Network


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

Hotspot


Dive into the research topics where Marian Mayhue is active.

Publication


Featured researches published by Marian Mayhue.


Oncogene | 2015

Regulation of MET receptor tyrosine kinase signaling by suppressor of cytokine signaling 1 in hepatocellular carcinoma

Y. Gui; Mehdi Yeganeh; Y. C. Donates; W. S. Tobelaim; W. Chababi; Marian Mayhue; A. Yoshimura; Sheela Ramanathan; Caroline Saucier; Subburaj Ilangumaran

Suppressor of cytokine signaling 1 (SOCS1) is considered as a tumor suppressor protein in hepatocellular carcinoma (HCC), but the underlying mechanisms remain unclear. Previously, we have shown that SOCS1-deficient hepatocytes displayed increased responsiveness to hepatocyte growth factor (HGF) due to enhanced signaling via the MET receptor tyrosine kinase. As aberrant MET activation occurs in many tumors including HCC, here we elucidated the mechanisms of SOCS1-mediated regulation. SOCS1 attenuated HGF-induced proliferation of human and mouse HCC cell lines and their growth as tumors in NOD.scid.gamma mice. Tumors formed by SOCS1 expressing HCC cells showed significantly reduced MET expression, indicating that SOCS1 not only attenuates MET signaling but also regulates MET expression. Mechanistically, SOCS1 interacted with MET via the Src homology 2 domain and this interaction was promoted by MET tyrosine kinase activity. The SOCS1-mediated reduction in MET expression does not require the juxtamembrane Y1003 residue implicated in Cbl-mediated downmodulation. Moreover, the proteasome inhibitor MG-132, but not the inhibitors of lysosomal degradation bafilomycin and chloroquine, reversed the SOCS1-mediated reduction in MET expression, indicating that this process is distinct from Cbl-mediated downmodulation. Accordingly, SOCS1 promoted polyubiquitination of MET via K48-dependent but not K63-mediated ubiquitin chain elongation. Furthermore, siRNA-mediated downmodulation of Cbl did not abolish SOCS1-mediated reduction in MET expression in HCC cells. SOCS1-dependent ubiquitination of endogenous MET receptor occurred rapidly following HGF stimulation in HCC cells, leading to proteasomal degradation of phosphorylated MET receptor. These findings indicate that SOCS1 mediates its tumor suppressor functions, at least partly, by binding to MET and interfering with downstream signaling pathways as well as by promoting the turnover of the activated MET receptor. We propose that loss of this control mechanism due to epigenetic repression of SOCS1 could contribute to oncogenic MET signaling in HCC and other cancers, and that MET inhibitors might be useful in treating these patients.


OncoImmunology | 2016

NLRC5 elicits antitumor immunity by enhancing processing and presentation of tumor antigens to CD8+ T lymphocytes

Galaxia M. Rodriguez; Diwakar Bobbala; Daniel Serrano; Marian Mayhue; Audrey Champagne; Caroline Saucier; Viktor Steimle; Thomas A. Kufer; Alfredo Menendez; Sheela Ramanathan; Subburaj Ilangumaran

ABSTRACT Cancers can escape immunesurveillance by diminishing the expression of MHC class-I molecules (MHC-I) and components of the antigen-processing machinery (APM). Developing new approaches to reverse these defects could boost the efforts to restore antitumor immunity. Recent studies have shown that the expression of MHC-I and antigen-processing molecules is transcriptionally regulated by NOD-like receptor CARD domain containing 5 (NLRC5). To investigate whether NLRC5 could be used to improve tumor immunogenicity, we established stable lines of B16-F10 melanoma cells expressing NLRC5 (B16-5), the T cell co-stimulatory molecule CD80 (B16-CD80) or both (B16-5/80). Cells harboring NLRC5 constitutively expressed MHC-I and LMP2, LMP7 and TAP1 genes of the APM. The B16-5 cells efficiently presented the melanoma antigenic peptide gp10025–33 to Pmel-1 TCR transgenic CD8+ T cells and induced their proliferation. In the presence of CD80, B16-5 cells stimulated Pmel-1 cells even without the addition of gp100 peptide, indicating that NLRC5 facilitated the processing and presentation of endogenous tumor antigen. Upon subcutaneous implantation, B16-5 cells showed markedly reduced tumor growth in C57BL/6 hosts but not in immunodeficient hosts, indicating that the NLRC5-expressing tumor cells elicited antitumor immunity. Following intravenous injection, B16-5 and B16-5/80 cells formed fewer lung tumor foci compared to control cells. In mice depleted of CD8+ T cells, B16-5 cells formed large subcutaneous and lung tumors. Finally, immunization with irradiated B16-5 cells conferred protection against challenge by parental B16 cells. Collectively, our findings indicate that NLRC5 could be exploited to restore tumor immunogenicity and to stimulate protective antitumor immunity.


Cytokine | 2016

Interleukin-15-mediated inflammation promotes non-alcoholic fatty liver disease.

Yuneivy Cepero-Donates; Gregory Lacraz; Farnaz Ghobadi; Volatiana Rakotoarivelo; Sakina Orkhis; Marian Mayhue; Yi-Guang Chen; Marek Rola-Pleszczynski; Alfredo Menendez; Subburaj Ilangumaran; Sheela Ramanathan

Interleukin-15 (IL-15) is essential for the homeostasis of lymphoid cells particularly memory CD8(+) T cells and NK cells. These cells are abundant in the liver, and are implicated in obesity-associated pathogenic processes. Here we characterized obesity-associated metabolic and cellular changes in the liver of mice lacking IL-15 or IL-15Rα. High fat diet-induced accumulation of lipids was diminished in the livers of mice deficient for IL-15 or IL-15Rα. Expression of enzymes involved in the transport of lipids in the liver showed modest differences. More strikingly, the liver tissues of IL15-KO and IL15Rα-KO mice showed decreased expression of chemokines CCl2, CCL5 and CXCL10 and reduced infiltration of mononuclear cells. In vitro, IL-15 stimulation induced chemokine gene expression in wildtype hepatocytes, but not in IL15Rα-deficient hepatocytes. Our results show that IL-15 is implicated in the high fat diet-induced lipid accumulation and inflammation in the liver, leading to fatty liver disease.


Cellular & Molecular Immunology | 2014

IL-15 trans-presentation regulates homeostasis of CD4 + T lymphocytes

Xi-Lin Chen; Diwakar Bobbala; Yuneivy Cepero Donates; Marian Mayhue; Subburaj Ilangumaran; Sheela Ramanathan

Interleukin-15 (IL-15) is essential for the survival of memory CD8+ and CD4+ T cell subsets, and natural killer and natural killer T cells. Here, we describe a hitherto unreported role of IL-15 in regulating homoeostasis of naive CD4+ T cells. Adoptive transfer of splenocytes from non-obese diabetic (NOD) mice results in increased homeostatic expansion of T cells in lymphopenic NOD.scid.Il15−/− mice when compared to NOD.scid recipients. The increased accumulation of CD4+ T cells is also observed in NOD.Il15−/− mice, indicating that IL-15-dependent regulation also occurs in the absence of lymphopenia. NOD.scid mice lacking the IL-15Rα chain, but not those lacking the common gamma chain, also show increased accumulation of CD4+ T cells. These findings indicate that the IL-15-mediated regulation occurs directly on CD4+ T cells and requires trans-presentation of IL-15. CD4+ T cells expanding in the absence of IL-15 signaling do not acquire the characteristics of classical regulatory T cells. Rather, CD4+ T cells expanding in the absence of IL-15 show impaired antigen-induced activation and IFN-γ production. Based on these findings, we propose that the IL-15-dependent regulation of the naive CD4+ T-cell compartment may represent an additional layer of control to thwart potentially autoreactive cells that escape central tolerance, while permitting the expansion of memory T cells.


Clinical and Experimental Immunology | 2013

Induction of autoimmune diabetes in non-obese diabetic mice requires interleukin-21-dependent activation of autoreactive CD8+ T cells

Xi Lin Chen; Diwakar Bobbala; Galaxia M. Rodriguez; Marian Mayhue; Yi-Guang Chen; Subburaj Ilangumaran; Sheela Ramanathan

Non‐obese diabetic (NOD) mice lacking interleukin (IL)‐21 or IL‐21 receptor do not develop autoimmune type 1 diabetes (T1D). We have shown recently that IL‐21 may promote activation of autoreactive CD8+ T cells by increasing their antigen responsiveness. To investigate the role of IL‐21 in activating diabetogenic CD8+ T cells in the NOD mouse, we generated IL‐21‐deficient NOD mice expressing the highly pathogenic major histocompatibility complex (MHC) class‐I‐restricted 8.3 transgenic T cell receptor (TCR). IL‐21 deficiency protected 8.3‐NOD mice completely from T1D. CD8+ T cells from the 8.3‐NOD.Il21−/− mice showed decreased antigen‐induced proliferation but displayed robust antigen‐specific cytolytic activity and production of effector cytokines. IL‐21‐deficient 8.3 T cells underwent efficient homeostatic proliferation, and previous antigen stimulation enabled these cells to cause diabetes in NOD.Scid recipients. The 8.3 T cells that developed in an IL‐21‐deficient environment showed impaired antigen‐specific proliferation in vivo even in IL‐21‐sufficient mice. These cells also showed impaired IL‐2 production and Il2 gene transcription following antigen stimulation. However, IL‐2 addition failed to reverse their impaired proliferation completely. These findings indicate that IL‐21 is required for efficient initial activation of autoreactive CD8+ T cells but is dispensable for the activated cells to develop effector functions and cause disease. Hence, therapeutic targeting of IL‐21 in T1D may inhibit activation of naive autoreactive CD8+ T cells, but may have to be combined with other strategies to inhibit already activated cells.


Cellular & Molecular Immunology | 2017

Trans-presentation of interleukin-15 by interleukin-15 receptor alpha is dispensable for the pathogenesis of autoimmune type 1 diabetes

Diwakar Bobbala; Marian Mayhue; Alfredo Menendez; Subburaj Ilangumaran; Sheela Ramanathan

Interleukin-15 (IL-15) is a pro-inflammatory cytokine that is required for the survival and activation of memory CD8+T cells, natural killer (NK) cells, innate lymphoid cells, macrophages and dendritic cells. IL-15 is implicated in the pathogenesis of various autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease, psoriasis and autoimmune type 1 diabetes (T1D). IL-15 receptor (IL-15R) consists of a specific α chain, the β chain that is shared with IL-2R and the common γ chain. IL-15 is unique in the manner in which it binds and signals through its receptor subunits. IL-15 that is complexed with IL-15Rα binds to the βγ receptor complex present on the responding cell to mediate its biological effects through a process referred to as trans-presentation. The trans-presented IL-15 is essential to mediate the biological effects on T lymphocytes and NK cells. Here we show that IL-15, but not IL-15Rα, is required for the development of spontaneous and virus-induced T1D, viral clearance and for antigen cross-presentation to CD8+ T lymphocytes. Our findings provide insight into the complexities of IL-15 signalling in the initiation and maintenance of CD8+ T cell-mediated immune responses.


PLOS ONE | 2016

Correction: Deficiency of Interleukin-15 Confers Resistance to Obesity by Diminishing Inflammation and Enhancing the Thermogenic Function of Adipose Tissues

Gregory Lacraz; Volatiana Rakotoarivelo; Sébastien M. Labbé; Mathieu Vernier; Christophe Noll; Marian Mayhue; Jana Stankova; Adel Schwertani; Guillaume Grenier; André C. Carpentier; Denis Richard; Gerardo Ferbeyre; Julie Fradette; Marek Rola-Pleszczynski; Alfredo Menendez; Marie-France Langlois; Subburaj Ilangumaran; Sheela Ramanathan

[This corrects the article DOI: 10.1371/journal.pone.0162995.].


Cytokine | 2016

Negative regulation of the hepatic fibrogenic response by suppressor of cytokine signaling 1

Rajani Kandhi; Diwakar Bobbala; Mehdi Yeganeh; Marian Mayhue; Alfredo Menendez; Subburaj Ilangumaran

Suppressor of cytokine signaling 1 (SOCS1) is an indispensable regulator of IFNγ signaling and has been implicated in the regulation of liver fibrosis. However, it is not known whether SOCS1 mediates its anti-fibrotic functions in the liver directly, or via modulating IFNγ, which has been implicated in attenuating hepatic fibrosis. Additionally, it is possible that SOCS1 controls liver fibrosis by regulating hepatic stellate cells (HSC), a key player in fibrogenic response. While the activation pathways of HSCs have been well characterized, the regulatory mechanisms are not yet clear. The goals of this study were to dissociate IFNγ-dependent and SOCS1-mediated regulation of hepatic fibrogenic response, and to elucidate the regulatory functions of SOCS1 in HSC activation. Liver fibrosis was induced in Socs1(-/-)Ifng(-/-) mice with dimethylnitrosamine or carbon tetrachloride. Ifng(-/-) and C57BL/6 mice served as controls. Following fibrogenic treatments, Socs1(-/-)Ifng(-/-) mice showed elevated serum ALT levels and increased liver fibrosis compared to Ifng(-/-) mice. The latter group showed higher ALT levels and fibrosis than C57BL/6 controls. The livers of SOCS1-deficient mice showed bridging fibrosis, which was associated with increased accumulation of myofibroblasts and abundant collagen deposition. SOCS1-deficient livers showed increased expression of genes coding for smooth muscle actin, collagen, and enzymes involved in remodeling the extracellular matrix, namely matrix metalloproteinases and tissue inhibitor of metalloproteinases. Primary HSCs from SOCS1-deficient mice showed increased proliferation in response to growth factors such as HGF, EGF and PDGF, and the fibrotic livers of SOCS1-deficient mice showed increased expression of the Pdgfb gene. Taken together, these data indicate that SOCS1 controls liver fibrosis independently of IFNγ and that part of this regulation may occur via regulating HSC proliferation and limiting growth factor availability.


PLOS ONE | 2015

GIMAP5 Deficiency Is Associated with Increased AKT Activity in T Lymphocytes

Xi-Lin Chen; Daniel Serrano; Marian Mayhue; Kasper Hoebe; Subburaj Ilangumaran; Sheela Ramanathan

Long-term survival of T lymphocytes in quiescent state is essential to maintain their cell numbers in secondary lymphoid organs. In mice and in rats, the loss of functional GTPase of the immune associated nucleotide binding protein 5 (GIMAP5) causes peripheral T lymphopenia due to spontaneous death of T cells. The underlying mechanism responsible for the disruption of quiescence in Gimap5 deficient T cells remains largely unknown. In this study, we show that loss of functional Gimap5 results in increased basal activation of mammalian target of rapamycin (mTOR), independent of protein phosphatase 2A (PP2A) or AMP-activated protein kinase (AMPK). Our results suggest that the constitutive activation of the phosphoinositide 3-kinase (PI3K) pathway may be one of the consequences of the absence of functional GIMAP5.


PLOS ONE | 2016

TCR and IL-7 Signaling Are Altered in the Absence of Functional GTPase of the Immune Associated Nucleotide Binding Protein 5 (GIMAP5)

Xi-Lin Chen; Daniel Serrano; Farnaz Ghobadi; Marian Mayhue; Kasper Hoebe; Subburaj Ilangumaran; Sheela Ramanathan

GTPase of the immune associated nucleotide binding protein (GIMAP) family of proteins are expressed essentially in cells of the hematopoietic system. Mutation in the founding member of this gene family, Gimap5, results in the lymphopenic phenotype in Bio-Breeding diabetes prone rats. In mice, deletion of functional Gimap5 gene affects the survival and renewal of hematopoietic stem cells in addition to the defects observed in T cells. Here we show that T cells from OTII TCR-transgenic Gimap5sph/sph mice do not proliferate in response to its cognate antigen. Furthermore, T cells from Gimap5 mutant rats and mice show decreased phosphorylation of STAT5 following stimulation with IL-7. Our results suggest that functional Gimap5 is required for optimal signaling through TCR and IL-7R in T cells.

Collaboration


Dive into the Marian Mayhue's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diwakar Bobbala

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi-Guang Chen

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xi Lin Chen

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar

Daniel Serrano

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar

Jana Stankova

Université de Sherbrooke

View shared research outputs
Top Co-Authors

Avatar

Xi-Lin Chen

Université de Sherbrooke

View shared research outputs
Researchain Logo
Decentralizing Knowledge