Network


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

Hotspot


Dive into the research topics where Benjamin M. Matta is active.

Publication


Featured researches published by Benjamin M. Matta.


Journal of Immunology | 2006

CD11b+/Gr-1+ Myeloid Suppressor Cells Cause T Cell Dysfunction after Traumatic Stress

Valeriya P. Makarenkova; Vishal Bansal; Benjamin M. Matta; Lori Perez; Juan B. Ochoa

T cell dysfunction that occurs after surgery or trauma is associated with a poor clinical outcome. We describe that myeloid suppressor cells expressing CD11b+/Gr-1+ markers invade the spleen after traumatic stress and suppress T cell function through the production of arginase 1. We created a consistent model of traumatic stress in C57BL/6 mice to perform this work. A significant number of CD11b+/Gr-1+ cells expressing arginase 1 accumulated in T cell zones around the germinal centers of the white pulp of the spleen within 6 h of trauma and lasted for at least 72 h. Increased arginase activity and arginase 1 expression, along with increased [3H]arginine uptake, l-arginine depletion, and l-ornithine accumulation in the culture medium, were observed exclusively in CD11b+/Gr-1+ cells after traumatic stress. Flow cytometry revealed CD11b+/Gr-1+ as a heterogeneous myeloid suppressor cell also expressing low levels of MHC class I and II, CD80, CD86, CD31, and others. When compared with controls, trauma-induced CD11b+/Gr-1+ cells significantly inhibited CD3/CD28-mediated T cell proliferation, TCR ζ-chain expression, and IL-2 production. The suppressive effects by trauma CD11b+/Gr-1+ cells were overcome with the arginase antagonist N-hydroxy-nor-l-arginine or extrasupplementation of medium with l-arginine. Poor Ag-presenting capacity of control and trauma-induced CD11b+/Gr-1+ cells was detected in allogeneic murine leukocyte reaction. This study demonstrates that CD11b+/Gr-1+ cells invade the spleen following traumatic stress and cause T cell dysfunction by an arginase-mediated mechanism, probably that of arginine depletion. Understanding the mechanism of immune suppression by these cells has important clinical implications in the treatment of immune dysfunction after trauma or surgery.


European Journal of Immunology | 2010

Tolerogenic plasmacytoid DC

Benjamin M. Matta; Antonino Castellaneta; Angus W. Thomson

Plasmacytoid DC (pDC) are type‐I IFN‐producing cells known for their capacity to promote anti‐viral innate and adaptive immune responses. Despite their potent anti‐viral function, when compared with conventional DC, pDC exhibit poor immunostimulatory ability and their interaction with T cells often favors the generation of Treg. pDC are activated primarily in response to ssRNA and ssDNA through TLR7 and TLR9, respectively, but also through TLR‐independent mechanisms. Non‐lymphoid tissue pDC, such as those residing in the airways, gut, and liver, play a significant role in regulating mucosal immunity and are critical for the development of tolerance to inhaled or ingested antigens. Herein we discuss properties that define tolerogenic pDC and how their unique characteristics translate into an ability to regulate immunity and promote the development of tolerance. We cover the importance of pDC during intrathymic Treg development and the maintenance of peripheral tolerance, as well as their regulatory role in transplantation, autoimmunity, and cancer. We highlight recent findings regarding danger‐associated molecular pattern and PAMP signaling in the regulation of pDC function, and how the ability of pDC to promote tolerance translates into the potential clinical applications of these cells as therapeutic targets to regulate immune reactivity.


Journal of Immunology | 2014

IL-33 Is an Unconventional Alarmin That Stimulates IL-2 Secretion by Dendritic Cells To Selectively Expand IL-33R/ST2+ Regulatory T Cells

Benjamin M. Matta; Jeremy M. Lott; Lisa Mathews; Quan Liu; Brian R. Rosborough; Bruce R. Blazar; Hēth R. Turnquist

IL-33 is a recently characterized IL-1 family member that is proposed to function as an alarmin, or endogenous signal of cellular damage, as well as act as a pleiotropic cytokine. The ability of IL-33 to potentiate both Th1 and Th2 immunity supports its role in pathogen clearance and disease immunopathology. Yet, IL-33 restrains experimental colitis and transplant rejection by expanding regulatory T cells (Treg) via an undefined mechanism. We sought to determine the influence of IL-33 on hematopoietic cells that drives Treg expansion and underlies the therapeutic benefit of IL-33 administration. In this study, we identify a feedback loop in which conventional mouse CD11c+ dendritic cells (DC) stimulated by IL-33 secrete IL-2 to selectively expand IL-33R(ST2+)– suppressive CD4+Foxp3+ Treg. Interestingly, this occurs in the absence of classical DC maturation, and DC-derived (innate) IL-2 increases ST2 expression on both DC and interacting Treg. ST2+ Treg represent an activated subset of Foxp3+ cells, demonstrated to be ICOShighCD44high compared with their ST2− counterparts. Furthermore, although studies have shown that IL-33–exposed DC promote Th2 responses, we reveal that ST2+ DC are required for IL-33–mediated in vitro and in vivo Treg expansion. Thus, we have uncovered a relationship between IL-33 and innate IL-2 that promotes the selective expansion of ST2+ Treg over non-Treg. These findings identify a novel regulatory pathway driven by IL-33 in immune cells that may be harnessed for therapeutic benefit or for robust expansion of Treg in vitro and in vivo.


Journal of Immunology | 2010

Mammalian Target of Rapamycin Inhibition and Alloantigen-Specific Regulatory T Cells Synergize To Promote Long-Term Graft Survival in Immunocompetent Recipients

Giorgio Raimondi; Tina L. Sumpter; Benjamin M. Matta; Mahesh Pillai; Natasha Corbitt; Yoram Vodovotz; Zhiliang Wang; Angus W. Thomson

Minimization of immunosuppression and donor-specific tolerance to MHC-mismatched organ grafts are important clinical goals. The therapeutic potential of regulatory T cells (Tregs) has been demonstrated, but conditions for optimizing their in vivo function posttransplant in nonlymphocyte-depleted hosts remain undefined. In this study, we address mechanisms through which inhibition of the mammalian target of rapamycin (Rapa) synergizes with alloantigen-specific Treg (AAsTreg) to permit long-term, donor-specific heart graft survival in immunocompetent hosts. Crucially, immature allogeneic dendritic cells allowed AAsTreg selection in vitro, with minimal expansion of unwanted (Th17) cells. The rendered Treg potently inhibited T cell proliferation in an Ag-specific manner. However, these AAsTreg remained unable to control T cells stimulated by allogeneic mature dendritic cells, a phenomenon dependent on the release of proinflammatory cytokines. In vivo, Rapa administration reduced danger-associated IL-6 production, T cell proliferation, and graft infiltration. Based on these observations, AAsTreg were administered posttransplant (day 7) in combination with a short course of Rapa and rendered >80% long-term (>150 d) graft survival, a result superior to that achieved with polyclonal Treg. Moreover, graft protection was alloantigen-specific. Significantly, long-term graft survival was associated with alloreactive T cell anergy. These findings delineate combination of transient mammalian target of Rapa inhibition with appropriate AAsTreg selection as an effective approach to promote long-term organ graft survival.


Blood | 2015

The IL-33/ST2 axis augments effector T cell responses during acute GVHD

Dawn K. Reichenbach; Vincent Schwarze; Benjamin M. Matta; Victor Tkachev; Elisabeth Lieberknecht; Quan Liu; Brent H. Koehn; Dietmar Pfeifer; Patricia A. Taylor; Gabriele Prinz; Heide Dierbach; Natalie Stickel; Yvonne Beck; Max Warncke; Tobias Junt; Annette Schmitt-Graeff; Susumu Nakae; Marie Follo; Tobias Wertheimer; Lukas Schwab; Jason Devlin; Simon C. Watkins; Justus Duyster; James L.M. Ferrara; Heth R. Turnquist; Robert Zeiser; Bruce R. Blazar

Interleukin (IL)-33 binding to the receptor suppression of tumorigenicity 2 (ST2) produces pro-inflammatory and anti-inflammatory effects. Increased levels of soluble ST2 (sST2) are a biomarker for steroid-refractory graft-versus-host disease (GVHD) and mortality. However, whether sST2 has a role as an immune modulator or only as a biomarker during GVHD was unclear. We show increased IL-33 production by nonhematopoietic cells in the gastrointestinal (GI) tract in mice post-conditioning and patients during GVHD. Exogenous IL-33 administration during the peak inflammatory response worsened GVHD. Conversely, GVHD lethality and tumor necrosis factor-α production was significantly reduced in il33(-/-) recipients. ST2 was upregulated on murine and human alloreactive T cells and sST2 increased as experimental GVHD progressed. Concordantly, st2(-/-) vs wild-type (WT) donor T cells had a marked reduction in GVHD lethality and GI histopathology. Alloantigen-induced IL-18 receptor upregulation was lower in st2(-/-) T cells, and linked to reduced interferon-γ production by st2(-/-) vs WT T cells during GVHD. Blockade of IL-33/ST2 interactions during allogeneic-hematopoietic cell transplantation by exogenous ST2-Fc infusions had a marked reduction in GVHD lethality, indicating a role of ST2 as a decoy receptor modulating GVHD. Together, these studies point to the IL-33/ST2 axis as a novel and potent target for GVHD therapy.


Journal of Immunology | 2012

IL-27 Production and STAT3-Dependent Upregulation of B7-H1 Mediate Immune Regulatory Functions of Liver Plasmacytoid Dendritic Cells

Benjamin M. Matta; Giorgio Raimondi; Brian R. Rosborough; Tina L. Sumpter; Angus W. Thomson

Plasmacytoid dendritic cells (pDCs) are highly specialized APCs that, in addition to their well-recognized role in anti-viral immunity, also regulate immune responses. Liver-resident pDCs are considerably less immunostimulatory than those from secondary lymphoid tissues and are equipped to promote immune tolerance/regulation through various mechanisms. IL-27 is an IL-12 family cytokine that regulates the function of both APCs and T cells, although little is known about its role in pDC immunobiology. In this study, we show that mouse liver pDCs express higher levels of IL-27p28 and EBV-induced protein 3 (Ebi3) compared with those of splenic pDCs. Both populations of pDCs express the IL-27Rα/WSX-1; however, only liver pDCs significantly upregulate expression of the coregulatory molecule B7 homolog-1 (B7-H1) in response to IL-27. Inhibition of STAT3 activation completely abrogates IL-27–induced upregulation of B7-H1 expression on liver pDCs. Liver pDCs treated with IL-27 increase the percentage of CD4+Foxp3+ T cells in MLR, which is dependent upon expression of B7-H1. pDCs from Ebi3-deficient mice lacking functional IL-27 show increased capacity to stimulate allogeneic T cell proliferation and IFN-γ production in MLR. Liver but not spleen pDCs suppress delayed-type hypersensitivity responses to OVA, an effect that is lost with Ebi3−/− and B7-H1−/− liver pDCs compared with wild-type liver pDCs. These data suggest that IL-27 signaling in pDCs promotes their immunoregulatory function and that IL-27 produced by pDCs contributes to their capacity to regulate immune responses in vitro and in vivo.


Journal of Parenteral and Enteral Nutrition | 2005

Interactions Between Fatty Acids and Arginine Metabolism: Implications for the Design of Immune-Enhancing Diets

Vishal Bansal; Kimberly M. Syres; Valeryia Makarenkova; Ryan Brannon; Benjamin M. Matta; Brian G. Harbrecht; Juan B. Ochoa

BACKGROUND Trauma increases the enzyme arginase, thus depleting arginine necessary for producing nitric oxide. Arginine and omega-3 fatty acids are components in immune-enhancing diets. These diets decrease infections in surgical patients, perhaps by preventing arginine deficiency. This study examines whether omega-3 fatty acids alter the metabolic fate of arginine. Thus, we hypothesized there could be differential effects of varying prostaglandins on regulation of arginase. METHODS Prostaglandins PGE1, PGE2, and PGE3 were tested using RAW 264.7 cells cultured in the presence of these prostaglandins for 24 hours. IL-13 (10 ng/mL) was added 24 hours later to induce arginase I. NO production was induced by adding LPS (2 microg/mL) to the cultures after another 24 hours. RESULTS Arginase activity (nmol/min/mg) was induced by all prostaglandins but significantly more by PGE1 (466.05+/-30.25) and PGE2 (248.45+/-15.05) than PGE3 (139.87+/-19.88; p < .002) when co-cultured with IL-13. Western blots correlated the increase in arginase I expression. Nitrate levels (microM) were inversely proportional to activity with PGE3 having the highest production (3.89+/-0.19) and PGE2 and PGE1 with the lowest (2.75+/-0.49 and 1.54+/-0.19, respectively). Inhibition of arginase I using nor-hydroxyarginine increased and equalized nitrate levels. CONCLUSIONS Different prostaglandins significantly alter the metabolism of arginine. Prostaglandins from omega-6 fatty acids increases arginase I expression. By decreasing arginase I expression, prostaglandins from omega-3 fatty acids may increase available arginine. The specific combinations of dietary fatty acids and arginine should be considered when tailoring dietary regimens.


Annals of Surgery | 2010

Stat 6-Dependent Induction of Myeloid Derived Suppressor Cells After Physical Injury Regulates Nitric Oxide Response to Endotoxin

Veronica Munera; Petar J. Popovic; Jodie Bryk; John P. Pribis; David Caba; Benjamin M. Matta; Mazen S. Zenati; Juan B. Ochoa

