Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Thomas B. Thornley is active.

Publication


Featured researches published by Thomas B. Thornley.


Journal of Immunology | 2006

TLR Agonists Abrogate Costimulation Blockade-Induced Prolongation of Skin Allografts

Thomas B. Thornley; Michael A. Brehm; Thomas G. Markees; Leonard D. Shultz; John P. Mordes; Raymond M. Welsh; Aldo A. Rossini; Dale L. Greiner

Costimulation blockade protocols are effective in prolonging allograft survival in animal models and are entering clinical trials, but how environmental perturbants affect graft survival remains largely unstudied. We used a costimulation blockade protocol consisting of a donor-specific transfusion and anti-CD154 mAb to address this question. We observed that lymphocytic choriomeningitis virus infection at the time of donor-specific transfusion and anti-CD154 mAb shortens allograft survival. Lymphocytic choriomeningitis virus 1) activates innate immunity, 2) induces allo-cross-reactive T cells, and 3) generates virus-specific responses, all of which may adversely affect allograft survival. To investigate the role of innate immunity, mice given costimulation blockade and skin allografts were coinjected with TLR2 (Pam3Cys), TLR3 (polyinosinic:polycytidylic acid), TLR4 (LPS), or TLR9 (CpG) agonists. Costimulation blockade prolonged skin allograft survival that was shortened after coinjection by TLR agonists. To investigate underlying mechanisms, we used “synchimeric” mice which circulate trace populations of anti-H2b transgenic alloreactive CD8+ T cells. In synchimeric mice treated with costimulation blockade, coadministration of all four TLR agonists prevented deletion of alloreactive CD8+ T cells and shortened skin allograft survival. These alloreactive CD8+ T cells 1) expressed the proliferation marker Ki-67, 2) up-regulated CD44, and 3) failed to undergo apoptosis. B6.TNFR2−/− and B6.IL-12R−/− mice treated with costimulation blockade plus LPS also exhibited short skin allograft survival whereas similarly treated B6.CD8α−/− and TLR4−/− mice exhibited prolonged allograft survival. We conclude that TLR signaling abrogates the effects of costimulation blockade by preventing alloreactive CD8+ T cell apoptosis through a mechanism not dependent on TNFR2 or IL-12R signaling.


Journal of Immunology | 2001

Skin Allograft Maintenance in a New Synchimeric Model System of Tolerance

Neal N. Iwakoshi; Thomas G. Markees; Nicole A. Turgeon; Thomas B. Thornley; Amy Cuthbert; Jean Leif; Nancy E. Phillips; John P. Mordes; Dale L. Greiner; Aldo A. Rossini

Treatment of mice with a single donor-specific transfusion plus a brief course of anti-CD154 mAb uniformly induces donor-specific transplantation tolerance characterized by the deletion of alloreactive CD8+ T cells. Survival of islet allografts in treated mice is permanent, but skin grafts eventually fail unless recipients are thymectomized. To analyze the mechanisms underlying tolerance induction, maintenance, and failure in euthymic mice we created a new analytical system based on allo-TCR-transgenic hemopoietic chimeric graft recipients. Chimeras were CBA (H-2k) mice engrafted with small numbers of syngeneic TCR-transgenic KB5 bone marrow cells. These mice subsequently circulated a self-renewing trace population of anti-H-2b-alloreactive CD8+ T cells maturing in a normal microenvironment. With this system, we studied the maintenance of H-2b allografts in tolerized mice. We documented that alloreactive CD8+ T cells deleted during tolerance induction slowly returned toward pretreatment levels. Skin allograft rejection in this system occurred in the context of 1) increasing numbers of alloreactive CD8+ cells; 2) a decline in anti-CD154 mAb concentration to levels too low to inhibit costimulatory functions; and 3) activation of the alloreactive CD8+ T cells during graft rejection following deliberate depletion of regulatory CD4+ T cells. Rejection of healed-in allografts in tolerized mice appears to be a dynamic process dependent on the level of residual costimulation blockade, CD4+ regulatory cells, and activated alloreactive CD8+ thymic emigrants that have repopulated the periphery after tolerization.


Journal of Immunology | 2007

Type 1 IFN Mediates Cross-Talk between Innate and Adaptive Immunity That Abrogates Transplantation Tolerance

Thomas B. Thornley; Nancy E. Phillips; Britte C. Beaudette-Zlatanova; Thomas G. Markees; Kapil Bahl; Michael A. Brehm; Leonard D. Shultz; Evelyn A. Kurt-Jones; John P. Mordes; Raymond M. Welsh; Aldo A. Rossini; Dale L. Greiner

TLR activation of innate immunity prevents the induction of transplantation tolerance and shortens skin allograft survival in mice treated with costimulation blockade. The mechanism by which TLR signaling mediates this effect has not been clear. We now report that administration of the TLR agonists LPS (TLR4) or polyinosinic:polycytidylic acid (TLR3) to mice treated with costimulation blockade prevents alloreactive CD8+ T cell deletion, primes alloreactive CTLs, and shortens allograft survival. The TLR4- and MyD88-dependent pathways are required for LPS to shorten allograft survival, whereas polyinosinic:polycytidylic acid mediates its effects through a TLR3-independent pathway. These effects are all mediated by signaling through the type 1 IFN (IFN-αβ) receptor. Administration of IFN-β recapitulates the detrimental effects of TLR agonists on transplantation tolerance. We conclude that the type 1 IFN generated as part of an innate immune response to TLR activation can in turn activate adaptive immune responses that abrogate transplantation tolerance. Blocking of type 1 IFN-dependent pathways in patients may improve allograft survival in the presence of exogenous TLR ligands.


Journal of Clinical Investigation | 2012

Allograft rejection is restrained by short-lived TIM-3 + PD-1 + Foxp3 + Tregs

Shipra Gupta; Thomas B. Thornley; Wenda Gao; Rafael A. Larocca; Laurence A. Turka; Vijay K. Kuchroo; Terry B. Strom

Tregs play a pivotal role in inducing and maintaining donor-specific transplant tolerance. The T cell immunoglobulin and mucin domain-3 protein (TIM-3) is expressed on many fully activated effector T cells. Along with program death 1 (PD-1), TIM-3 is used as a marker for exhausted effector T cells, and interaction with its ligand, galectin-9, leads to selective death of TIM-3+ cells. We report herein the presence of a galectin-9-sensitive CD4+FoxP3+TIM-3+ population of T cells, which arose from CD4+FoxP3+TIM-3- proliferating T cells in vitro and in vivo and were often PD-1+. These cells became very prominent among graft-infiltrating Tregs during allograft response. The frequency and number of TIM-3+ Tregs peaked at the time of graft rejection and declined thereafter. Moreover, these cells also arise in a tolerance-promoting donor-specific transfusion model, representing a pool of proliferating, donor-specific Tregs. Compared with TIM-3- Tregs, TIM-3+ Tregs, which are often PD-1+ as well, exhibited higher in vitro effector function and more robust expression of CD25, CD39, CD73, CTLA-4, IL-10, and TGF-β but not galectin-9. However, these TIM-3+ Tregs did not flourish when passively transferred to newly transplanted hosts. These data suggest that a heretofore unrecognized graft-infiltrating, short-lived subset of Tregs can restrain rejection.


European Journal of Immunology | 2014

Leptin deficiency impairs maturation of dendritic cells and enhances induction of regulatory T and Th17 cells

Pedro M. Moraes-Vieira; Rafael A. Larocca; Ênio José Bassi; Jean Pierre Schatzmann Peron; Vinicius Andrade-Oliveira; Frederick Wasinski; Ronaldo C. Araujo; Thomas B. Thornley; Francisco J. Quintana; Alexandre S. Basso; Terry B. Strom; Niels Olsen Saraiva Câmara

Leptin is an adipose‐secreted hormone that plays an important role in both metabolism and immunity. Leptin has been shown to induce Th1‐cell polarization and inhibit Th2‐cell responses. Additionally, leptin induces Th17‐cell responses, inhibits regulatory T (Treg) cells and modulates autoimmune diseases. Here, we investigated whether leptin mediates its activity on T cells by influencing dendritic cells (DCs) to promote Th17 and Treg‐cell immune responses in mice. We observed that leptin deficiency (i) reduced the expression of DC maturation markers, (ii) decreased DC production of IL‐12, TNF‐α, and IL‐6, (iii) increased DC production of TGF‐β, and (iv) limited the capacity of DCs to induce syngeneic CD4+ T‐cell proliferation. As a consequence of this unique phenotype, DCs generated under leptin‐free conditions induced Treg or TH17 cells more efficiently than DCs generated in the presence of leptin. These data indicate important roles for leptin in DC homeostasis and the initiation and maintenance of inflammatory and regulatory immune responses by DCs.


PLOS ONE | 2013

Adipose tissue-derived mesenchymal stem cells increase skin allograft survival and inhibit Th-17 immune response.

Rafael A. Larocca; Pedro M. Moraes-Vieira; Ênio José Bassi; Patricia Semedo; Danilo Candido de Almeida; Marina Burgos da Silva; Thomas B. Thornley; Alvaro Pacheco-Silva; Niels Olsen Saraiva Câmara

Adipose tissue-derived mesenchymal stem cells (ADSC) exhibit immunosuppressive capabilities both in vitro and in vivo. Their use for therapy in the transplant field is attractive as they could render the use of immunosuppressive drugs unnecessary. The aim of this study was to investigate the effect of ADSC therapy on prolonging skin allograft survival. Animals that were treated with a single injection of donor allogeneic ADSC one day after transplantation showed an increase in donor skin graft survival by approximately one week. This improvement was associated with preserved histological morphology, an expansion of CD4+ regulatory T cells (Treg) in draining lymph nodes, as well as heightened IL-10 expression and down-regulated IL-17 expression. In vitro, ADSC inhibit naïve CD4+ T cell proliferation and constrain Th-1 and Th-17 polarization. In summary, infusion of ADSC one day post-transplantation dramatically increases skin allograft survival by inhibiting the Th-17 pathogenic immune response and enhancing the protective Treg immune response. Finally, these data suggest that ADSC therapy will open new opportunities for promoting drug-free allograft survival in clinical transplantation.


American Journal of Transplantation | 2010

Allografts Stimulate Cross-Reactive Virus-Specific Memory CD8 T Cells with Private Specificity

Michael A. Brehm; Keith A. Daniels; Bhavana Priyadharshini; Thomas B. Thornley; Dale L. Greiner; Aldo A. Rossini; Raymond M. Welsh

Viral infections have been associated with the rejection of transplanted allografts in humans and mice, and the induction of tolerance to allogeneic tissues in mice is abrogated by an ongoing viral infection and inhibited in virus‐immune mice. One proposed mechanism for this ‘heterologous immunity’ is the induction of alloreactive T cell responses that cross‐react with virus‐derived antigens. These cross‐reactive CD8 T cells are generated during acute viral infection and survive into memory, but their ability to partake in the immune response to allografts in vivo is not known. We show here that cross‐reactive, virus‐specific memory CD8 T cells from mice infected with LCMV proliferated in response to allografts. CD8 T cells specific to several LCMV epitopes proliferated in response to alloantigens, with the magnitude and hierarchy of epitope‐specific responses varying with the private specificities of the host memory T cell repertoire, as shown by adoptive transfer studies. Last, we show that purified LCMV‐specific CD8 T cells rejected skin allografts in SCID mice. These findings therefore implicate a potential role for heterologous immunity in virus‐induced allograft rejection.


Journal of Clinical Investigation | 2014

Fragile TIM-4–expressing tissue resident macrophages are migratory and immunoregulatory

Thomas B. Thornley; Zemin Fang; Savithri Balasubramanian; Rafael A. Larocca; Weihua Gong; Shipra Gupta; Eva Csizmadia; Nicolas Degauque; Beom Seok Kim; Maria Koulmanda; Vijay K. Kuchroo; Terry B. Strom

Macrophages characterized as M2 and M2-like regulate immune responses associated with immune suppression and healing; however, the relationship of this macrophage subset to CD169+ tissue-resident macrophages and their contribution to shaping alloimmune responses is unknown. Here we identified a population of M2-like tissue-resident macrophages that express high levels of the phosphatidylserine receptor TIM-4 and CD169 (TIM-4hiCD169+). Labeling and tracking of TIM-4hiCD169+ macrophages in mice revealed that this population is a major subset of tissue-resident macrophages, homes to draining LNs following oxidative stress, exhibits an immunoregulatory and hypostimulatory phenotype that is maintained after migration to secondary lymphoid organs, favors preferential induction of antigen-stimulated Tregs, and is highly susceptible to apoptosis. Moreover, CD169+ tissue-resident macrophages were resistant to oxidative stress-induced apoptosis in mice lacking TIM-4. Compared with heart allografts from WT mice, Tim4-/- heart allografts survived much longer and were more easily tolerized by non-immunosuppressed recipients. Furthermore, Tim4-/- allograft survival was associated with the infiltration of Tregs into the graft. Together, our data provide evidence that M2-like TIM-4hiCD169+ tissue-resident macrophages are immunoregulatory and promote engraftment of cardiac allografts, but their influence is diminished by TIM-4-dependent programmed cell death.


Cell Stress & Chaperones | 2008

Protein kinase C signaling during T cell activation induces the endoplasmic reticulum stress response

Steven C. Pino; Bryan O’Sullivan-Murphy; Erich A. Lidstone; Thomas B. Thornley; Agata Jurczyk; Fumihiko Urano; Dale L. Greiner; John P. Mordes; Aldo A. Rossini; Rita Bortell

T cell receptor (TCR) ligation (signal one) in the presence of co-stimulation (signal two) results in downstream signals that increase protein production enabling naïve T cells to fully activate and gain effector function. Enhanced production of proteins by a cell requires an increase in endoplasmic reticulum (ER) chaperone expression, which is accomplished through activation of a cellular mechanism known as the ER stress response. The ER stress response is initiated during the cascade of events that occur for the activation of many cells; however, this process has not been comprehensively studied for T cell function. In this study, we used primary T cells and mice circulating TCR transgenic CD8+ T cells to investigate ER chaperone expression in which TCR signaling was initiated in the presence or absence of co-stimulation. In the presence of both signals, in vitro and in vivo analyses demonstrated induction of the ER stress response, as evidenced by elevated expression of GRP78 and other ER chaperones. Unexpectedly, ER chaperones were also increased in T cells exposed only to signal one, a treatment known to cause T cells to enter the ‘nonresponsive’ states of anergy and tolerance. Treatment of T cells with an inhibitor to protein kinase C (PKC), a serine/threonine protein kinase found downstream of TCR signaling, indicated PKC is involved in the induction of the ER stress response during the T cell activation process, thus revealing a previously unknown role for this signaling protein in T cells. Collectively, these data suggest that induction of the ER stress response through PKC signaling is an important component for the preparation of a T cell response to antigen.


Journal of Immunology | 2009

TLR Agonists Prevent the Establishment of Allogeneic Hematopoietic Chimerism in Mice Treated with Costimulation Blockade

David Miller; Thomas B. Thornley; Todd Pearson; Annie J. Kruger; Masahiro Yamazaki; Leonard D. Shultz; Raymond M. Welsh; Michael A. Brehm; Aldo A. Rossini; Dale L. Greiner

Activation of TLR4 by administration of LPS shortens the survival of skin allografts in mice treated with costimulation blockade through a CD8 T cell-dependent, MyD88-dependent, and type I IFN receptor-dependent pathway. The effect of TLR activation on the establishment of allogeneic hematopoietic chimerism in mice treated with costimulation blockade is not known. Using a costimulation blockade protocol based on a donor-specific transfusion (DST) and a short course of anti-CD154 mAb, we show that LPS administration at the time of DST matures host alloantigen-presenting dendritic cells, prevents the establishment of mixed allogeneic hematopoietic chimerism, and shortens survival of donor-specific skin allografts. LPS mediates its effects via a mechanism that involves both CD4+ and CD8+ T cells and results from signaling through either the MyD88 or the type I IFN receptor pathways. We also document that timing of LPS administration is critical, as injection of LPS 24 h before treatment with DST and anti-CD154 mAb does not prevent hematopoietic engraftment but administration the day after bone marrow transplantation does. We conclude that TLR4 activation prevents the induction of mixed allogeneic hematopoietic chimerism through type I IFN receptor and MyD88-dependent signaling, which leads to the up-regulation of costimulatory molecules on host APCs and the generation of alloreactive T cells. These data suggest that distinct but overlapping cellular and molecular mechanisms control the ability of TLR agonists to block tolerance induction to hematopoietic and skin allografts in mice treated with costimulation blockade.

Collaboration


Dive into the Thomas B. Thornley's collaboration.

Top Co-Authors

Avatar

Dale L. Greiner

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Aldo A. Rossini

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Michael A. Brehm

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Terry B. Strom

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Raymond M. Welsh

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Thomas G. Markees

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eva Csizmadia

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

John P. Mordes

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Keith A. Daniels

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge