Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 18(6): 1127-1136, 2019 06.
Article in English | MEDLINE | ID: mdl-30926634

ABSTRACT

Integrin ß1 receptor, expressed on the surface of tumor cells and macrophages in the tumor microenvironment (TME), has been implicated in both tumor progression and resistance to multiple modalities of therapy. OS2966 is the first clinical-ready humanized monoclonal antibody to block integrin ß1 and was recently orphan designated by the FDA Office of Orphan Products Development. Here, we tested therapeutic potential of OS2966-mediated integrin ß1 blockade to enhance the efficacy of oncolytic herpes simplex virus-1 (oHSV) through evaluation of virus replication, tumor cell killing efficiency, effect on the antiviral signaling pathway, co-culture assays of oHSV-infected cells with macrophages, and in vivo bioluminescence imaging on mammary fat pad triple-negative breast cancer xenograft and subcutaneous and intracranial glioma xenografts. OS2966 treatment decreased interferon signaling and proinflammatory cytokine induction in oHSV-treated tumor cells and inhibited migration of macrophages, resulting in enhanced oHSV replication and cytotoxicity. OS2966 treatment also significantly enhanced oHSV replication and oHSV-mediated antitumor efficacy in orthotopic xenograft models, including triple-negative breast cancer and glioblastoma. The results demonstrated the synergistic potential of the combinatory treatment approach with OS2966 to improve antitumor efficacy of conventional oHSV therapy.


Subject(s)
Antibodies, Blocking/therapeutic use , Herpesvirus 1, Human/physiology , Integrin beta1/immunology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Movement/immunology , Coculture Techniques , Combined Modality Therapy/methods , Female , Glioma/metabolism , Glioma/pathology , Glioma/therapy , Humans , Macrophages/metabolism , Mice , Mice, Nude , RAW 264.7 Cells , Virus Replication/immunology , Xenograft Model Antitumor Assays
3.
J Vis Exp ; (117)2016 11 08.
Article in English | MEDLINE | ID: mdl-27911384

ABSTRACT

Elevated lipogenesis is a common characteristic of cancer and metabolic diseases. Sterol regulatory element-binding proteins (SREBPs), a family of membrane-bound transcription factors controlling the expression of genes important for the synthesis of cholesterol, fatty acids and phospholipids, are frequently upregulated in these diseases. In the process of SREBP nuclear translocation, SREBP-cleavage activating protein (SCAP) plays a central role in the trafficking of SREBP from the endoplasmic reticulum (ER) to the Golgi and in subsequent proteolysis activation. Recently, we uncovered that glucose-mediated N-glycosylation of SCAP is a prerequisite condition for the exit of SCAP/SREBP from the ER and movement to the Golgi. N-glycosylation stabilizes SCAP and directs SCAP/SREBP trafficking. Here, we describe a protocol for the isolation of membrane fractions in human cells and for the preparation of the samples for the detection of SCAP N-glycosylation and total protein by using western blot. We further provide a method to monitor SCAP trafficking by using confocal microscopy. This protocol is appropriate for the investigation of SCAP N-glycosylation and trafficking in mammalian cells.


Subject(s)
Cell Culture Techniques , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Cell Membrane , Cholesterol , DNA-Binding Proteins , Endoplasmic Reticulum , Glycosylation , Golgi Apparatus , Humans , Protein Transport , Transcription Factors
4.
Cell Rep ; 16(6): 1527-1535, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27477273

ABSTRACT

Dysregulated lipid metabolism is a characteristic of malignancies. Sterol regulatory element binding protein 1 (SREBP-1), a transcription factor playing a central role in lipid metabolism, is highly activated in malignancies. Here, we unraveled a link between miR-29 and the SCAP (SREBP cleavage-activating protein)/SREBP-1 pathway in glioblastoma (GBM) growth. Epidermal growth factor receptor (EGFR) signaling enhances miR-29 expression in GBM cells via upregulation of SCAP/SREBP-1, and SREBP-1 activates miR-29 expression via binding to specific sites in its promoter. In turn, miR-29 inhibits SCAP and SREBP-1 expression by interacting with their 3' UTRs. miR-29 transfection suppressed lipid synthesis and GBM cell growth, which were rescued by the addition of fatty acids or N-terminal SREBP-1 expression. Xenograft studies showed that miR-29 mimics significantly inhibit GBM growth and prolong the survival of GBM-bearing mice. Our study reveals a previously unrecognized negative feedback loop in SCAP/SREBP-1 signaling mediated by miR-29 and suggests that miR-29 treatment may represent an effective means to target GBM.


Subject(s)
ErbB Receptors/genetics , Glioblastoma , MicroRNAs/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Animals , Feedback , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Lipid Metabolism/genetics , Mice, Nude , Promoter Regions, Genetic/genetics
5.
Clin Cancer Res ; 22(21): 5337-5348, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27281560

ABSTRACT

PURPOSE: Elevated lipogenesis regulated by sterol regulatory element-binding protein-1 (SREBP-1), a transcription factor playing a central role in lipid metabolism, is a novel characteristic of glioblastoma (GBM). The aim of this study was to identify effective approaches to suppress GBM growth by inhibition of SREBP-1. As SREBP activation is negatively regulated by endoplasmic reticulum (ER) cholesterol, we sought to determine whether suppression of sterol O-acyltransferase (SOAT), a key enzyme converting ER cholesterol to cholesterol esters (CE) to store in lipid droplets (LDs), effectively suppressed SREBP-1 and blocked GBM growth. EXPERIMENTAL DESIGN: The presence of LDs in glioma patient tumor tissues was analyzed using immunofluorescence, immunohistochemistry, and electronic microscopy. Western blotting and real-time PCR were performed to analyze protein levels and gene expression of GBM cells, respectively. Intracranial GBM xenografts were used to determine the effects of genetically silencing SOAT1 and SREBP-1 on tumor growth. RESULTS: Our study unraveled that cholesterol esterification and LD formation are signature of GBM, and human patients with glioma possess elevated LDs that correlate with GBM progression and poor survival. We revealed that SOAT1 is highly expressed in GBM and functions as a key player in controlling the cholesterol esterification and storage in GBM. Targeting SOAT1 suppresses GBM growth and prolongs survival in xenograft models via inhibition of SREBP-1-regulated lipid synthesis. CONCLUSIONS: Cholesterol esterification and storage in LDs are novel characteristics of GBM, and inhibiting SOAT1 to block cholesterol esterification is a promising therapeutic strategy to treat GBM by suppressing SREBP-1. Clin Cancer Res; 22(21); 5337-48. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Glioblastoma/drug therapy , Glioblastoma/metabolism , Lipogenesis/drug effects , Sterol O-Acyltransferase/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cholesterol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Female , Gene Expression Regulation/drug effects , Glioma/drug therapy , Glioma/metabolism , Humans , Lipid Metabolism/drug effects , Male , Mice , Mice, Nude , Middle Aged , Young Adult
6.
Cancer Cell ; 28(5): 569-581, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26555173

ABSTRACT

Tumorigenesis is associated with increased glucose consumption and lipogenesis, but how these pathways are interlinked is unclear. Here, we delineate a pathway in which EGFR signaling, by increasing glucose uptake, promotes N-glycosylation of sterol regulatory element-binding protein (SREBP) cleavage-activating protein (SCAP) and consequent activation of SREBP-1, an ER-bound transcription factor with central roles in lipid metabolism. Glycosylation stabilizes SCAP and reduces its association with Insig-1, allowing movement of SCAP/SREBP to the Golgi and consequent proteolytic activation of SREBP. Xenograft studies reveal that blocking SCAP N-glycosylation ameliorates EGFRvIII-driven glioblastoma growth. Thus, SCAP acts as key glucose-responsive protein linking oncogenic signaling and fuel availability to SREBP-dependent lipogenesis. Targeting SCAP N-glycosylation may provide a promising means of treating malignancies and metabolic diseases.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Blotting, Western , Cell Line , Female , Glucose/metabolism , Glucose/pharmacology , Glycosylation/drug effects , Golgi Apparatus/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , MCF-7 Cells , Membrane Proteins/genetics , Mice, Nude , Microscopy, Confocal , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , RNA Interference , Sterol Regulatory Element Binding Protein 1/genetics , Survival Analysis , Transplantation, Heterologous , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...