Network


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

Hotspot


Dive into the research topics where Matthew J. Riese is active.

Publication


Featured researches published by Matthew J. Riese.


Molecular Microbiology | 2002

Intracellular localization modulates targeting of ExoS, a type III cytotoxin, to eukaryotic signalling proteins.

Kristin J. Pederson; Rebecca Krall; Matthew J. Riese; Joseph T. Barbieri

ExoS is a bifunctional type III cytotoxin produced by Pseudomonas aeruginosa. Residues 96–232 comprise the Rho GTPase activating protein (Rho GAP) domain, whereas residues 233–453 comprise the 14‐3‐3‐dependent ADP‐ribosyltransferase domain. Earlier studies showed that the N‐terminus targeted ExoS to intracellular membranes within eukaryotic cells. This N‐terminal targeting region is now characterized for cellular and biological contributions to intoxications by ExoS. An ExoS(1–107)–green fluorescent protein (GFP) fusion protein co‐localized with α‐mannosidase, which indicated that the fusion protein localized near the Golgi. Residues 51–72 of ExoS (termed the membrane localization domain, MLD) were necessary and sufficient for membrane localization within eukaryotic cells. Deletion of the MLD did not inhibit type III secretion of ExoS from P. aeruginosa or type III delivery of ExoS into eukaryotic cells. Type III‐delivered ExoS(ΔMLD) localized within the cytosol of eukaryotic cells, whereas type III‐delivered ExoS was membrane associated. Although type III‐delivered ExoS(ΔMLD) stimulated the reorganization of the actin cytoskeleton (a Rho GAP activity), it did not ADP‐ribosylate Ras. Type III‐delivered ExoS(ΔMLD) and ExoS showed similar capacities for eliciting a cytotoxic response in CHO cells, which uncoupled the ADP‐ribosylation of Ras from the cytotoxicity elicited by ExoS.


Cancer Research | 2013

Enhanced Effector Responses in Activated CD8+ T Cells Deficient in Diacylglycerol Kinases

Matthew J. Riese; Liang-Chuan S. Wang; Edmund Moon; Rohan P. Joshi; Anjana Ranganathan; Carl H. June; Gary A. Koretzky; Steven M. Albelda

Recent clinical trials have shown promise in the use of chimeric antigen receptor (CAR)-transduced T cells; however, augmentation of their activity may broaden their clinical use and improve their efficacy. We hypothesized that because CAR action requires proteins essential for T-cell receptor (TCR) signal transduction, deletion of negative regulators of these signaling pathways would enhance CAR signaling and effector T-cell function. We tested CAR activity and function in T cells that lacked one or both isoforms of diacylglycerol kinase (dgk) expressed highly in T cells, dgkα and dgkζ, enzymes that metabolize the second messenger diacylglycerol (DAG) and limit Ras/ERK activation. We found that primary murine T cells transduced with CARs specific for the human tumor antigen mesothelin showed greatly enhanced cytokine production and cytotoxicity when cocultured with a murine mesothelioma line that stably expresses mesothelin. In addition, we found that dgk-deficient CAR-transduced T cells were more effective in limiting the growth of implanted tumors, both concurrent with and after establishment of tumor. Consistent with our studies in mice, pharmacologic inhibition of dgks also augments function of primary human T cells transduced with CARs. These results suggest that deletion of negative regulators of TCR signaling enhances the activity and function of CAR-expressing T cells and identify dgks as potential targets for improving the clinical potential of CARs.


Molecular and Cellular Biology | 2012

PERK Utilizes Intrinsic Lipid Kinase Activity To Generate Phosphatidic Acid, Mediate Akt Activation, and Promote Adipocyte Differentiation

Ekaterina Bobrovnikova-Marjon; Dariusz Pytel; Matthew J. Riese; Laura Pontano Vaites; N. Singh; Gary A. Koretzky; Eric S. Witze; J. A. Diehl

ABSTRACT The endoplasmic reticulum (ER) resident PKR-like kinase (PERK) is necessary for Akt activation in response to ER stress. We demonstrate that PERK harbors intrinsic lipid kinase, favoring diacylglycerol (DAG) as a substrate and generating phosphatidic acid (PA). This activity of PERK correlates with activation of mTOR and phosphorylation of Akt on Ser473. PERK lipid kinase activity is regulated in a phosphatidylinositol 3-kinase (PI3K) p85α-dependent manner. Moreover, PERK activity is essential during adipocyte differentiation. Because PA and Akt regulate many cellular functions, including cellular survival, proliferation, migratory responses, and metabolic adaptation, our findings suggest that PERK has a more extensive role in insulin signaling, insulin resistance, obesity, and tumorigenesis than previously thought.


Journal of Biological Chemistry | 2002

Auto-ADP-ribosylation of Pseudomonas aeruginosa ExoS

Matthew J. Riese; Udo-Michael Goehring; Mary Ehrmantraut; Joel Moss; Joseph T. Barbieri; Klaus Aktories; Gudula Schmidt

Pseudomonas aeruginosa Exoenzyme S (ExoS) is a bifunctional type-III cytotoxin. The N terminus possesses a Rho GTPase-activating protein (GAP) activity, whereas the C terminus comprises an ADP-ribosyltransferase domain. We investigated whether the ADP-ribosyltransferase activity of ExoS influences its GAP activity. Although the ADP-ribosyltransferase activity of ExoS is dependent upon FAS, a 14-3-3 family protein, factor-activating ExoS (FAS) had no influence on the activity of the GAP domain of ExoS (ExoS-GAP). In the presence of NAD and FAS, the GAP activity of full-length ExoS was reduced about 10-fold, whereas NAD and FAS did not affect the activity of the ExoS-GAP fragment. Using [32P]NAD, ExoS-GAP was identified as a substrate of the ADP-ribosyltransferase activity of ExoS. Site-directed mutagenesis revealed that auto-ADP-ribosylation of Arg-146 of ExoS was crucial for inhibition of GAP activityin vitro. To reveal the auto-ADP-ribosylation of ExoS in intact cells, tetanolysin was used to produce pores in the plasma membrane of Chinese hamster ovary (CHO) cells to allow the intracellular entry of [32P]NAD, the substrate for ADP-ribosylation. After a 3-h infection of CHO cells withPseudomonas aeruginosa, proteins of 50 and 25 kDa were preferentially ADP-ribosylated. The 50-kDa protein was determined to be auto-ADP-ribosylated ExoS, whereas the 25-kDa protein appeared to represent a group of proteins that included Ras.


Journal of Biological Chemistry | 2011

Decreased Diacylglycerol Metabolism Enhances ERK Activation and Augments CD8+ T Cell Functional Responses

Matthew J. Riese; Jashanpreet Grewal; Jayajit Das; Tao Zou; Vineet Patil; Arup K. Chakraborty; Gary A. Koretzky

Modulation of T cell receptor signal transduction in CD8+ T cells represents a novel strategy toward enhancing the immune response to tumor. Recently, levels of guanine exchange factors, RasGRP and SOS, within T cells have been shown to represent a key determinant in the regulation of the analog to the digital activation threshold of Ras. One important for regulating activation levels of RasGRP is diacylglycerol (DAG), and its levels are influenced by diacylglycerol kinase-ζ (DGKζ), which metabolizes DAG into phosphatidic acid, terminating DAG-mediated Ras signaling. We sought to determine whether DGKζ-deficient CD8+ T cells demonstrated enhanced in vitro responses in a manner predicted by the current model of Ras activation and to evaluate whether targeting this threshold confers enhanced CD8+ T cell responsiveness to tumor. We observed that DGKζ-deficient CD8+ T cells conform to most predictions of the current model of how RasGRP levels influence Ras activation. But our results differ in that the EC50 value of stimulation is not altered for any T cell receptor stimulus, a finding that suggests a further degree of complexity to how DGKζ deficiency affects signals important for Ras and ERK activation. Additionally, we found that DGKζ-deficient CD8+ T cells demonstrate enhanced responsiveness in a subcutaneous lymphoma model, implicating the analog to a digital conversion threshold as a novel target for potential therapeutic manipulation.


Science Signaling | 2013

The ζ Isoform of Diacylglycerol Kinase Plays a Predominant Role in Regulatory T Cell Development and TCR-Mediated Ras Signaling

Rohan P. Joshi; Amanda M. Schmidt; Jayajit Das; Dariusz Pytel; Matthew J. Riese; Lester M; J. A. Diehl; Edward M. Behrens; Taku Kambayashi; Gary A. Koretzky

Despite its relatively low abundance, DGKζ exerts a major inhibitory effect on T cell receptor signaling. Getting More Activity for Less DGK Strong antigen-dependent activation of the T cell receptor (TCR) in thymocytes leads to the development of natural regulatory T (nTreg) cells through a pathway involving diacylglycerol (DAG). Metabolism of DAG by diacylglycerol kinases (DGKs) to generate phosphatidic acid (PA) limits DAG signaling. In the second of a pair of papers, Joshi et al. characterized the relative contributions of DGKα and DGKζ to TCR-dependent signaling. Although DGKα was more abundant than DGKζ in the cytosol and at the interface between T cells and antigen-presenting cells, only loss of DGKζ enhanced TCR signaling and increased generation of nTreg cells in mice. Mathematical modeling suggested that these results might be explained by the differential catalytic activities of the DGK isoforms, and experiments in T cells showed that DGKζ produced the greatest amounts of PA. Together, these results suggest that the relatively low abundance of DGKζ in T cells belies its importance in inhibiting TCR signaling. Diacylglycerol (DAG) is a critical second messenger that mediates T cell receptor (TCR)–stimulated signaling. The abundance of DAG is reduced by the diacylglycerol kinases (DGKs), which catalyze the conversion of DAG to phosphatidic acid (PA) and thus inhibit DAG-mediated signaling. In T cells, the predominant DGK isoforms are DGKα and DGKζ, and deletion of the genes encoding either isoform enhances DAG-mediated signaling. We found that DGKζ, but not DGKα, suppressed the development of natural regulatory T (Treg) cells and predominantly mediated Ras and Akt signaling downstream of the TCR. The differential functions of DGKα and DGKζ were not attributable to differences in protein abundance in T cells or in their localization to the contact sites between T cells and antigen-presenting cells. RasGRP1, a key DAG-mediated activator of Ras signaling, associated to a greater extent with DGKζ than with DGKα; however, in silico modeling of TCR-stimulated Ras activation suggested that a difference in RasGRP1 binding affinity was not sufficient to cause differences in the functions of each DGK isoform. Rather, the model suggested that a greater catalytic rate for DGKζ than for DGKα might lead to DGKζ exhibiting increased suppression of Ras-mediated signals compared to DGKα. Consistent with this notion, experimental studies demonstrated that DGKζ was more effective than DGKα at catalyzing the metabolism of DAG to PA after TCR stimulation. The enhanced effective enzymatic production of PA by DGKζ is therefore one possible mechanism underlying the dominant functions of DGKζ in modulating Treg cell development.


Journal of Immunology | 2015

Regulatory T Cells Require TCR Signaling for Their Suppressive Function

Amanda M. Schmidt; Wen Lu; Vishal J. Sindhava; Yanping Huang; Janis K. Burkhardt; Enjun Yang; Matthew J. Riese; Jonathan S. Maltzman; Martha S. Jordan; Taku Kambayashi

Regulatory T cells (Tregs) are a subset of CD4+ T cells that maintain immune tolerance in part by their ability to inhibit the proliferation of conventional CD4+ T cells (Tconvs). The role of the TCR and the downstream signaling pathways required for this suppressive function of Tregs are not fully understood. To yield insight into how TCR-mediated signals influence Treg suppressive function, we assessed the ability of Tregs with altered TCR-mediated signaling capacity to inhibit Tconv proliferation. Mature Tregs deficient in Src homology 2 domain containing leukocyte protein of 76 kDa (SLP-76), an adaptor protein that nucleates the proximal signaling complex downstream of the TCR, were unable to inhibit Tconv proliferation, suggesting that TCR signaling is required for Treg suppressive function. Moreover, Tregs with defective phospholipase C γ (PLCγ) activation due to a Y145F mutation of SLP-76 were also defective in their suppressive function. Conversely, enhancement of diacylglycerol-mediated signaling downstream of PLCγ by genetic ablation of a negative regulator of diacylglycerol kinase ζ increased the suppressive ability of Tregs. Because SLP-76 is also important for integrin activation and signaling, we tested the role of integrin activation in Treg-mediated suppression. Tregs lacking the adaptor proteins adhesion and degranulation promoting adapter protein or CT10 regulator of kinase/CT10 regulator of kinase–like, which are required for TCR-mediated integrin activation, inhibited Tconv proliferation to a similar extent as wild-type Tregs. Together, these data suggest that TCR-mediated PLCγ activation, but not integrin activation, is required for Tregs to inhibit Tconv proliferation.


Infection and Immunity | 2002

Membrane Localization Contributes to the In Vivo ADP-Ribosylation of Ras by Pseudomonas aeruginosa ExoS

Matthew J. Riese; Joseph T. Barbieri

ABSTRACT Type III-delivered exoenzyme S (ExoS) preferentially ADP-ribosylated membrane-associated His6HRas, relative to its cytosolic derivative His6HRasΔCAAX. This indicates that the subcellular protein distribution contributes to in vivo ADP-ribosylation by ExoS.


Molecular Immunology | 2015

IQGAP1: insights into the function of a molecular puppeteer.

Alex M. Abel; Kristina M. Schuldt; Kamalakannan Rajasekaran; David Hwang; Matthew J. Riese; Sridhar Rao; Monica S. Thakar; Subramaniam Malarkannan

The intracellular spatiotemporal organization of signaling events is critical for normal cellular function. In response to environmental stimuli, cells utilize highly organized signaling pathways that are subject to multiple layers of regulation. However, the molecular mechanisms that coordinate these complex processes remain an enigma. Scaffolding proteins (scaffolins) have emerged as critical regulators of signaling pathways, many of which have well-described functions in immune cells. IQGAP1, a highly conserved cytoplasmic scaffold protein, is able to curb, compartmentalize, and coordinate multiple signaling pathways in a variety of cell types. IQGAP1 plays a central role in cell-cell interaction, cell adherence, and movement via actin/tubulin-based cytoskeletal reorganization. Evidence also implicates IQGAP1 as an essential regulator of the MAPK and Wnt/β-catenin signaling pathways. Here, we summarize the recent advances on the cellular and molecular biology of IQGAP1. We also describe how this pleiotropic scaffolin acts as a true molecular puppeteer, and highlight the significance of future research regarding the role of IQGAP1 in immune cells.


Cancer Research | 2015

Abstract CT137: Combination of agonistic CD40 monoclonal antibody CP-870,893 and anti-CTLA-4 antibody tremelimumab in patients with metastatic melanoma

David L. Bajor; Rosemarie Mick; Matthew J. Riese; Lee P. Richman; Xiaowei Xu; Drew A. Torigian; Erietta Stelekati; Martha Sweeney; Brendan Sullivan; Lynn M. Schuchter; Ravi K. Amaravadi; E. John Wherry; Robert H. Vonderheide

Purpose: Combining immunostimulatory monoclonal (mAb) and checkpoint inhibition may improve response rates and overall survival in patients with metastatic melanoma. CP-870,893 is an agonistic fully human IgG2 mAb targeting the immune stimulatory molecule CD40 (αCD40). Tremelimumab (treme) is a fully human IgG2 mAb targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA4), a negative co-stimulatory molecule found on T cells. Methods: Patients with metastatic melanoma were enrolled on a single-arm open-label phase I study. αCD40 and treme were dosed i.v. every 3 weeks and 12 weeks, respectively. Dose escalation followed a 3 + 3 strategy with alternating increases of αCD40 and treme in each cohort. Outcomes were scored using the Common Terminology Criteria for Adverse Events v3.0 and RECIST 1.0. Flow cytometric analysis of baseline blood samples was compared to samples obtained at least 3 weeks after the last dose of αCD40 and treme. Results: Toxicity: Twenty-four patients were enrolled and treated at 4 different dose levels. Two patients were replaced per protocol due to rapid, symptomatic progression of disease; these patients were not evaluated for response but were included in toxicity analysis. Dose-limiting toxicities (DLT), considered to be likely due to treatment, were colitis (n = 1) and hypophysitis (n = 1) and uveitis (n = 1). Cytokine release syndrome (CRS), grade 1-2, occurring within 24 hours of CP administration, was the most common treatment-related toxicity occurring in 19 (79.2%) patients. CRS symptoms were controlled with standard supportive care. All episodes of CRS resolved within 24 hours of αCD40 administration. The estimated maximum tolerated dose was 0.2 mg/kg of αCD40 and 10 mg/kg of treme. Outcomes: Overall objective response rate (ORR) was 27.3% (best response): two patients (9.1%) had complete responses to treatment and four (18.2%) patients had partial responses. The median follow-up was 22 months with a median progression free survival of 2.5 months and a median overall survival (OS) of 26.1 months. Correlative studies: Flow cytometric analysis of peripheral blood lymphocytes revealed changes in CD8+ T cell phenotypes. Double positive granzyme B+ and Ki-67+ cells increased from 0.56 ± 0.17% (mean ± standard error) for all CD8+ T cells at baseline to 1.01 ± 0.23% post-treatment (p = 0.03). CD8+ T cells expressing both programmed cell death 1 and eomesodermin were also higher after treatment, increasing from 7.7 ± 1.1% to 10.4 ±1.8% (p = 0.04) of all CD8+ T cells. Conclusion: Combination therapy with αCD40 and treme was well-tolerated in patients with metastatic melanoma, with rates of CRS and other toxicity similar to previously reported rates for αCD40 or treme treatments alone. Overall ORR was 27.3%, with median OS of 26.1 months. Given the tolerability, antitumor activity, and biomarker evidence of immune activation, expanded study of this combination should be pursued. Citation Format: David L. Bajor, Rosemarie Mick, Matthew J. Riese, Lee P. Richman, Xiaowei Xu, Drew A. Torigian, Erietta Stelekati, Martha Sweeney, Brendan Sullivan, Lynn M. Schuchter, Ravi Amaravadi, E. John Wherry, Robert H. Vonderheide. Combination of agonistic CD40 monoclonal antibody CP-870,893 and anti-CTLA-4 antibody tremelimumab in patients with metastatic melanoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT137. doi:10.1158/1538-7445.AM2015-CT137

Collaboration


Dive into the Matthew J. Riese's collaboration.

Top Co-Authors

Avatar

Gary A. Koretzky

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Joseph T. Barbieri

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Steven M. Albelda

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Bryon D. Johnson

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Sridhar Rao

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Debra K. Newman

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Amanda M. Schmidt

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Brendan Sullivan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David L. Bajor

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge