Network


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

Hotspot


Dive into the research topics where Jason Foley is active.

Publication


Featured researches published by Jason Foley.


Science Translational Medicine | 2011

The PDL1-PD1 axis converts human TH1 cells into regulatory T cells.

Shoba Amarnath; Courtney W. Mangus; James Cm M. Wang; Fang Wei; Alice He; Veena Kapoor; Jason Foley; Paul R. Massey; Tania C. Felizardo; James L. Riley; Bruce L. Levine; Carl H. June; Jeffrey A. Medin; Daniel H. Fowler

The inhibitory ligand PDL1 transforms immune cells from attackers into regulators. PDL1: Restoring the Peace With great power comes great responsibility. In superhero lore, special powers don’t separate the saviors from the evil villains they fight; instead, what matters is how the person behind the mask uses those powers. Immune cells are the superheroes of the body—they fight off infection and patrol the body for cancer. However, sometimes, even protective cells “go bad,” causing autoimmunity or graft-versus-host disease after transplant. Amarnath et al. now show that an inhibitory protein called programmed death ligand 1 (PDL1) can regulate renegade immune cells by converting immune response–promoting T helper type 1 (TH1) cells to regulatory T (Treg) cells—agents that selectively suppress activation of the immune system. TH1 cells secrete proinflammatory cytokines and are critical for the immune response to infection and cancer cells. In contrast to other subsets of TH cells, researchers believed TH1 cells to be relatively stable. However, PDL1 caused human TH1 cells to convert to Treg cells both in vitro and in vivo. These TH1-derived Treg cells inhibited graft-versus-host disease in mice after transplant. Moreover, inhibiting Treg differentiation by blocking the PDL1 receptor PD1 or pharmacologically inhibiting SHP1 and SHP2, which are signaling molecules that act downstream of PD1 activation, restored graft-versus-host disease in mice. These data provide the basis for future therapies: Because PDL1 is highly expressed on many cancers, inhibiting this pathway may restore T cell–mediated cancer surveillance; alternately, accentuating signaling through this pathway may prevent autoimmunity or graft-versus-host disease. With this knowledge, scientists and doctors may be able to ensure that T cells are the superheroes they are meant to be. Immune surveillance by T helper type 1 (TH1) cells is not only critical for the host response to tumors and infection, but also contributes to autoimmunity and graft-versus-host disease (GVHD) after transplantation. The inhibitory molecule programmed death ligand 1 (PDL1) has been shown to anergize human TH1 cells, but other mechanisms of PDL1-mediated TH1 inhibition such as the conversion of TH1 cells to a regulatory phenotype have not been well characterized. We hypothesized that PDL1 may cause TH1 cells to manifest differentiation plasticity. Conventional T cells or irradiated K562 myeloid tumor cells overexpressing PDL1 converted TBET+ TH1 cells into FOXP3+ regulatory T (Treg) cells in vivo, thereby preventing human-into-mouse xenogeneic GVHD (xGVHD). Either blocking PD1 expression on TH1 cells by small interfering RNA targeting or abrogation of PD1 signaling by SHP1/2 pharmacologic inhibition stabilized TH1 cell differentiation during PDL1 challenge and restored the capacity of TH1 cells to mediate lethal xGVHD. PD1 signaling therefore induces human TH1 cells to manifest in vivo plasticity, resulting in a Treg phenotype that severely impairs cell-mediated immunity. Converting human TH1 cells to a regulatory phenotype with PD1 signaling provides a potential way to block GVHD after transplantation. Moreover, because this conversion can be prevented by blocking PD1 expression or pharmacologically inhibiting SHP1/2, this pathway provides a new therapeutic direction for enhancing T cell immunity to cancer and infection.


Journal of Clinical Oncology | 2008

Association of Serum Interleukin-7 Levels With the Development of Acute Graft-Versus-Host Disease

Robert Dean; Terry J. Fry; Crystal L. Mackall; Seth M. Steinberg; Fran Hakim; Daniel H. Fowler; Jeanne Odom; Jason Foley; Ronald E. Gress; Michael R. Bishop

PURPOSE Morbidity from acute graft-versus-host disease (GVHD) limits the success of allogeneic hematopoietic stem-cell transplantation (HSCT) to treat malignancy. Interleukin-7 (IL-7), the principal homeostatic cytokine for T cells, is required for acute GVHD in murine models. In contrast to inflammatory cytokines (eg, IL-2, tumor necrosis factor alpha), IL-7 has not been studied extensively in the clinical transplant setting relative to its relationship with acute GVHD. PATIENTS AND METHODS We evaluated the association of serum IL-7 levels with acute GVHD in 31 patients who were uniformly treated in a prospective clinical trial with reduced-intensity allogeneic HSCT from human leukocyte antigen-identical siblings. GVHD prophylaxis consisted of cyclosporine and methotrexate. Serum IL-7 levels and lymphocyte populations were determined at enrollment, the day of transplantation before the allograft infusion, and at specified intervals through 12 months post-transplantation. RESULTS As expected, IL-7 levels were inversely correlated with T-cell populations (P < .00001). Acute GVHD was significantly associated with higher IL-7 levels at day +7 (P = .01) and day +14 (P = .00003) post-transplantation as well as with the allograft CD34(+) cell dose (P = .01). IL-7 levels at day +14 also correlated with the severity of acute GVHD (P < .0001). In logistic regression models, these factors were highly sensitive (up to 86%) and specific (100%) for classifying whether patients developed acute GVHD. CONCLUSION These data support preclinical observations that IL-7 plays a critical role in inducing acute GVHD and provide a rational basis for novel approaches to prevent and treat acute GVHD through modulation of the IL-7 pathway.


Journal of Immunology | 2005

Ex Vivo Rapamycin Generates Donor Th2 Cells That Potently Inhibit Graft-versus-Host Disease and Graft-versus-Tumor Effects via an IL-4-Dependent Mechanism

Jason Foley; Unsu Jung; Angel Miera; Todd Borenstein; Jacopo Mariotti; Michael Eckhaus; Barbara E. Bierer; Daniel H. Fowler

Rapamycin (sirolimus) inhibits graft-vs-host disease (GVHD) and polarizes T cells toward Th2 cytokine secretion after allogeneic bone marrow transplantation (BMT). Therefore, we reasoned that ex vivo rapamycin might enhance the generation of donor Th2 cells capable of preventing GVHD after fully MHC-disparate murine BMT. Using anti-CD3 and anti-CD28 costimulation, CD4+ Th2 cell expansion was preserved partially in high-dose rapamycin (10 μM; Th2.rapa cells). Th2.rapa cells secreted IL-4 yet had reduced IL-5, IL-10, and IL-13 secretion relative to control Th2 cells. BMT cohorts receiving wild-type (WT) Th2.rapa cells, but not Th2.rapa cells generated from IL-4-deficient (knockout) donors, had marked Th2 skewing post-BMT and greatly reduced donor anti-host T cell alloreactivity. Histologic studies demonstrated that Th2.rapa cell recipients had near complete abrogation of skin, liver, and gut GVHD. Overall survival in recipients of WT Th2.rapa cells, but not IL-4 knockout Th2.rapa cells, was constrained due to marked attenuation of an allogeneic graft-vs-tumor (GVT) effect against host-type breast cancer cells. Delay in Th2.rapa cell administration until day 4, 7, or 14 post-BMT enhanced GVT effects, moderated GVHD, and improved overall survival. Therefore, ex vivo rapamycin generates enhanced donor Th2 cells for attempts to balance GVHD and GVT effects.


PLOS ONE | 2009

A central role for Foxp3+ regulatory T cells in K-Ras-driven lung tumorigenesis

Courtney A. Granville; Regan M. Memmott; Andria Balogh; Jacopo Mariotti; Shigeru Kawabata; Wei Han; Jaclyn LoPiccolo; Jason Foley; David J. Liewehr; Seth M. Steinberg; Daniel H. Fowler; M. Christine Hollander; Phillip A. Dennis

Background K-Ras mutations are characteristic of human lung adenocarcinomas and occur almost exclusively in smokers. In preclinical models, K-Ras mutations are necessary for tobacco carcinogen-driven lung tumorigenesis and are sufficient to cause lung adenocarcinomas in transgenic mice. Because these mutations confer resistance to commonly used cytotoxic chemotherapies and targeted agents, effective therapies that target K-Ras are needed. Inhibitors of mTOR such as rapamycin can prevent K-Ras-driven lung tumorigenesis and alter the proportion of cytotoxic and Foxp3+ regulatory T cells, suggesting that lung-associated T cells might be important for tumorigenesis. Methods Lung tumorigenesis was studied in three murine models that depend on mutant K-Ras; a tobacco carcinogen-driven model, a syngeneic inoculation model, and a transgenic model. Splenic and lung-associated T cells were studied using flow cytometry and immunohistochemistry. Foxp3+ cells were depleted using rapamycin, an antibody, or genetic ablation. Results Exposure of A/J mice to a tobacco carcinogen tripled lung-associated Foxp3+ cells prior to tumor development. At clinically relevant concentrations, rapamycin prevented this induction and reduced lung tumors by 90%. In A/J mice inoculated with lung adenocarcinoma cells resistant to rapamycin, antibody-mediated depletion of Foxp3+ cells reduced lung tumorigenesis by 80%. Likewise, mutant K-Ras transgenic mice lacking Foxp3+ cells developed 75% fewer lung tumors than littermates with Foxp3+ cells. Conclusions Foxp3+ regulatory T cells are required for K-Ras-mediated lung tumorigenesis in mice. These studies support clinical testing of rapamycin or other agents that target Treg in K-Ras driven human lung cancer.


