Network


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

Hotspot


Dive into the research topics where Serge Y. Fuchs is active.

Publication


Featured researches published by Serge Y. Fuchs.


Cell Reports | 2015

DNA-Damage-Induced Type I Interferon Promotes Senescence and Inhibits Stem Cell Function

Qiujing Yu; Yuliya V. Katlinskaya; Christopher J. Carbone; Bin Zhao; Kanstantsin V. Katlinski; Hui Zheng; Manti Guha; Ning Li; Qijun Chen; Ting Yang; Christopher J. Lengner; Roger A. Greenberg; F. Brad Johnson; Serge Y. Fuchs

Expression of type I interferons (IFNs) can be induced by DNA-damaging agents, but the mechanisms and significance of this regulation are not completely understood. We found that the transcription factor IRF3, activated in an ATM-IKKα/β-dependent manner, stimulates cell-autonomous IFN-β expression in response to double-stranded DNA breaks. Cells and tissues with accumulating DNA damage produce endogenous IFN-β and stimulate IFN signaling in vitro and in vivo. In turn, IFN acts to amplify DNA-damage responses, activate the p53 pathway, promote senescence, and inhibit stem cell function in response to telomere shortening. Inactivation of the IFN pathway abrogates the development of diverse progeric phenotypes and extends the lifespan of Terc knockout mice. These data identify DNA-damage-response-induced IFN signaling as a critical mechanism that links accumulating DNA damage with senescence and premature aging.


Journal of Clinical Investigation | 2015

ATF4-dependent induction of heme oxygenase 1 prevents anoikis and promotes metastasis

Souvik Dey; Carly M. Sayers; Ioannis I. Verginadis; Stacey L. Lehman; Yi Cheng; George J. Cerniglia; Stephen W. Tuttle; Michael Feldman; Paul J. Zhang; Serge Y. Fuchs; J. Alan Diehl; Constantinos Koumenis

The integrated stress response (ISR) is a critical mediator of cancer cell survival, and targeting the ISR inhibits tumor progression. Here, we have shown that activating transcription factor 4 (ATF4), a master transcriptional effector of the ISR, protects transformed cells against anoikis - a specialized form of apoptosis - following matrix detachment and also contributes to tumor metastatic properties. Upon loss of attachment, ATF4 activated a coordinated program of cytoprotective autophagy and antioxidant responses, including induced expression of the major antioxidant enzyme heme oxygenase 1 (HO-1). HO-1 upregulation was the result of simultaneous activation of ATF4 and the transcription factor NRF2, which converged on the HO1 promoter. Increased levels of HO-1 ameliorated oxidative stress and cell death. ATF4-deficient human fibrosarcoma cells were unable to colonize the lungs in a murine model, and reconstitution of ATF4 or HO-1 expression in ATF4-deficient cells blocked anoikis and rescued tumor lung colonization. HO-1 expression was higher in human primary and metastatic tumors compared with noncancerous tissue. Moreover, HO-1 expression correlated with reduced overall survival of patients with lung adenocarcinoma and glioblastoma. These results establish HO-1 as a mediator of ATF4-dependent anoikis resistance and tumor metastasis and suggest ATF4 and HO-1 as potential targets for therapeutic intervention in solid tumors.


Cancer Cell | 2017

Inactivation of Interferon Receptor Promotes the Establishment of Immune Privileged Tumor Microenvironment

Kanstantsin V. Katlinski; Jun Gui; Yuliya V. Katlinskaya; Angelica Ortiz; Riddhita Chakraborty; Sabyasachi Bhattacharya; Christopher J. Carbone; Daniel P. Beiting; Melanie A. Girondo; Amy R. Peck; Ellen Puré; Priya Chatterji; Anil K. Rustgi; J. Alan Diehl; Constantinos Koumenis; Hallgeir Rui; Serge Y. Fuchs

Refractoriness of solid tumors, including colorectal cancers (CRCs), to immunotherapies is attributed to the immunosuppressive tumor microenvironment that protects malignant cells from cytotoxic T lymphocytes (CTLs). We found that downregulation of the type I interferon receptor chain IFNAR1 occurs in human CRC and mouse models of CRC. Downregulation of IFNAR1 in tumor stroma stimulated CRC development and growth, played a key role in formation of the immune-privileged niche, and predicted poor prognosis inxa0human CRC patients. Genetic stabilization of IFNAR1 improved CTL survival and increased the efficacy of the chimeric antigen receptor Txa0cell transfer and PD-1 inhibition. Likewise, pharmacologic stabilization of IFNAR1 suppressed tumor growth providing the rationale for upregulating IFNAR1 to improve anti-cancer therapies.


Nature Communications | 2016

miR-216b regulation of c-Jun mediates GADD153/CHOP-dependent apoptosis

Zhenhua Xu; Yiwen Bu; Nilesh Chitnis; Costas Koumenis; Serge Y. Fuchs; J. Alan Diehl

The ability of the unfolded protein response, UPR, to regulate cell homeostasis through both gene expression and protein synthesis has been well documented. One primary pro-apoptotic protein that responds to both PERK and Ire1 signalling is the CHOP/GADD153 transcription factor. Although CHOP deficiency delays onset of cell death, questions remain regarding how CHOP regulates apoptosis. Here, we provide evidence demonstrating that CHOP/GADD153-dependent apoptosis reflects expression of micro-RNA, miR-216b. MiR-216b accumulation requires PERK-dependent induction of CHOP/GADD153, which then directly regulates miR-216b expression. As maximal expression of miR-216b is antagonized by Ire1, miR-216b accumulation reflects the convergence of PERK and Ire1 activities. Functionally, miR-216b directly targets c-Jun, thereby reducing AP-1-dependent transcription and sensitizing cells to ER stress-dependent apoptosis. These results provide direct insight into the molecular mechanisms of CHOP/GADD153-dependent cell death.


Journal of Biological Chemistry | 2015

Type I Interferon Controls Propagation of Long Interspersed Element-1

Qiujing Yu; Christopher J. Carbone; Yuliya V. Katlinskaya; Hui Zheng; Ke Zheng; Mengcheng Luo; P. Jeremy Wang; Roger A. Greenberg; Serge Y. Fuchs

Background: Type 1 interferons (IFN1) mediate defense against viruses but their role in regulating retrotransposon activities is unknown. Results: LINE-1 retrotransposon induces IFN1, which in turn inhibits LINE-1 retrotransposition. Conclusion: IFN1 regulate activities and propagation of LINE-1. Significance: Given that retrotransposons alter the genome, IFN1 play a role in maintenance of genomic integrity. Type I interferons (IFN) including IFNα and IFNβ are critical for the cellular defense against viruses. Here we report that increased levels of IFNβ were found in testes from mice deficient in MOV10L1, a germ cell-specific RNA helicase that plays a key role in limiting the propagation of retrotransposons including Long Interspersed Element-1 (LINE-1). Additional experiments revealed that activation of LINE-1 retrotransposons increases the expression of IFNβ and of IFN-stimulated genes. Conversely, pretreatment of cells with IFN suppressed the replication of LINE-1. Furthermore, the efficacy of LINE-1 replication was increased in isogenic cell lines harboring inactivating mutations in diverse elements of the IFN signaling pathway. Knockdown of the IFN receptor chain IFNAR1 also stimulated LINE-1 propagation in vitro. Finally, a greater accumulation of LINE-1 was found in mice that lack IFNAR1 compared with wild type mice. We propose that LINE-1-induced IFN plays an important role in restricting LINE-1 propagation and discuss the putative role of IFN in preserving the genome stability.


Journal of Virology | 2016

Hemagglutinin of Influenza A Virus Antagonizes Type I Interferon (IFN) Responses by Inducing Degradation of Type I IFN Receptor 1

Chuan Xia; Madhuvanthi Vijayan; Curtis J. Pritzl; Serge Y. Fuchs; Adrian B. McDermott; Bumsuk Hahm

ABSTRACT Influenza A virus (IAV) employs diverse strategies to circumvent type I interferon (IFN) responses, particularly by inhibiting the synthesis of type I IFNs. However, it is poorly understood if and how IAV regulates the type I IFN receptor (IFNAR)-mediated signaling mode. In this study, we demonstrate that IAV induces the degradation of IFNAR subunit 1 (IFNAR1) to attenuate the type I IFN-induced antiviral signaling pathway. Following infection, the level of IFNAR1 protein, but not mRNA, decreased. Indeed, IFNAR1 was phosphorylated and ubiquitinated by IAV infection, which resulted in IFNAR1 elimination. The transiently overexpressed IFNAR1 displayed antiviral activity by inhibiting virus replication. Importantly, the hemagglutinin (HA) protein of IAV was proved to trigger the ubiquitination of IFNAR1, diminishing the levels of IFNAR1. Further, influenza A viral HA1 subunit, but not HA2 subunit, downregulated IFNAR1. However, viral HA-mediated degradation of IFNAR1 was not caused by the endoplasmic reticulum (ER) stress response. IAV HA robustly reduced cellular sensitivity to type I IFNs, suppressing the activation of STAT1/STAT2 and induction of IFN-stimulated antiviral proteins. Taken together, our findings suggest that IAV HA causes IFNAR1 degradation, which in turn helps the virus escape the powerful innate immune system. Thus, the research elucidated an influenza viral mechanism for eluding the IFNAR signaling pathway, which could provide new insights into the interplay between influenza virus and host innate immunity. IMPORTANCE Influenza A virus (IAV) infection causes significant morbidity and mortality worldwide and remains a major health concern. When triggered by influenza viral infection, host cells produce type I interferon (IFN) to block viral replication. Although IAV was shown to have diverse strategies to evade this powerful, IFN-mediated antiviral response, it is not well-defined if IAV manipulates the IFN receptor-mediated signaling pathway. Here, we uncovered that influenza viral hemagglutinin (HA) protein causes the degradation of type I IFN receptor subunit 1 (IFNAR1). HA promoted phosphorylation and polyubiquitination of IFNAR1, which facilitated the degradation of this receptor. The HA-mediated elimination of IFNAR1 notably decreased the cells sensitivities to type I IFNs, as demonstrated by the diminished expression of IFN-induced antiviral genes. This discovery could help us understand how IAV regulates the host innate immune response to create an environment optimized for viral survival in host cells.


Cell Reports | 2016

Suppression of Type I Interferon Signaling Overcomes Oncogene-Induced Senescence and Mediates Melanoma Development and Progression

Yuliya V. Katlinskaya; Kanstantsin V. Katlinski; Qiujing Yu; Angelica Ortiz; Daniel P. Beiting; Angela K Brice; Diwakar Davar; Cindy Sanders; John M. Kirkwood; Hallgeir Rui; Xiaowei Xu; Constantinos Koumenis; J. Alan Diehl; Serge Y. Fuchs

Oncogene activation induces DNA damage responses and cell senescence. We report a key role of type I interferons (IFNs) in oncogene-induced senescence. IFN signaling-deficient melanocytes expressing activated Braf do not exhibit senescence and develop aggressive melanomas. Restoration of IFN signaling in IFN-deficient melanoma cells induces senescence and suppresses melanoma progression. Additional data from human melanoma patients and mouse transplanted tumor models suggest the importance of non-cell-autonomous IFN signaling. Inactivation of the IFN pathway is mediated by the IFN receptor IFNAR1 downregulation that invariably occurs during melanoma development. Mice harboring an IFNAR1 mutant, which is partially resistant to downregulation, delay melanoma development, suppress metastatic disease, and better respond to BRAF or PD-1 inhibitors. These results suggest that IFN signaling is an important tumor-suppressive pathway that inhibits melanoma development and progression and argue for targeting IFNAR1 downregulation to prevent metastatic disease and improve the efficacy of molecularly target and immune-targeted melanoma therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Type I interferons mediate pancreatic toxicities of PERK inhibition

Qiujing Yu; Bin Zhao; Jun Gui; Kanstantsin V. Katlinski; Angela K Brice; Yan Gao; Changhong Li; Jake A. Kushner; Constantinos Koumenis; J. Alan Diehl; Serge Y. Fuchs

Significance Inactivating pancreatic endoplasmic reticulum kinase (PERK) mutations cause pancreatic degeneration and diabetes in patients with Wolcott-Rallison syndrome. Pancreatic injury is also observed in mice upon PERK genetic ablation or treatment with PERK inhibitors. This toxicity (the mechanisms of which are poorly understood) impedes the clinical development of PERK inhibitors, which show promise against cancers and neurodegenerative diseases. Here we demonstrate that activation of type 1 interferon signaling occurs upon PERK ablation and is responsible for pancreatic injury and the loss of exocrine and endocrine tissues and functions. Neutralization of interferon signaling protects the pancreas from deleterious effects of PERK inhibitors. Temporally targeting the interferon pathway may help with the treatment of patients with Wolcott-Rallison syndrome and the use of PERK inhibitors against other diseases. The great preclinical promise of the pancreatic endoplasmic reticulum kinase (PERK) inhibitors in neurodegenerative disorders and cancers is marred by pancreatic injury and diabetic syndrome observed in PERK knockout mice and humans lacking PERK function and suffering from Wolcott-Rallison syndrome. PERK mediates many of the unfolded protein response (UPR)-induced events, including degradation of the type 1 interferon (IFN) receptor IFNAR1 in vitro. Here we report that whole-body or pancreas-specific Perk ablation in mice leads to an increase in IFNAR1 protein levels and signaling in pancreatic tissues. Concurrent IFNAR1 deletion attenuated the loss of PERK-deficient exocrine and endocrine pancreatic tissues and prevented the development of diabetes. Experiments using pancreas-specific Perk knockouts, bone marrow transplantation, and cultured pancreatic islets demonstrated that stabilization of IFNAR1 and the ensuing increased IFN signaling in pancreatic tissues represents a major driver of injury triggered by Perk loss. Neutralization of IFNAR1 prevented pancreatic toxicity of PERK inhibitor, indicating that blocking the IFN pathway can mitigate human genetic disorders associated with PERK deficiency and help the clinical use of PERK inhibitors.


Molecular and Cellular Biology | 2016

Type I Interferons Control Proliferation and Function of the Intestinal Epithelium

Yuliya V. Katlinskaya; Kanstantsin V. Katlinski; Audrey Lasri; Ning Li; Daniel P. Beiting; Amy C. Durham; Ting Yang; Eli Pikarsky; Christopher J. Lengner; F. Brad Johnson; Yinon Ben-Neriah; Serge Y. Fuchs

ABSTRACT Wnt pathway-driven proliferation and renewal of the intestinal epithelium must be tightly controlled to prevent development of cancer and barrier dysfunction. Although type I interferons (IFN) produced in the gut under the influence of microbiota are known for their antiproliferative effects, the role of these cytokines in regulating intestinal epithelial cell renewal is largely unknown. Here we report a novel role for IFN in the context of intestinal knockout of casein kinase 1α (CK1α), which controls the ubiquitination and degradation of both β-catenin and the IFNAR1 chain of the IFN receptor. Ablation of CK1α leads to the activation of both β-catenin and IFN pathways and prevents the unlimited proliferation of intestinal epithelial cells despite constitutive β-catenin activity. IFN signaling contributes to the activation of the p53 pathway and the appearance of apoptotic and senescence markers in the CK1α-deficient gut. Concurrent genetic ablation of CK1α and IFNAR1 leads to intestinal hyperplasia, robust attenuation of apoptosis, and rapid and lethal loss of barrier function. These data indicate that IFN play an important role in controlling the proliferation and function of the intestinal epithelium in the context of β-catenin activation.


PLOS Genetics | 2016

PERK Is a Haploinsufficient Tumor Suppressor: Gene Dose Determines Tumor-Suppressive Versus Tumor Promoting Properties of PERK in Melanoma.

Dariusz Pytel; Yan Gao; Katarzyna Mackiewicz; Yuliya V. Katlinskaya; Kirk A. Staschke; Maria Cristina Garcia Paredes; Akihiro Yoshida; Shuo Qie; Gao Zhang; Olga S. Chajewski; Lawrence Wu; Ireneusz Majsterek; Meenhard Herlyn; Serge Y. Fuchs; J. Alan Diehl

The unfolded protein response (UPR) regulates cell fate following exposure of cells to endoplasmic reticulum stresses. PERK, a UPR protein kinase, regulates protein synthesis and while linked with cell survival, exhibits activities associated with both tumor progression and tumor suppression. For example, while cells lacking PERK are sensitive to UPR-dependent cell death, acute activation of PERK triggers both apoptosis and cell cycle arrest, which would be expected to contribute tumor suppressive activity. We have evaluated these activities in the BRAF-dependent melanoma and provide evidence revealing a complex role for PERK in melanoma where a 50% reduction is permissive for BrafV600E-dependent transformation, while complete inhibition is tumor suppressive. Consistently, PERK mutants identified in human melanoma are hypomorphic with dominant inhibitory function. Strikingly, we demonstrate that small molecule PERK inhibitors exhibit single agent efficacy against BrafV600E-dependent tumors highlighting the clinical value of targeting PERK.

Collaboration


Dive into the Serge Y. Fuchs's collaboration.

Top Co-Authors

Avatar

J. Alan Diehl

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qiujing Yu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angelica Ortiz

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Bin Zhao

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Daniel P. Beiting

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Hallgeir Rui

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge