a source of mature T cells. In adoptive transfer (AT) experiments, B6 Rag1−/− or B6 Rag2−/−/Pfp−/− (perforin deficient) recipients received 30 × 106 splenocytes. Data are represented as mean ± SEM. Results In lethally irradiated WT recipients, Balb/c→B6 caused severe GVHD that was fatal in all recipients by 24 days after BMT. 129→B6 led to a milder GVHD, where 50% of recipients survived to 35 days after BMT, and a few survived long-term (>100 days). Both major and minor mismatch GVHD were characterized by 15–25% weight loss, clinical symptoms including decreased activity, hunched posture, ruffled fur, and hair loss, and target organ histopathology. To test the requirement of irradiation conditioning in GVHD, we transferred Balb/c splenocytes into unconditioned B6 Rag1−/− recipients. No signs of GVHD developed. However, donor T cells were virtually undetectable 5 weeks after AT, indicating graft rejection. To test if this was due to recipient natural killer (NK) cells, a major mismatch AT was performed into B6 Rag2−/−/Pfp−/− recipients, which lack fully functional NKs. Balb/c→B6 Rag2−/−/Pfp−/− resulted in GVHD, including 10–15% weight loss, clinical symptoms, and target organ histopathology, although the disease was not as severe as that following lethal irradiation of WT recipients, consistent with a facilitative role for conditioning in disease progression. In contrast to the above, a 129 splenocyte AT did not cause GVHD in Rag1−/− or Rag2−/−/Pfp−/− recipients, despite donor T cell engraftment. This suggested that conditioning was required for minor mismatch GVHD. To test this, we sublethally irradiated Rag1−/− recipients prior to 129 AT. This resulted in GVHD, with 10–15% weight loss, clinical symptoms, and histopathology. At day 7 after AT, serum IFNγ, TNF, and IL-6 were significantly greater in mice that received irradiation and AT (compared to AT alone, p<0.05). Donor cells within spleens of mice that received irradiation and AT had a reduced fraction of Foxp3+/CD4+ (9.5%±2.3) compared to AT alone (16.8%±1.6, p=0.06), and increased intracellular CD8+ IFNγ expression (%IFNγ+:47.7±2.4 vs. 41.6±1.8, p<0.05; MFI:15722±2003 vs. MFI:8025±319, p<0.05). To determine whether more alloreactive donor cells were primed after irradiation, an in vivo killing assay was performed with recipient-specific targets. Combined irradiation and AT resulted in 66.3±13.5% killing efficiency while AT only had an 18.9±11.7% killing efficiency. Conclusions We conclude that irradiation conditioning is required for minor, but not major mismatch GVHD. The conditioning regimen creates the necessary proinflammatory milieu to prime sufficient numbers of alloreactive cells for GVHD. While this milieu can enhance development of major mismatch GVHD, it is not required for development of disease. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 109 (9) ◽  
pp. 4097-4104 ◽  
Author(s):  
Yanhui Xu ◽  
Andrew S. Flies ◽  
Dallas B. Flies ◽  
Gefeng Zhu ◽  
Sudarshan Anand ◽  
...  

Abstract Decoy lymphotoxin β receptor (LTβR) has potent immune inhibitory activities and thus represents a promising biologic for the treatment of inflammation, autoimmune diseases, and graft-versus-host disease (GVHD). As this reagent interrupts multiple molecular interactions, including LTβ-LTβR and LIGHT-HVEM/LTβR, underlying molecular mechanisms have yet to be fully understood. In this study, we demonstrate that blockade of the LIGHT-HVEM pathway is sufficient to induce amelioration of GVHD in mouse models. Anti–host cytotoxic T lymphocyte (CTL) activity following in vivo transfer of allogeneic lymphocytes was completely abrogated when LIGHT- or HVEM-deficient (KO) T cells were used as donor cells. Accordingly, survival of the recipient mice following the transfer of allogeneic bone marrow cells plus LIGHT-KO or HVEM-KO T cells was significantly prolonged. In the absence of LIGHT-HVEM costimulation, alloreactive donor T cells undergo vigorous apoptosis while their proliferative potential remains intact. Furthermore, we prepared a neutralizing monoclonal antibody (mAb) specific to HVEM and showed that administration of anti–HVEM mAb profoundly ameliorated GVHD and led to complete hematopoietic chimerism with donor cells. Collectively, our results demonstrate an indispensable role of LIGHT-HVEM costimulation in the pathogenesis of GVHD and illustrate a novel target for selective immunotherapy in allogeneic bone marrow transplantation.


Sign in / Sign up

Export Citation Format

Share Document