Stem Cells | 2015

Bone marrow-derived mesenchymal stromal cells harness purinergenic signaling to tolerize human Th1 cells in vivo.

Shoba Amarnath; Jason Foley; Don E. Farthing; Ronald E. Gress; Arian Laurence; Michael A. Eckhaus; Jean-Yves Métais; Jeremy J. Rose; Frances T. Hakim; Tania C. Felizardo; Austin V. Cheng; Pamela Gehron Robey; David E. Stroncek; Marianna Sabatino; Minoo Battiwalla; Sawa Ito; Daniel H. Fowler; A.J. Barrett

The use of bone marrow‐derived mesenchymal stromal cells (BMSC) in the treatment of alloimmune and autoimmune conditions has generated much interest, yet an understanding of the therapeutic mechanism remains elusive. We therefore explored immune modulation by a clinical‐grade BMSC product in a model of human‐into‐mouse xenogeneic graft‐versus‐host disease (x‐GVHD) mediated by human CD4+ Th1 cells. BMSC reversed established, lethal x‐GVHD through marked inhibition of Th1 cell effector function. Gene marking studies indicated BMSC engraftment was limited to the lung; furthermore, there was no increase in regulatory T cells, thereby suggesting a paracrine mechanism of BMSC action. BMSC recipients had increased serum CD73 expressing exosomes that promoted adenosine accumulation ex vivo. Importantly, immune modulation mediated by BMSC was fully abrogated by pharmacologic therapy with an adenosine A2A receptor antagonist. To investigate the potential clinical relevance of these mechanistic findings, patient serum samples collected pre‐ and post‐BMSC treatment were studied for exosome content: CD73 expressing exosomes promoting adenosine accumulation were detected in post‐BMSC samples. In conclusion, BMSC effectively modulate experimental GVHD through a paracrine mechanism that promotes adenosine‐based immune suppression. Stem Cells 2015;33:1200–1212 Stem Cells 2015;33:1200–1212


Biology of Blood and Marrow Transplantation | 2008

Th2 cell therapy of established acute graft-versus-host disease requires IL-4 and IL-10 and is abrogated by IL-2 or host-type antigen-presenting cells.

Jason Foley; Jacopo Mariotti; Kaitlyn Ryan; Michael A. Eckhaus; Daniel H. Fowler

Delayed donor Th2 cell infusion permits a graft-versus-tumor (GVT) effect to occur with subsequent amelioration of established graft-versus-host disease (GVHD). Relative to GVHD controls (B6-into-BALB/c model), recipients of delayed Th2 cells (day 14 post-BMT) had increased survival (3/3 experiments [exp]; each exp P < .0001) and reduced GVHD by histology analysis 5 days post-Th2 infusion without increased tumor burden (3 of 3 exp; each exp P < or = .02). Th2 cell-mediated amelioration of GVHD was associated with greatly reduced allospecific IFN-gamma secretion, in vivo augmentation of allospecific IL-4 and IL-10 secretion, and reduction in donor CD8(+) T cell number post-BMT (3 of 3 exp; each comparison, P < or = .003). To better understand the molecular mechanism of this GVHD therapy, Th2 cells were generated from wild-type (WT), IL-4 deficient (KO), or IL-10 KO donors: remarkably, recipients of IL-4 or IL-10 KO Th2 cells had no survival advantage, no improvement in GVHD by histology, no reduction in CD8(+) T cell expansion post-BMT, and no in vivo shift toward type II cytokines. We reasoned that IL-2 and alloantigen availability may be limiting factors for Th2 cell therapy, and as such, evaluated whether coadministration of IL-2 or coinfusion of host-type antigen-presenting cells (APC) might intensify the anti-GVHD effect. However, contrary to these hypotheses, concomitant IL-2 therapy or APC administration fully abrogated the Th2 cell-mediated survival advantage and histology-defined GVHD reduction, reduced Th2 cell expansion in vivo while promoting CD8(+) T cell expansion from cells originating from the initial allograft, and impaired type II polarization in vivo. In conclusion, Th2 cell therapy can rapidly ameliorate severe GVHD via IL-4 and IL-10 mediated mechanisms, and potentially, via IL-2 consumption and APC modulation mechanisms.


Clinical Gastroenterology and Hepatology | 2004

Clinical “cytokine storm” as revealed by monocyte intracellular flow cytometry: correlation of tumor necrosis factor α with severe gut graft-versus-host disease

Daniel H. Fowler; Jason Foley; Jeannie Whit Shan Hou; Jeanne Odom; Kate Castro; Seth M. Steinberg; Juan Gea-Banacloche; Claude Kasten-Sportes; Ronald E. Gress; Michael R. Bishop

BACKGROUND & AIMS Gut graft-versus-host disease (GVHD) contributes significantly to lethality after allogeneic hematopoietic stem-cell transplantation (HSCT). In murine models, macrophage secretion of interleukin 1alpha (IL-1alpha) and tumor necrosis factor alpha (TNF-alpha) contributes to gut GVHD pathogenesis. To help characterize whether human gut GVHD has similar biological characteristics, monocyte IL-1alpha and TNF-alpha production were evaluated after HSCT. METHODS Patients with refractory hematologic malignancy (n = 17) underwent reduced-intensity conditioning, HLA-matched sibling HSCT, and cyclosporine A GVHD prophylaxis. After HSCT, monocyte IL-1alpha and TNF-alpha levels were measured using intracellular flow cytometry (IC-FCM), and results were correlated with clinical GVHD. RESULTS Incidences of acute GVHD were none (n = 3), grades I-II (n = 9), or grades III-IV (n = 5; each case with stage 2-3 gut GVHD). Posttransplantation monocyte IL-1alpha production (percentage of CD14(+)IL-1(+) cells) increased significantly from 8.7% +/- 3.7% (week 2) to 40.3% +/- 7.3% (week 4; P = 0.0065) and was not associated with GVHD severity (P = 1.00). Conversely, increases in monocyte TNF-alpha were quantitatively reduced and temporally delayed, from 0.6% +/- 0.2% (week 2) to 3.6% +/- 1.4% (week 6; P = 0.076). Most importantly, elevation of monocyte TNF-alpha level correlated with increased gut GVHD severity (P = 0.0041); increases in monocyte TNF-alpha levels typically preceded the onset of gut GVHD symptoms. CONCLUSIONS Human gut GVHD after reduced-intensity allogeneic HSCT is associated with monocyte cytokine secretion initially involving IL-1alpha, followed by TNF-alpha. Serial measurement of monocyte cytokines, in particular, TNF-alpha, by IC-FCM may represent a noninvasive method for GVHD monitoring, potentially allowing the identification of patients appropriate for early-intervention strategies.


Blood | 2008

Graft rejection as a Th1-type process amenable to regulation by donor Th2-type cells through an interleukin-4/STAT6 pathway.

Jacopo Mariotti; Jason Foley; Kaitlyn Ryan; Nicole Buxhoeveden; Veena Kapoor; Shoba Amarnath; Daniel H. Fowler

Graft rejection has been defined as the mirror image of graft-versus-host disease, which is biologically characterized primarily as a Th1-type process. As such, we reasoned that graft rejection would represent a Th1 response amenable to Th2 modulation. Indeed, adoptive transfer of host Th1-type cells mediated rejection of fully MHC-disparate murine bone marrow allografts more effectively than host Th2-type cells. Furthermore, STAT1-deficient host T cells did not differentiate into Th1-type cells in vivo and failed to mediate rejection. We next hypothesized that donor Th2 cell allograft augmentation would prevent rejection by modulation of the host Th1/Th2 balance. In the setting of donor Th2 cell therapy, host-anti-donor allospecific T cells acquired Th2 polarity, persisted posttransplantation, and did not mediate rejection. Abrogation of rejection required donor Th2 cell IL-4 secretion and host T-cell STAT6 signaling. In conclusion, T cell-mediated marrow graft rejection primarily resembles a Th1-type process that can be abrogated by donor Th2 cell therapy that promotes engraftment through a novel mechanism whereby cytokine polarization is transferred to host T cells.


Journal of Immunology | 2008

Ex Vivo Rapamycin Generates Apoptosis-Resistant Donor Th2 Cells That Persist In Vivo and Prevent Hemopoietic Stem Cell Graft Rejection

Jacopo Mariotti; Jason Foley; Unsu Jung; Todd Borenstein; Nermina Kantardzic; Soo Han; Joshua T. Hanson; Elaine Wong; Nicole Buxhoeveden; Jane B. Trepel; Antonio Tito Fojo; William G. Telford; Daniel H. Fowler

Because ex vivo rapamycin generates murine Th2 cells that prevent Graft-versus-host disease more potently than control Th2 cells, we hypothesized that rapamycin would generate Th2/Tc2 cells (Th2/Tc2.R cells) that abrogate fully MHC-disparate hemopoietic stem cell rejection more effectively than control Th2/Tc2 cells. In a B6-into-BALB/c graft rejection model, donor Th2/Tc2.R cells were indeed enriched in their capacity to prevent rejection; importantly, highly purified CD4+ Th2.R cells were also highly efficacious for preventing rejection. Rapamycin-generated Th2/Tc2 cells were less likely to die after adoptive transfer, accumulated in vivo at advanced proliferative cycles, and were present in 10-fold higher numbers than control Th2/Tc2 cells. Th2.R cells had a multifaceted, apoptosis-resistant phenotype, including: 1) reduced apoptosis after staurosporine addition, serum starvation, or CD3/CD28 costimulation; 2) reduced activation of caspases 3 and 9; and 3) increased anti-apoptotic Bcl-xL expression and reduced proapoptotic Bim and Bid expression. Using host-versus-graft reactivity as an immune correlate of graft rejection, we found that the in vivo efficacy of Th2/Tc2.R cells 1) did not require Th2/Tc2.R cell expression of IL-4, IL-10, perforin, or Fas ligand; 2) could not be reversed by IL-2, IL-7, or IL-15 posttransplant therapy; and 3) was intact after therapy with Th2.R cells relatively devoid of Foxp3 expression. We conclude that ex vivo rapamycin generates Th2 cells that are resistant to apoptosis, persist in vivo, and effectively prevent rejection by a mechanism that may be distinct from previously described graft-facilitating T cells.


Autophagy | 2010

Rapamycin generates anti-apoptotic human Th1/Tc1 cells via autophagy for induction of xenogeneic GVHD.

Shoba Amarnath; Francis A. Flomerfelt; Carliann M. Costanzo; Jason Foley; Jacopo Mariotti; Daniel M. Konecki; Anu Gangopadhyay; Michael Eckhaus; Susan Wong; Bruce L. Levine; Carl H. June; Daniel H. Fowler

Murine T cells exposed to rapamycin maintain flexibility towards Th1/Tc1 differentiation, thereby indicating that rapamycin promotion of regulatory T cells (Tregs) is conditional. The degree to which rapamycin might inhibit human Th1/Tc1 differentiation has not been evaluated. In the presence of rapamycin, T cell costimulation and polarization with IL-12 or IFN-α permitted human CD4+ and CD8+ T cell differentiation towards a Th1/Tc1 phenotype; activation of STAT1 and STAT4 pathways essential for Th1/Tc1 polarity was preserved during mTOR blockade but instead abrogated by PI3 kinase inhibition. Such rapamycin-resistant human Th1/Tc1 cells: (1) were generated through autophagy (increased LC3BII expression; phenotype reversion by autophagy inhibition via 3-MA or siRNA for Beclin1); (2) expressed anti-apoptotic bcl-2 family members (reduced Bax, Bak; increased phospho-Bad); (3) maintained mitochondrial membrane potentials; and (4) displayed reduced apoptosis. In vivo, type I polarized and rapamycin-resistant human T cells caused increased xenogeneic graft-versus-host disease (x-GVHD). Murine recipients of rapamycin-resistant human Th1/Tc1 cells had: (1) persistent T cell engraftment; (2) increased T cell cytokine and cytolytic effector function; and (3) T cell infiltration of skin, gut, and liver. Rapamycin therefore does not impair human T cell capacity for type I differentiation. Rather, rapamycin yields an anti-apoptotic Th1/Tc1 effector phenotype by promoting autophagy.

Collaboration


Dive into the Jason Foley's collaboration.

Top Co-Authors

Avatar

Daniel H. Fowler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jacopo Mariotti

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shoba Amarnath

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Unsu Jung

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Todd Borenstein

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kaitlyn Ryan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael A. Eckhaus

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nicole Buxhoeveden

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

D.H. Fowler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tania C. Felizardo

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge