Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 19(24): 6802-11, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24048331

ABSTRACT

PURPOSE: The ER chaperone GRP78 translocates to the surface of tumor cells and promotes survival, metastasis, and resistance to therapy. An oncogenic function of cell surface GRP78 has been attributed to the activation of the phosphoinositide 3-kinase (PI3K) pathway. We intend to use a novel anti-GRP78 monoclonal antibody (MAb159) to attenuate PI3K signaling and inhibit tumor growth and metastasis. EXPERIMENTAL DESIGN: MAb159 was characterized biochemically. Antitumor activity was tested in cancer cell culture, tumor xenograft models, tumor metastasis models, and spontaneous tumor models. Cancer cells and tumor tissues were analyzed for PI3K activity. MAb159 was humanized and validated for diagnostic and therapeutic application. RESULTS: MAb159 specifically recognized surface GRP78, triggered GRP78 endocytosis, and localized to tumors but not to normal organs in vivo. MAb159 inhibited tumor cell proliferation and enhanced tumor cell death both in vitro and in vivo. In MAb159-treated tumors, PI3K signaling was inhibited without compensatory MAPK pathway activation. Furthermore, MAb159 halted or reversed tumor progression in the spontaneous PTEN-loss-driven prostate and leukemia tumor models, and inhibited tumor growth and metastasis in xenograft models. Humanized MAb159, which retains high affinity, tumor specific localization, and the antitumor activity, was nontoxic in mice, and had desirable pharmacokinetics. CONCLUSIONS: GRP78-specific antibody MAb159 modulates the PI3K pathway and inhibits tumor growth and metastasis. Humanized MAb159 will enter human trials shortly.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal/administration & dosage , Heat-Shock Proteins/genetics , Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Elafin/genetics , Endoplasmic Reticulum Chaperone BiP , HT29 Cells , Heat-Shock Proteins/immunology , Humans , Mice , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/immunology , Xenograft Model Antitumor Assays
2.
J Am Chem Soc ; 134(14): 6344-53, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22404648

ABSTRACT

Methods to visualize, track, and modify proteins in living cells are central for understanding the spatial and temporal underpinnings of life inside cells. Although fluorescent proteins have proven to be extremely useful for in vivo studies of protein function, their utility is inherently limited because their spectral and structural characteristics are interdependent. These limitations have spurred the creation of alternative approaches for the chemical labeling of proteins. We report in this work the use of fluorescence resonance emission transfer (FRET)-quenched DnaE split inteins for the site-specific labeling and concomitant fluorescence activation of proteins in living cells. We have successfully employed this approach for the site-specific in-cell labeling of the DNA binding domain (DBD) of the transcription factor YY1 using several human cell lines. Moreover, we have shown that this approach can be also used for modifying proteins to control their cellular localization and potentially alter their biological activity.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Inteins , Proteins/chemistry , YY1 Transcription Factor/chemistry , Amino Acid Sequence , Biochemistry/methods , Cell Line , Cell Line, Tumor , DNA/chemistry , DNA Polymerase III/chemistry , Green Fluorescent Proteins/chemistry , HeLa Cells , Humans , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Sequence Homology, Amino Acid
3.
Cancer Res ; 71(8): 2848-57, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21467168

ABSTRACT

Glucose-regulated protein 78 (GRP78)/BiP is a multifunctional protein which plays a major role in endoplasmic reticulum (ER) protein processing, protein quality control, maintaining ER homeostasis, and controlling cell signaling and viability. Previously, using a transgene-induced mammary tumor model, we showed that Grp78 heterozygosity impeded cancer growth through suppression of tumor cell proliferation and promotion of apoptosis and the Grp78(+/-) mice exhibited dramatic reduction (70%) in the microvessel density (MVD) of the endogenous mammary tumors, while having no effect on the MVD of normal organs. This observation suggests that GRP78 may critically regulate the function of the host vasculature within the tumor microenvironment. In this article, we interrogated the role of GRP78 in the tumor microenvironment. In mouse tumor models in which wild-type (WT), syngeneic mammary tumor cells were injected into the host, we showed that Grp78(+/-) mice suppressed tumor growth and angiogenesis during the early phase but not during the late phase of tumor growth. Growth of metastatic lesions of WT, syngeneic melanoma cells in the Grp78(+/-) mice was potently suppressed. We created conditional heterozygous knockout of GRP78 in the host endothelial cells and showed severe reduction of tumor angiogenesis and metastatic growth, with minimal effect on normal tissue MVD. Furthermore, knockdown of GRP78 expression in immortalized human endothelial cells showed that GRP78 is a critical mediator of angiogenesis by regulating cell proliferation, survival, and migration. Our findings suggest that concomitant use of current chemotherapeutic agents and novel therapies against GRP78 may offer a powerful dual approach to arrest cancer initiation, progression, and metastasis.


Subject(s)
Heat-Shock Proteins/genetics , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Animals , Apoptosis/physiology , Cell Growth Processes/physiology , Cell Movement/physiology , Endoplasmic Reticulum Chaperone BiP , Endothelial Cells/cytology , Endothelial Cells/physiology , Female , Heterozygote , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Tumor Microenvironment
4.
PLoS One ; 5(5): e10852, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20520781

ABSTRACT

Glucose-regulated protein 94 (GRP94) is one of the most abundant endoplasmic reticulum (ER) resident proteins and is the ER counterpart of the cytoplasmic heat shock protein 90 (HSP90). GRP94, a component of the GRP78 chaperone system in protein processing, has pro-survival properties with implicated function in cancer progression and autoimmune disease. Previous studies on the loss of GRP94 function showed that it is required for embryonic development, regulation of toll-like receptors and innate immunity of macrophages. Here we report the creation of mouse models targeting exon 2 of the Grp94 allele that allows both traditional and conditional knockout (KO) of Grp94. In this study, we utilized the viable Grp94+/+ and +/- mice, as well as primary mouse embryonic fibroblasts generated from them as experimental tools to study its role in ER chaperone balance and ER stress signaling. Our studies reveal that while Grp94 heterozygosity reduces GRP94 level it does not alter ER chaperone levels or the ER stress response. To study the effect of complete loss of GRP94 function, since homozygous GRP94 KO leads to embryonic lethality, we generated Grp94-/- embryonic stem cells. In contrast to Grp94 heterozygosity, complete knockout of GRP94 leads to compensatory upregulation of the ER chaperones GRP78, calnexin and calreticulin but not protein disulphide isomerase. Unexpectedly, loss of GRP94 leads to significant decrease in the level of ER-stress induced spliced form of XBP-1 protein, a downstream target of the IRE1 signaling pathway. Furthermore, from analysis of microarray database and immunohistochemical staining, we present predictions where GRP94 may play an important role in specific adult organ homeostasis and function.


Subject(s)
Embryonic Development/genetics , Endoplasmic Reticulum/pathology , Gene Targeting , Membrane Glycoproteins/genetics , Mutation/genetics , Signal Transduction/genetics , Stress, Physiological/genetics , Alleles , Alternative Splicing/genetics , Animals , DNA-Binding Proteins/genetics , Embryo Loss/metabolism , Embryo Loss/pathology , Embryonic Stem Cells/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression Profiling , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heterozygote , Homozygote , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Molecular Chaperones/metabolism , Regulatory Factor X Transcription Factors , Suppression, Genetic , Transcription Factors/genetics , Up-Regulation , X-Box Binding Protein 1
5.
Mol Cancer Ther ; 8(5): 1086-94, 2009 May.
Article in English | MEDLINE | ID: mdl-19417144

ABSTRACT

Histone deacetylase (HDAC) inhibitors are emerging as effective therapies in the treatment of cancer, and the role of HDACs in the regulation of promoters is rapidly expanding. GRP78/BiP is a stress inducible endoplasmic reticulum (ER) chaperone with antiapoptotic properties. We present here the mechanism for repression of the Grp78 promoter by HDAC1. Our studies reveal that HDAC inhibitors specifically induce GRP78, and the induction level is amplified by ER stress. Through mutational analysis, we have identified the minimal Grp78 promoter and specific elements responsible for HDAC-mediated repression. We show the involvement of HDAC1 in the negative regulation of the Grp78 promoter not only by its induction in the presence of the HDAC inhibitors trichostatin A and MS-275 but also by exogenous overexpression and small interfering RNA knockdown of specific HDACs. We present the results of chromatin immunoprecipitation analysis that reveals the binding of HDAC1 to the Grp78 promoter before, but not after, ER stress. Furthermore, overexpression of GRP78 confers resistance to HDAC inhibitor-induced apoptosis in cancer cells, and conversely, suppression of GRP78 sensitizes them to HDAC inhibitors. These results define HDAC inhibitors as new agents that up-regulate GRP78 without concomitantly inducing the ER or heat shock stress response, and suppression of GRP78 in tumors may provide a novel, adjunctive option to enhance anticancer therapies that use these compounds.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Animals , Endoplasmic Reticulum Chaperone BiP , Female , Gene Knockdown Techniques , Gene Order , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Xenograft Model Antitumor Assays
6.
Mol Cancer Res ; 6(8): 1268-75, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18708359

ABSTRACT

The tumor vasculature is essential for tumor growth and survival and is a key target for anticancer therapy. Glioblastoma multiforme, the most malignant form of brain tumor, is highly vascular and contains abnormal vessels, unlike blood vessels in normal brain. Previously, we showed that primary cultures of human brain endothelial cells, derived from blood vessels of malignant glioma tissues (TuBEC), are physiologically and functionally different from endothelial cells derived from nonmalignant brain tissues (BEC) and are substantially more resistant to apoptosis. Resistance of TuBEC to a wide range of current anticancer drugs has significant clinical consequences as it represents a major obstacle toward eradication of residual brain tumor. We report here that the endoplasmic reticulum chaperone GRP78/BiP is generally highly elevated in the vasculature derived from human glioma specimens, both in situ in tissue and in vitro in primary cell cultures, compared with minimal GRP78 expression in normal brain tissues and blood vessels. Interestingly, TuBEC constitutively overexpress GRP78 without concomitant induction of other major unfolded protein response targets. Resistance of TuBEC to chemotherapeutic agents such as CPT-11, etoposide, and temozolomide can be overcome by knockdown of GRP78 using small interfering RNA or chemical inhibition of its catalytic site. Conversely, overexpression of GRP78 in BEC rendered these cells resistant to drug treatments. Our findings provide the proof of principle that targeting GRP78 will sensitize the tumor vasculature to chemotherapeutic drugs, thus enhancing the efficacy of these drugs in combination therapy for glioma treatment.


Subject(s)
Brain Neoplasms/pathology , Drug Resistance, Neoplasm , Endothelial Cells/pathology , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Antineoplastic Agents/pharmacology , Brain Neoplasms/blood supply , Brain Neoplasms/enzymology , Caspases/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Death/drug effects , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Chaperone BiP , Endothelial Cells/drug effects , Heat-Shock Proteins/antagonists & inhibitors , Humans , Molecular Chaperones/antagonists & inhibitors , Protein Folding , RNA, Small Interfering/metabolism
7.
Cancer Res ; 68(2): 498-505, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18199545

ABSTRACT

The unfolded protein response (UPR) is an evolutionarily conserved mechanism that activates both proapoptotic and survival pathways to allow eukaryotic cells to adapt to endoplasmic reticulum (ER) stress. Although the UPR has been implicated in tumorigenesis, its precise role in endogenous cancer remains unclear. A major UPR protective response is the induction of the ER chaperone GRP78/BiP, which is expressed at high levels in a variety of tumors and confers drug resistance in both proliferating and dormant cancer cells. To determine the physiologic role of GRP78 in in situ-generated tumor and the consequence of its suppression on normal organs, we used a genetic model of breast cancer in the Grp78 heterozygous mice where GRP78 expression level was reduced by about half, mimicking anti-GRP78 agents that achieve partial suppression of GRP78 expression. Here, we report that Grp78 heterozygosity has no effect on organ development or antibody production but prolongs the latency period and significantly impedes tumor growth. Our results reveal three major mechanisms mediated by GRP78 for cancer progression: enhancement of tumor cell proliferation, protection against apoptosis, and promotion of tumor angiogenesis. Importantly, although partial reduction of GRP78 in the Grp78 heterozygous mice substantially reduces the tumor microvessel density, it has no effect on vasculature of normal organs. Our findings establish that a key UPR target GRP78 is preferably required for pathophysiologic conditions, such as tumor proliferation, survival, and angiogenesis, underscoring its potential value as a novel therapeutic target for dual antitumor and antiangiogenesis activity.


Subject(s)
Cell Proliferation , Heat-Shock Proteins/physiology , Mammary Neoplasms, Experimental/pathology , Molecular Chaperones/physiology , Neovascularization, Pathologic/genetics , Animals , Antibody Formation/genetics , Apoptosis/genetics , Caspases/genetics , Cell Survival , Endoplasmic Reticulum Chaperone BiP , Female , Gene Expression Regulation, Neoplastic , Heat-Shock Proteins/genetics , Heterozygote , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Molecular Chaperones/genetics , Transcription Factor CHOP/genetics , Transgenes/physiology , Tumor Burden/genetics
8.
Ann N Y Acad Sci ; 1113: 58-71, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17483206

ABSTRACT

Stress is the imbalance of homeostasis, which can be sensed even at the subcellular level. The stress-sensing capability of various organelles including the endoplasmic reticulum (ER) has been described. It has become evident that acute or prolonged ER stress plays an important role in many human diseases; especially those involving organs/tissues specialized in protein secretion. This article summarizes the emerging role of ER stress in diverse human pathophysiological conditions such as carcinogenesis and tumor progression, cerebral ischemia, plasma cell maturation and apoptosis, obesity, insulin resistance, and type 2 diabetes. Certain components of the ER stress response machinery are identified as biomarkers of the diseases or as possible targets for therapeutic intervention.


Subject(s)
Endoplasmic Reticulum/physiology , Heat-Shock Response/physiology , Animals , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Heat-Shock Proteins/physiology , Humans , Molecular Chaperones/biosynthesis , Molecular Chaperones/genetics , Molecular Chaperones/physiology
9.
Cancer Res ; 65(13): 5785-91, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15994954

ABSTRACT

Therapeutic targeting of the tumor vasculature that destroys preexisting blood vessels of the tumor and antiangiogenesis therapy capitalize on the requirement of tumor cells on an intact vascular supply for oxygen and nutrients for growth, expansion and metastasis to the distal organs. Whereas these classes of agents show promise in delaying tumor progression, they also create glucose and oxygen deprivation conditions within the tumor that could trigger unintended prosurvival responses. The glucose-regulated protein GRP78, a major endoplasmic reticulum chaperone, is inducible by severe glucose depletion, anoxia, and acidosis. Here we report that in a xenograft model of human breast cancer, treatment with the vascular targeting agent, combretastatin A4P, or the antiangiogenic agent, contortrostatin, promotes transcriptional activation of the Grp78 promoter and elevation of GRP78 protein in surviving tumor cells. We further show that GRP78 is overexpressed in a panel of human breast cancer cells that has developed resistance to a variety of drug treatment regimens. Suppression of GRP78 through the use of lentiviral vector expressing small interfering RNA sensitizes human breast cancer cells to etoposide-mediated cell death. Our studies imply that antivascular and antiangiogenesis therapy that results in severe glucose and oxygen deprivation will induce GRP78 expression that could lead to drug resistance.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Disintegrins/pharmacology , Heat-Shock Proteins/biosynthesis , Molecular Chaperones/biosynthesis , Stilbenes/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Endoplasmic Reticulum Chaperone BiP , Etoposide/pharmacology , Female , Glucose/deficiency , Glucose/metabolism , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Humans , Mice , Mice, Nude , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Neovascularization, Pathologic/drug therapy , Oxygen/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Transcriptional Activation/drug effects , Transfection , Xenograft Model Antitumor Assays
11.
Hum Gene Ther ; 15(6): 553-61, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15212714

ABSTRACT

GRP78 is a stress-inducible chaperone protein with antiapoptotic properties that is overexpressed in transformed cells and cells under glucose starvation, acidosis, and hypoxic conditions that persist in poorly vascularized tumors. Previously we demonstrated that the Grp78 promoter is able to eradicate tumors using murine cells in immunocompetent models by driving expression of the HSV-tk suicide gene. Here, through the use of positron emission tomography (PET) imaging, we provide direct evidence of spontaneous in vivo activation of the HSV-tk suicide gene driven by the Grp78 promoter in growing tumors and its activation by photodynamic therapy (PDT) in a controlled manner. In this report, we evaluated whether this promoter can be applied to human cancer therapy. We observed that the Grp78 promoter, in the context of a retroviral vector, was highly activated by stress and PDT in three different types of human breast carcinomas independent of estrogen receptor and p53. Complete regression of sizable human tumors was observed after prodrug ganciclovir treatment of the xenografts in immunodeficient mice. In addition, the Grp78 promoter-driven suicide gene is strongly expressed in a variety of human tumors, including human osteosarcoma. In contrast, the activity of the murine leukemia virus (MuLV) long-terminal repeat (LTR) promoter varied greatly in different human breast carcinoma cell lines, and in some cases, stress resulted in partial suppression of the LTR promoter activity. In transgenic mouse models, the Grp78 promoter-driven transgene is largely quiescent in major adult organs but highly active in cancer cells and cancer-associated macrophages, which can diffuse to tumor necrotic sites devoid of vascular supply and facilitate cell-based therapy. Thus, transcriptional control through the use of the Grp78 promoter offers multiple novel approaches for human cancer gene therapy.


Subject(s)
Genes, Transgenic, Suicide/genetics , Heat-Shock Proteins/genetics , Mammary Neoplasms, Animal/therapy , Molecular Chaperones/genetics , Osteosarcoma/therapy , Promoter Regions, Genetic/drug effects , Thymidine Kinase/genetics , Animals , Antiviral Agents/therapeutic use , Endoplasmic Reticulum Chaperone BiP , Female , Ganciclovir/therapeutic use , Gene Expression , Genetic Therapy , Glucose/metabolism , Heat-Shock Proteins/metabolism , Humans , Leukemia Virus, Murine/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Nude , Molecular Chaperones/metabolism , Osteosarcoma/pathology , Photochemotherapy , Positron-Emission Tomography , Receptors, Estrogen/metabolism , Simplexvirus/genetics , Stress, Physiological , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
12.
Cancer Res ; 62(24): 7207-12, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12499260

ABSTRACT

A major challenge in treating cancer is the difficulty of bringing therapy to poorly perfused areas of solid tumors, which are often most resistant to chemotherapy and radiation. GRP94 is a chaperone protein localized in the endoplasmic reticulum with antiapoptotic properties. We report here that in vitro the proximal murine grp94 promoter is regulated differently from the hypoxia response element fused to the SV40 minimal promoter, with glucose starvation as an inducer of grp94 but a potent repressor of the hypoxia response element/SV40 fusion promoter. Through the use of transgenic mouse models, we showed that LacZ transgene expression driven by the grp94 promoter was strongly activated not only in spontaneous but also in a variety of chemically induced tumors. We additionally discovered that macrophages in the vicinity of malignant tumor showed a high level of transgene expression, consistent with intense beta-galactosidase staining at boundaries between viable tumor cells and necrotic areas. Isolated macrophages also showed grp94 mRNA and transgene activation under glucose starvation in vitro. In contrast, transgene activity was not detected in the normal tissue counterparts of any of the malignant tumors examined or macrophages associated with normal organs. These studies provide direct evidence that the tumor microenvironment is a potent physiological inducer of the grp94 promoter. The unique properties of the grp94 promoter suggest that it may offer a novel tool for directing transcription of therapeutic agents to tumors particularly in resistant regions bordering necrotic areas, delivered through standard vectors or macrophages.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , HSP70 Heat-Shock Proteins/genetics , Macrophages/physiology , Membrane Proteins/genetics , Transgenes/genetics , Animals , Cell Hypoxia/physiology , Cells, Cultured , Female , Glucose/administration & dosage , Humans , Lac Operon , Mice , Molecular Chaperones/genetics , Promoter Regions, Genetic , RNA, Messenger/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...