Objective:To delineate the role of T-helper 2 (Th2) cytokines in the induction of trauma induced myeloid suppressor cells (TIMSC) and the regulation of nitric oxide production. Background:Trauma induces myeloid cells that express CD11b+/Gr1+ and arginase 1 and exhibit an immune suppressing activity. This article explores the mechanisms that induce TIMSC and the effects on nitric oxide production in response to endotoxin. Methods:TIMSC were studied in response to Th2 cytokines and a subsequent challenge to endotoxin. The role of Th2 cytokines was studied in STAT6−/− mice. Accumulation of TIMSC in spleens was studied using flow cytometry and immunhistochemistry. Plasma was recovered to measure accumulation of nitric oxide metabolites. Results:TIMSC accumulated in the spleen of injured mice and were particularly sensitive to IL-4 and IL-13 with large inductions of arginase activity. Significant blunting in both the accumulation of TIMSC in the spleen and induction of arginase 1 was observed in STAT6−/− mice after physical injury. Accumulation of nitric oxide metabolites to endotoxin was observed in STAT6−/− mice. Conclusion:This study shows that induction of CD11b+/Gr1+ cells after physical injury play an essential role in the regulation of nitric oxide production after a septic challenge. The accumulation and induction of arginase 1 in TIMSC is Th2 cytokine dependent. To our knowledge, the role of TIMSC in the regulation of nitric oxide is a novel finding. This observation adds to the possibility that TIMSC could play an important role in immunosuppression observed after physical injury.


Blood | 2013

Murine dendritic cell rapamycin-resistant and rictor-independent mTOR controls IL-10, B7-H1, and regulatory T-cell induction

Brian R. Rosborough; Dàlia Raïch-Regué; Benjamin M. Matta; Keunwook Lee; Boyi Gan; Ronald A. DePinho; Holger Hackstein; Mark Boothby; Hēth R. Turnquist; Angus W. Thomson

Mammalian target of rapamycin (mTOR) is an important, yet poorly understood integrative kinase that regulates immune cell function. mTOR functions in 2 independent complexes: mTOR complex (mTORC) 1 and 2. The immunosuppressant rapamycin (RAPA) inhibits mTORC1 but not mTORC2 and causes a paradoxical reduction in anti-inflammatory interleukin (IL) 10 and B7-homolog 1 (B7-H1) expression by dendritic cells (DCs). Using catalytic mTOR inhibitors and DCs lacking mTORC2, we show that restraint of signal transducer and activator of transcription 3-mediated IL-10 and B7-H1 expression during DC maturation involves a RAPA-insensitive and mTORC2-independent mTOR mechanism. Relatedly, catalytic mTOR inhibition promotes B7-H1-dependent and IL-1β-dependent DC induction of regulatory T cells (Tregs). Thus, we define an immunoregulatory pathway in which RAPA-sensitive mTORC1 in DCs promotes effector T-cell expansion and RAPA-insensitive mTORC1 restrains T(reg) induction. These findings identify the first known RAPA-insensitive mTOR pathway that is not mediated solely by mTORC2 and have implications for the use of catalytic mTOR inhibitors in inflammatory disease settings.


Journal of Immunology | 2012

Hepatic Stellate Cells Undermine the Allostimulatory Function of Liver Myeloid Dendritic Cells via STAT3-Dependent Induction of IDO

Tina L. Sumpter; Anil Dangi; Benjamin M. Matta; Chao Huang; Donna B. Stolz; Yoram Vodovotz; Angus W. Thomson; Chandrashekhar R. Gandhi

Hepatic stellate cells (HSCs) are critical for hepatic wound repair and tissue remodeling. They also produce cytokines and chemokines that may contribute to the maintenance of hepatic immune homeostasis and the inherent tolerogenicity of the liver. The functional relationship between HSCs and the professional migratory APCs in the liver, that is, dendritic cells (DCs), has not been evaluated. In this article, we report that murine liver DCs colocalize with HSCs in vivo under normal, steady-state conditions, and cluster with HSCs in vitro. In vitro, HSCs secrete high levels of DC chemoattractants, such as MΙP-1α and MCP-1, as well as cytokines that modulate DC activation, including TNF-α, IL-6, and IL-1β. Culture of HSCs with conventional liver myeloid (m) DCs resulted in increased IL-6 and IL-10 secretion compared with that of either cell population alone. Coculture also resulted in enhanced expression of costimulatory (CD80, CD86) and coinhibitory (B7-H1) molecules on mDCs. HSC-induced mDC maturation required cell–cell contact and could be blocked, in part, by neutralizing MΙP-1α or MCP-1. HSC-induced mDC maturation was dependent on activation of STAT3 in mDCs and, in part, on HSC-secreted IL-6. Despite upregulation of costimulatory molecules, mDCs conditioned by HSCs demonstrated impaired ability to induce allogeneic T cell proliferation, which was independent of B7-H1, but dependent upon HSC-induced STAT3 activation and subsequent upregulation of IDO. In conclusion, by promoting IDO expression, HSCs may act as potent regulators of liver mDCs and function to maintain hepatic homeostasis and tolerogenicity.

Collaboration


Dive into the Benjamin M. Matta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa Mathews

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Quan Liu

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giorgio Raimondi

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jeremy M. Lott

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge