Network


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

Hotspot


Dive into the research topics where Kanstantsin V. Katlinski is active.

Publication


Featured researches published by Kanstantsin V. Katlinski.


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.


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.


Journal of Investigative Dermatology | 2016

Therapeutic Elimination of the Type 1 Interferon Receptor for Treating Psoriatic Skin Inflammation

Jun Gui; Michael D. Gober; Xiaoping Yang; Kanstantsin V. Katlinski; Christine Marshall; Meena R. Sharma; Victoria P. Werth; Darren P. Baker; Hallgeir Rui; John T. Seykora; Serge Y. Fuchs

Phototherapy with UV light is a standard treatment for psoriasis, yet the mechanisms underlying the therapeutic effects are not well understood. Studies in human and mouse keratinocytes and in the skin tissues from human patients and mice showed that UV treatment triggers ubiquitination and downregulation of the type I IFN receptor chain IFNAR1, leading to suppression of IFN signaling and an ensuing decrease in the expression of inflammatory cytokines and chemokines. The severity of imiquimod-induced psoriasiform inflammation was greatly exacerbated in skin of mice deficient in IFNAR1 ubiquitination (Ifnar1(SA)). Furthermore, these mice did not benefit from UV phototherapy. Pharmacologic induction of IFNAR1 ubiquitination and degradation by an antiprotozoal agent halofuginone also relieved psoriasiform inflammation in wild-type but not in Ifnar1(SA) mice. These data identify downregulation of IFNAR1 by UV as a major mechanism of the UV therapeutic effects against the psoriatic inflammation and provide a proof of principle for future development of agents capable of inducing IFNAR1 ubiquitination and downregulation for the treatment of psoriasis.


Clinical Cancer Research | 2017

A potent in vivo anti-tumor efficacy of novel recombinant type I interferon.

kangjian zhang; Xiao-Fei Yin; Yuan-Qin Yang; Hui-Ling Li; Yan-Ni Xu; Lie-Yang Chen; Xi Jun Liu; Sujing Yuan; Xianlong Fang; Jing Xiao; Shuai Wu; Haineng Xu; Liang Chu; Kanstantsin V. Katlinski; Yuliya V. Katlinskaya; Rong-Bing Guo; Guang-Wen Wei; Da-Cheng Wang; Xinyuan Liu; Serge Y. Fuchs

Purpose: Antiproliferative, antiviral, and immunomodulatory activities of endogenous type I IFNs (IFN1) prompt the design of recombinant IFN1 for therapeutic purposes. However, most of the designed IFNs exhibited suboptimal therapeutic efficacies against solid tumors. Here, we report evaluation of the in vitro and in vivo antitumorigenic activities of a novel recombinant IFN termed sIFN-I. Experimental Design: We compared primary and tertiary structures of sIFN-I with its parental human IFNα-2b, as well as affinities of these ligands for IFN1 receptor chains and pharmacokinetics. These IFN1 species were also compared for their ability to induce JAK–STAT signaling and expression of the IFN1-stimulated genes and to elicit antitumorigenic effects. Effects of sIFN-I on tumor angiogenesis and immune infiltration were also tested in transplanted and genetically engineered immunocompetent mouse models. Results: sIFN-I displayed greater affinity for IFNAR1 (over IFNAR2) chain of the IFN1 receptor and elicited a greater extent of IFN1 signaling and expression of IFN-inducible genes in human cells. Unlike IFNα-2b, sIFN-I induced JAK–STAT signaling in mouse cells and exhibited an extended half-life in mice. Treatment with sIFN-I inhibited intratumoral angiogenesis, increased CD8+ T-cell infiltration, and robustly suppressed growth of transplantable and genetically engineered tumors in immunodeficient and immunocompetent mice. Conclusions: These findings define sIFN-I as a novel recombinant IFN1 with potent preclinical antitumorigenic effects against solid tumor, thereby prompting the assessment of sIFN-I clinical efficacy in humans. Clin Cancer Res; 23(8); 2038–49. ©2016 AACR.


Cancer Research | 2016

Abstract A63: IFNAR1 downregulation during melanoma progression upregulates αv expression and promotes metastasis

Angelica Ortiz; Yuliya V. Katlinskaya; Kanstantsin V. Katlinski; Serge Y. Fuchs

The anti-metastatic effects of type 1 interferon (IFN1) prompted its use for adjuvant therapy in malignant melanoma patients at high risk for developing metastasis. While IFN1 adjuvant therapy continues to be the preferred treatment, many patients fail to elicit response to IFN1 treatment and progress to develop distal metastases often resulting in a lethal outcome. We observed downregulation of the IFNAR1 chain of the IFN1 receptor and suppression of IFN1 signaling in genetically engineered mouse melanomas driven by melanocyte-specific activation of Braf and inactivation of Pten . Induction of melanoma in animals expressing IFNAR1 that is deficient in ubiquitination and degradation led to a delayed onset of the disease and decreased number of local metastases. Remarkably, no distant metastases were found in these mice despite substantial primary tumor burden. Gene set enrichment analysis demonstrated a dramatic loss of the αv integrin pathway signature in mice harboring stabilized IFNAR1. Conversely, melanomas induced in Ifnar1 knockout mice yielded transplantable cell lines that displayed high levels of αv expression. Restoring IFNAR1 expression in these cells led to the loss of αv expression. We discuss the role of these mechanisms in melanoma metastasis and refractoriness to IFN1-based therapy. Citation Format: Angelica Ortiz, Yuliya V. Katlinskaya, Kanstantsin V. Katlinski, Serge Y. Fuchs. IFNAR1 downregulation during melanoma progression upregulates αv expression and promotes metastasis. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Metastasis; 2015 Nov 30-Dec 3; Austin, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(7 Suppl):Abstract nr A63.


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


Cell Reports | 2013

A BRISC-SHMT Complex Deubiquitinates IFNAR1 and Regulates Interferon Responses

Hui Zheng; Vibhor Gupta; Jeffrey Patterson-Fortin; Sabyasachi Bhattacharya; Kanstantsin V. Katlinski; Junmin Wu; Bentley Varghese; Christopher J. Carbone; Bernadette Aressy; Serge Y. Fuchs; Roger A. Greenberg


Cytokine | 2009

Human soluble CXCR2 in health and disease

Sviatlana Akalovich; Kanstantsin V. Katlinski; Yuliya V. Katlinskaya; Tatyana Doroshenko; Nikolai N. Voitenok

Collaboration


Dive into the Kanstantsin V. Katlinski's collaboration.

Top Co-Authors

Avatar

Serge Y. Fuchs

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angelica Ortiz

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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
Top Co-Authors

Avatar

J. Alan Diehl

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jun Gui

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Qiujing Yu

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge