Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
PLoS Genet ; 12(12): e1006518, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27977682

ABSTRACT

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.


Subject(s)
Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Tumor Suppressor Proteins/genetics , eIF-2 Kinase/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Gene Dosage/genetics , Haploinsufficiency/genetics , Humans , Melanoma/drug therapy , Melanoma/pathology , Mutation , Signal Transduction/drug effects , Signal Transduction/genetics , Small Molecule Libraries/administration & dosage , Tumor Suppressor Proteins/biosynthesis , Unfolded Protein Response/genetics , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/biosynthesis
2.
J Biol Chem ; 290(16): 10191-9, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25716322

ABSTRACT

Type I interferons (IFN) including IFNα and IFNß are critical for the cellular defense against viruses. Here we report that increased levels of IFNß 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 IFNß 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.


Subject(s)
Fibroblasts/metabolism , Interferon-alpha/genetics , Interferon-beta/genetics , Long Interspersed Nucleotide Elements , Animals , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/immunology , Gene Expression Regulation , Genomic Instability , HeLa Cells , Humans , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Primary Cell Culture , RNA Helicases/deficiency , RNA Helicases/genetics , RNA Helicases/immunology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction , Testis/cytology , Testis/immunology , Testis/metabolism
3.
Cancer Discov ; 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38975874

ABSTRACT

KRAS inhibitors demonstrate clinical efficacy in pancreatic ductal adenocarcinoma (PDAC); however, resistance is common. Among patients with KRASG12C-mutant PDAC treated with adagrasib or sotorasib, mutations in PIK3CA and KRAS, and amplifications of KRASG12C, MYC, MET, EGFR, and CDK6 emerged at acquired resistance. In PDAC cell lines and organoid models treated with the KRASG12D inhibitor MRTX1133, epithelial-to-mesenchymal transition and PI3K-AKT-mTOR signaling associate with resistance to therapy. MRTX1133 treatment of the KrasLSL-G12D/+;Trp53LSL-R172H/+;p48-Cre (KPC) mouse model yielded deep tumor regressions, but drug resistance ultimately emerged, accompanied by amplifications of Kras, Yap1, Myc, and Cdk6/Abcb1a/b, and co-evolution of drug-resistant transcriptional programs. Moreover, in KPC and PDX models, mesenchymal and basal-like cell states displayed increased response to KRAS inhibition compared to the classical state. Combination treatment with KRASG12D inhibition and chemotherapy significantly improved tumor control in PDAC mouse models. Collectively, these data elucidate co-evolving resistance mechanisms to KRAS inhibition and support multiple combination therapy strategies.

4.
Cancer Cell ; 35(1): 33-45.e6, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30645975

ABSTRACT

Tumor-derived extracellular vesicles (TEV) "educate" healthy cells to promote metastases. We found that melanoma TEV downregulated type I interferon (IFN) receptor and expression of IFN-inducible cholesterol 25-hydroxylase (CH25H). CH25H produces 25-hydroxycholesterol, which inhibited TEV uptake. Low CH25H levels in leukocytes from melanoma patients correlated with poor prognosis. Mice incapable of downregulating the IFN receptor and Ch25h were resistant to TEV uptake, TEV-induced pre-metastatic niche, and melanoma lung metastases; however, ablation of Ch25h reversed these phenotypes. An anti-hypertensive drug, reserpine, suppressed TEV uptake and disrupted TEV-induced formation of the pre-metastatic niche and melanoma lung metastases. These results suggest the importance of CH25H in defense against education of normal cells by TEV and argue for the use of reserpine in adjuvant melanoma therapy.


Subject(s)
Extracellular Vesicles/metabolism , Lung Neoplasms/secondary , Melanoma/pathology , Receptor, Interferon alpha-beta/metabolism , Steroid Hydroxylases/metabolism , Animals , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Interferons/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Melanoma/metabolism , Mice , Neoplasm Metastasis , Oxysterols/metabolism , Reserpine/administration & dosage , Reserpine/pharmacology , Steroid Hydroxylases/genetics , THP-1 Cells
5.
Cancer Cell ; 31(2): 194-207, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28196594

ABSTRACT

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 in human CRC patients. Genetic stabilization of IFNAR1 improved CTL survival and increased the efficacy of the chimeric antigen receptor T cell transfer and PD-1 inhibition. Likewise, pharmacologic stabilization of IFNAR1 suppressed tumor growth providing the rationale for upregulating IFNAR1 to improve anti-cancer therapies.


Subject(s)
Colorectal Neoplasms/immunology , Receptor, Interferon alpha-beta/physiology , Animals , Cell Survival , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Down-Regulation , Humans , Immune Tolerance , Mice , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/analysis , Receptor, Interferon alpha-beta/genetics , Signal Transduction , T-Lymphocytes, Cytotoxic/physiology , Tumor Microenvironment
6.
Clin Cancer Res ; 23(8): 2038-2049, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27683179

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Interferon-alpha/chemistry , Interferon-alpha/pharmacology , Animals , Female , Flow Cytometry , Humans , Immunoblotting , Interferon alpha-2 , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms, Experimental/drug therapy , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Surface Plasmon Resonance , Xenograft Model Antitumor Assays
7.
Cell Rep ; 15(1): 171-180, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27052162

ABSTRACT

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.


Subject(s)
Cellular Senescence , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/genetics , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Down-Regulation , Female , HEK293 Cells , Humans , Interferon Type I/metabolism , Male , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Mutation , Proto-Oncogene Proteins B-raf/metabolism , Receptor, Interferon alpha-beta/genetics
8.
Mol Cell Biol ; 36(7): 1124-35, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26811327

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 ß-catenin and the IFNAR1 chain of the IFN receptor. Ablation of CK1α leads to the activation of both ß-catenin and IFN pathways and prevents the unlimited proliferation of intestinal epithelial cells despite constitutive ß-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 ß-catenin activation.


Subject(s)
Interferon Type I/physiology , Intestinal Mucosa/cytology , Receptor, Interferon alpha-beta/metabolism , Animals , Apoptosis , Casein Kinase Ialpha/genetics , Casein Kinase Ialpha/metabolism , Cell Proliferation , DNA Damage , Intestinal Mucosa/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Ubiquitination
9.
Cancer Biol Ther ; 16(8): 1214-9, 2015.
Article in English | MEDLINE | ID: mdl-26046815

ABSTRACT

The major known function of cytokines that belong to type I interferons (IFN, including IFNα and IFNß) is to mount the defense against viruses. This function also protects the genetic information of host cells from alterations in the genome elicited by some of these viruses. Furthermore, recent studies demonstrated that IFN also restrict proliferation of damaged cells by inducing cell senescence. Here we investigated the subsequent role of IFN in elimination of the senescent cells. Our studies demonstrate that endogenous IFN produced by already senescent cells contribute to increased expression of the natural killer (NK) receptor ligands, including MIC-A and ULBP2. Furthermore, neutralization of endogenous IFN or genetic ablation of its receptor chain IFNAR1 compromises the recognition of senescent cells and their clearance in vitro and in vivo. We discuss the role of IFN in protecting the multi-cellular host from accumulation of damaged senescent cells and potential significance of this mechanism in human cancers.


Subject(s)
Cellular Senescence , Fibroblasts/pathology , Interferon Type I/physiology , Animals , Cells, Cultured , Cellular Senescence/drug effects , Fibroblasts/drug effects , GPI-Linked Proteins/genetics , Histocompatibility Antigens Class I/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Interferon Type I/pharmacology , Interferon-beta/immunology , Interferon-beta/metabolism , Interferon-beta/pharmacology , Mice, Inbred C57BL , Mice, Mutant Strains , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Progeria/pathology , Receptor, Interferon alpha-beta/genetics , Werner Syndrome/pathology
10.
Cell Rep ; 11(5): 785-797, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921537

ABSTRACT

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/ß-dependent manner, stimulates cell-autonomous IFN-ß expression in response to double-stranded DNA breaks. Cells and tissues with accumulating DNA damage produce endogenous IFN-ß 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.


Subject(s)
Cellular Senescence , DNA Damage , Interferon-beta/metabolism , Animals , Antibodies, Neutralizing/immunology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line , DNA Damage/drug effects , Humans , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/antagonists & inhibitors , Interferon-beta/genetics , Intestinal Mucosa/metabolism , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , RNA Interference , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Telomerase/deficiency , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism
11.
EMBO Mol Med ; 6(3): 384-97, 2014 03.
Article in English | MEDLINE | ID: mdl-24480543

ABSTRACT

Type 1 interferons (IFN) protect the host against viruses by engaging a cognate receptor (consisting of IFNAR1/IFNAR2 chains) and inducing downstream signaling and gene expression. However, inflammatory stimuli can trigger IFNAR1 ubiquitination and downregulation thereby attenuating IFN effects in vitro. The significance of this paradoxical regulation is unknown. Presented here results demonstrate that inability to stimulate IFNAR1 ubiquitination in the Ifnar1(SA) knock-in mice renders them highly susceptible to numerous inflammatory syndromes including acute and chronic pancreatitis, and autoimmune and toxic hepatitis. Ifnar1(SA) mice (or their bone marrow-receiving wild type animals) display persistent immune infiltration of inflamed tissues, extensive damage and gravely inadequate tissue regeneration. Pharmacologic stimulation of IFNAR1 ubiquitination is protective against from toxic hepatitis and fulminant generalized inflammation in wild type but not Ifnar1(SA) mice. These results suggest that endogenous mechanisms that trigger IFNAR1 ubiquitination for limiting the inflammation-induced tissue damage can be purposely mimicked for therapeutic benefits.


Subject(s)
Receptor, Interferon alpha-beta/metabolism , Acute Disease , Animals , Bone Marrow Transplantation , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/surgery , Chemical and Drug Induced Liver Injury/veterinary , Chronic Disease , Female , Gene Knock-In Techniques , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Liver/physiology , Mice , Mice, Inbred C57BL , Pancreas/physiology , Pancreatitis/chemically induced , Pancreatitis/pathology , Pancreatitis/surgery , Receptor, Interferon alpha-beta/genetics , Regeneration , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL