Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Immunol ; 14: 1256491, 2023.
Article in English | MEDLINE | ID: mdl-38022678

ABSTRACT

Transfer of autologous tumor infiltrating lymphocytes (TIL) to patients with refractory melanoma has shown clinical efficacy in a number of trials. However, extending the clinical benefit to patients with other cancers poses a challenge. Inefficient costimulation in the tumor microenvironment can lead to T cell anergy and exhaustion resulting in poor anti-tumor activity. Here, we describe a chimeric costimulatory antigen receptor (CoStAR) comprised of FRα-specific scFv linked to CD28 and CD40 intracellular signaling domains. CoStAR signaling alone does not activate T cells, while the combination of TCR and CoStAR signaling enhances T cell activity resulting in less differentiated T cells, and augmentation of T cell effector functions, including cytokine secretion and cytotoxicity. CoStAR activity resulted in superior T cell proliferation, even in the absence of exogenous IL-2. Using an in vivo transplantable tumor model, CoStAR was shown to improve T cell survival after transfer, enhanced control of tumor growth, and improved host survival. CoStAR could be reliably engineered into TIL from multiple tumor indications and augmented TIL activity against autologous tumor targets both in vitro and in vivo. CoStAR thus represents a general approach to improving TIL therapy with synthetic costimulation.


Subject(s)
Melanoma , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , CD28 Antigens , Lymphocytes, Tumor-Infiltrating , Folate Receptor 1 , Receptors, Chimeric Antigen/genetics , CD40 Antigens , Tumor Microenvironment
2.
J Transl Med ; 20(1): 426, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36138468

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) are closely related to the occurrence and development of cancer. Abnormally expressed lncRNA can be used as a diagnostic marker for cancer. In this study, we aim to investigate the clinical significance of MIR99AHG expression in lung adenocarcinoma (LUAD), and its biological roles in LUAD progression. METHODS: The relative expression of MIR99AHG in LUAD tissues and cell lines was analyzed using public databases and RT-qPCR. The biological functions of MIR99AHG were investigated using a loss-of-function approach. The effect of MIR99AHG on lung fibrosis was assessed by scratch assay, invasion assay and lung fibrosis rat model. FISH, luciferase reporter assay and immunofluorescence were performed to elucidate the underlying molecular mechanisms. RESULTS: LncRNA MIR99AHG expression level was downregulated in LUAD tissues and cell lines. Low MIR99AHG levels were associated with poorer patient overall survival. Functional analysis showed that MIR99AHG is associated with the LUAD malignant phenotype in vitro and in vivo. Further mechanistic studies showed that, MIR99AHG functions as a competitive endogenous RNA (ceRNA) to antagonize miR-136-5p-mediated ubiquitin specific protease 4 (USP4) degradation, thereby unregulated the expression of angiotensin-converting enzyme 2 (ACE2), a downstream target gene of USP4, which in turn affected alveolar type II epithelial cell fibrosis and epithelial-mesenchymal transition (EMT). In summary, the MIR99AHG/miR-136-5p/USP4/ACE2 signalling axis regulates lung fibrosis and EMT, thus inhibiting LUAD progression. CONCLUSION: This study showed that downregulated MIR99AHG leads to the development of pulmonary fibrosis. Therefore, overexpression of MIR99AHG may provide a new approach to preventing LUAD progression.


Subject(s)
Adenocarcinoma , Lung Neoplasms , MicroRNAs , Pulmonary Fibrosis , RNA, Long Noncoding , Adenocarcinoma/genetics , Angiotensin-Converting Enzyme 2 , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Pulmonary Fibrosis/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Rats , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
3.
Aging (Albany NY) ; 13(10): 13876-13897, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33962392

ABSTRACT

Bladder cancer (BLCA) is one of the common malignant tumors of the urinary system. The poor prognosis of BLCA patients is due to the lack of early diagnosis and disease recurrence after treatment. Increasing evidence suggests that gene products of the nuclear factor of activated T-cells (NFAT) family are involved in BLCA progression and subsequent interaction(s) with immune surveillance. In this study, we carried out a pan-cancer analysis of the NFAT family and found that NFAT2 is an independent prognostic factor for BLCA. We then screened for differentially expressed genes (DEGs) and further analyzed such candidate gene loci using gene ontology enrichment to curate the KEGG database. We then used Lasso and multivariate Cox regression to identify 4 gene loci (FER1L4, RNF128, EPHB6, and FN1) which were screened together with NFAT2 to construct a prognostic model based on using Kaplan-Meier analysis to predict the overall survival of BLCA patients. Moreover, the accuracy of our proposed model is supported by deposited datasets in the Gene Expression Omnibus (GEO) database. Finally, a nomogram of this prognosis model for BLCA was established which could help to provide better disease management and treatment.


Subject(s)
Models, Biological , NFATC Transcription Factors/metabolism , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Kaplan-Meier Estimate , NFATC Transcription Factors/genetics , Oncogenes , Prognosis , Proportional Hazards Models , Protein Interaction Maps/genetics , Reproducibility of Results , Risk Factors , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
4.
Cell Commun Signal ; 17(1): 93, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409371

ABSTRACT

BACKGROUND: Breast cancer is the leading cause of cancer death in women worldwide which is closely related to metastasis. But the exact molecular mechanism of ERα-36 and STAT3 on metastasis is still not fully understood. METHODS: MCF-7 and MDA-MB-231 human breast cancer cell lines and MCF-10A were overexpressioned or knockdown ERα-36 and STAT3 and tested for migration, invasion and proliferation assays. Direct interaction of STAT3 and ERα-36 were analyzed by coimmunoprecipitation assays. The effect of STAT3 and ERα-36 on MMP2/9 expression was analyzed by qPCR and western blotting. STAT3 phospholyation and acetylation by ERα-36 and p300 were observed and quantified by coimmunoprecipitation assays and western blotting. RESULTS: Cross-talk between ERα-36 and STAT3 was demonstrated to mediate through a direct physical association between the two proteins. Furthermore, the interaction between ERα-36 and STAT3 was demonstrated to give rise to functional changes in their signaling events. Both MMP2 and MMP9 expression require the binding of the newly identified protein complex, ERα-36-STAT3, to its promoter, the second phase, which is more robust, depends on ERα-mediated recruitment of p300 onto the complex and the subsequent acetylation of STAT3. In addition, STAT3 is tyrosine-phosphorylated in a biphasic manner, and the late phase requires ERα-36-mediated p300-dependent acetylation. Furthermore, interference with acetylation of STAT3 by overexpression of acetylation null STAT3 mutant led to the loss of MMP2 and MMP9 expression. ChIP analysis and reporter gene assays revealed that ERα-36-STAT3 complex binding to the MMP2 and MMP9 promoter led to an enhanceosome formation and facilitated MMP2 and MMP9 expression. CONCLUSIONS: Our studies demonstrate for the first time that the function of MMP2 and MMP9 in breast cancer cell migration, which is mediated by interactions between ERα-36 and STAT3.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , STAT3 Transcription Factor/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , Humans , MCF-7 Cells , Mutation , STAT3 Transcription Factor/genetics , Signal Transduction
5.
Nature ; 567(7746): 49-55, 2019 03.
Article in English | MEDLINE | ID: mdl-30814735

ABSTRACT

The colonic epithelium facilitates host-microorganism interactions to control mucosal immunity, coordinate nutrient recycling and form a mucus barrier. Breakdown of the epithelial barrier underpins inflammatory bowel disease (IBD). However, the specific contributions of each epithelial-cell subtype to this process are unknown. Here we profile single colonic epithelial cells from patients with IBD and unaffected controls. We identify previously unknown cellular subtypes, including gradients of progenitor cells, colonocytes and goblet cells within intestinal crypts. At the top of the crypts, we find a previously unknown absorptive cell, expressing the proton channel OTOP2 and the satiety peptide uroguanylin, that senses pH and is dysregulated in inflammation and cancer. In IBD, we observe a positional remodelling of goblet cells that coincides with downregulation of WFDC2-an antiprotease molecule that we find to be expressed by goblet cells and that inhibits bacterial growth. In vivo, WFDC2 preserves the integrity of tight junctions between epithelial cells and prevents invasion by commensal bacteria and mucosal inflammation. We delineate markers and transcriptional states, identify a colonic epithelial cell and uncover fundamental determinants of barrier breakdown in IBD.


Subject(s)
Colon/cytology , Colon/pathology , Epithelial Cells/classification , Epithelial Cells/cytology , Health , Inflammatory Bowel Diseases/pathology , Ion Channels/metabolism , Animals , Biomarkers/analysis , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/microbiology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Genetic Predisposition to Disease/genetics , Goblet Cells/cytology , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Hydrogen-Ion Concentration , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Natriuretic Peptides/metabolism , Proteins/metabolism , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/pathology , Tight Junctions/metabolism , Transcription, Genetic , WAP Four-Disulfide Core Domain Protein 2
6.
J Cell Biochem ; 120(4): 6542-6554, 2019 04.
Article in English | MEDLINE | ID: mdl-30368881

ABSTRACT

Tumor cells metabolize more glucose to lactate in aerobic or hypoxic conditions than normal cells. Pyruvate kinase isoenzyme type M2 (PKM2) is crucial for tumor cell aerobic glycolysis. We established a role for let-7a-5p/Stat3/hnRNP-A1/PKM2 signaling in breast cancer cell glucose metabolism. PKM2 depletion via small interfering RNA (siRNA) inhibits cell proliferation and aerobic glycolysis in breast cancer cells. Signal transducer and activator of transcription 3 (Stat3) promotes upregulation of heterogeneous nuclear ribonucleoprotein (hnRNP)-A1 expression, hnRNP-A1 binding to pyruvate kinase isoenzyme (PKM) pre messenger RNA, and the subsequent formation of PKM2. This pathway is downregulated by the microRNA let-7a-5p, which functionally targets Stat3, whereas hnRNP-A1 blocks the biogenesis of let-7a-5p to counteract its ability to downregulate the Stat3/hnRNP-A1/PKM2 signaling pathway. The downregulation of Stat3/hnRNP-A1/PKM2 by let-7a-5p is verified using a breast cancer. These results suggest that let-7a-5p, Stat3, and hnRNP-A1 form a feedback loop, thereby regulating PKM2 expression to modulate glucose metabolism of breast cancer cells. These findings elucidate a new pathway mediating aerobic glycolysis in breast cancers and provide an attractive potential target for breast cancer therapeutic intervention.


Subject(s)
Breast Neoplasms/pathology , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Thyroid Hormones/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Cell Proliferation , Feedback, Physiological , Female , Glycolysis , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Humans , Membrane Proteins/genetics , MicroRNAs/genetics , Prognosis , STAT3 Transcription Factor/genetics , Thyroid Hormones/genetics , Tumor Cells, Cultured , Thyroid Hormone-Binding Proteins
7.
Sci Rep ; 7(1): 16869, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203900

ABSTRACT

Preventing the protein-protein interaction of the cellular chromatin binding protein Lens Epithelium-Derived Growth Factor (LEDGF) and human immunodeficiency virus (HIV) integrase is an important possible strategy for anti-viral treatment for AIDS. We have used Intracellular Antibody Capture technology to isolate a single VH antibody domain that binds to LEDGF. The crystal structure of the LEDGF-VH complex reveals that the single domain antibody mimics the effect of binding of HIV integrase to LEDGF which is crucial for HIV propagation. CD4-expressing T cell lines were constructed to constitutively express the LEDGF-binding VH and these cells showed interference with HIV viral replication, assayed by virus capsid protein p24 production. Therefore, pre-conditioning cells to express antibody fragments confers effective intracellular immunization for preventing chronic viral replication and can be a way to prevent HIV spread in infected patients. This raises the prospect that intracellular immunization strategies that focus on cellular components of viral integrase protein interactions can be used to combat the problems associated with latent HIV virus re-emergence in patients. New genome editing development, such as using CRISPR/cas9, offer the prospect intracellularly immunized T cells in HIV+ patients.


Subject(s)
HIV Infections/pathology , HIV Integrase/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Single-Domain Antibodies/immunology , Amino Acid Sequence , Animals , Binding Sites , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Crystallography, X-Ray , HIV Core Protein p24/metabolism , HIV Infections/immunology , HIV Integrase/chemistry , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Jurkat Cells , Mice , Molecular Dynamics Simulation , Protein Binding , Sequence Alignment , Single-Domain Antibodies/chemistry , Two-Hybrid System Techniques , Virus Replication
8.
Exp Cell Res ; 359(2): 394-404, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28822708

ABSTRACT

Breast cancer is the leading cause of cancer death in women worldwide which is closely related to metastasis. Our previous study has shown that MRTF-A promote the migration of MDA-MB-231 cells and WDR1 promotes breast cancer cell migration. But the exact molecular mechanism on metastasis is still not fully understood, we now report that WDR1 enhanced the effect of MRTF-A induced-MDA-MB-231 cell migration by promoting the expression of the EMT markers and migration markers via RhoA-MRTF-A signaling pathway. Importantly, WDR1 promoted the nuclear importion of MRTF-A by affecting the expression of nuclear transport protein importin. But WDR1 did not affect the expression of MRTF-A. Interestingly, MRTF-A promoted the expression of miR-206 via its promoter CArG box but miR-206 inhibits the migration of breast cancer cells through suppressing the expression of WDR1 and MRTF-A via targeted their 3'UTR. Our data thus provide important and novel insights into MRTF-A-miR-206-WDR1 form feedback loop to regulate breast cancer cell migration.


Subject(s)
Feedback, Physiological , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Microfilament Proteins/genetics , Trans-Activators/genetics , 3' Untranslated Regions , Base Sequence , Binding Sites , Cell Movement , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Female , Humans , Karyopherins/genetics , Karyopherins/metabolism , MCF-7 Cells , MicroRNAs/metabolism , Microfilament Proteins/metabolism , Promoter Regions, Genetic , Signal Transduction , Trans-Activators/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
9.
Gene ; 616: 22-30, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28342807

ABSTRACT

Myocardin is regarded as a key mediator for the change of smooth muscle phenotype. The gap junction protein connexin 43 (Cx43) has been shown to be involved in vascular smooth muscle cells (VSMCs) proliferation and the development of atherosclerosis. However, the role of myocardin on gap junction of cell communication and the relation between myocardin and Cx43 in VSMC phenotypic switch has not been investigated. The goal of the present study is to investigate the molecular mechanism by which myocardin affects Cx43-regulated VSMC proliferation. Data presented in this study demonstrated that inhibition of the Cx43 activation process impaired VSMC proliferation. On the other hand, overexpression miR-206 inhibited VSMC proliferation. In additon, miR-206 silences the expression of Cx43 via targeting Cx43 3' Untranslated Regions. Importantly, myocardin can significantly promote the expression of miR-206. Cx43 regulates VSMCs' proliferation and metastasis through miR-206, which could be promoted by myocardin and used as a marker for diagnosis and a target for therapeutic intervention. Thus myocardin affected the gap junction by inhibited Cx43 and myocardin-miR-206-Cx43 formed a loop to regulate VSMC phenotypic switch.


Subject(s)
Connexin 43/genetics , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , 3' Untranslated Regions , Cell Differentiation , Cell Line , Cell Proliferation , Connexin 43/metabolism , Humans , Muscle, Smooth, Vascular/metabolism , Phenotype
10.
Sci Rep ; 7: 44899, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28322325

ABSTRACT

Haemangioblastoma is a rare malignancy of the CNS where vascular proliferation causes lesions due to endothelial propagation. We found that conditionally expressing mutant Kras, using Rag1-Cre, gave rise to CNS haemangioblastoma in the cortex and cerebellum in mice that present with highly vascular tumours with stromal cells similar to human haemangioblastomas. The aberrant haemangioblastoma endothelial cells do not express mutant Kras but rather the mutant oncogene is expressed in CNS interstitial cells, including neuronal cells and progeny. This demonstrates a non-cell autonomous origin of this disease that is unexpectedly induced via Rag1-Cre expression in CNS interstitial cells. This is the first time that mutant RAS has been shown to stimulate non-cell autonomous proliferation in malignancy and suggests that mutant RAS can control endothelial cell proliferation in neo-vascularisation when expressed in certain cells.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Genes, ras , Hemangioblastoma/genetics , Hemangioblastoma/pathology , Mutation , Animals , Cerebellar Neoplasms/mortality , Disease Models, Animal , Gene Expression , Genes, Reporter , Hemangioblastoma/mortality , Humans , Incidence , Mice , Mice, Transgenic
12.
J Biol Chem ; 290(43): 25907-19, 2015 10 23.
Article in English | MEDLINE | ID: mdl-26370078

ABSTRACT

We have reported previously that a missense mutation in the mitochondrial fission gene Dynamin-related protein 1 (Drp1) underlies the Python mouse model of monogenic dilated cardiomyopathy. The aim of this study was to investigate the consequences of the C452F mutation on Drp1 protein function and to define the cellular sequelae leading to heart failure in the Python monogenic dilated cardiomyopathy model. We found that the C452F mutation increased Drp1 GTPase activity. The mutation also conferred resistance to oligomer disassembly by guanine nucleotides and high ionic strength solutions. In a mouse embryonic fibroblast model, Drp1 C452F cells exhibited abnormal mitochondrial morphology and defective mitophagy. Mitochondria in C452F mouse embryonic fibroblasts were depolarized and had reduced calcium uptake with impaired ATP production by oxidative phosphorylation. In the Python heart, we found a corresponding progressive decline in oxidative phosphorylation with age and activation of sterile inflammation. As a corollary, enhancing autophagy by exposure to a prolonged low-protein diet improved cardiac function in Python mice. In conclusion, failure of Drp1 disassembly impairs mitophagy, leading to a downstream cascade of mitochondrial depolarization, aberrant calcium handling, impaired ATP synthesis, and activation of sterile myocardial inflammation, resulting in heart failure.


Subject(s)
Biopolymers/physiology , Dynamins/physiology , Heart Failure/etiology , Mitophagy , Myocarditis/etiology , Animals , Biopolymers/genetics , Biopolymers/metabolism , Cells, Cultured , Dynamins/genetics , Dynamins/metabolism , Heart Failure/physiopathology , Mice , Mutation , Myocarditis/physiopathology , Oxidative Phosphorylation
13.
J Biol Chem ; 290(32): 19641-52, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26100622

ABSTRACT

The JAK-STAT3 signaling pathway is one of the critical pathways regulating cell proliferation, differentiation, and apoptosis. Myocardin is regarded as a key mediator for the change of smooth muscle phenotypes. However, the relationship between STAT3 and myocardin in the vascular smooth muscle cell (VSMC) phenotypic switch has not been investigated. The goal of this study was to investigate the molecular mechanism by which STAT3 affects the myocardin-regulated VSMC phenotypic switch. Data presented in this study demonstrated that STAT3 was rapidly up-regulated after stimulation with VEGF. Inhibition of the STAT3 activation process impaired VSMC proliferation and enhanced the expression of VSMC contractile genes by increasing serum-response factor binding to the CArG-containing regions of VSMC-specific contractile genes. In contrast, the interaction between serum-response factor and its co-activator myocardin was reduced by overexpression of STAT3. In addition, treated VEGF inhibited the transcription activity of myocardin, and overexpression of STAT3 inhibited myocardin-induced up-regulation of VSMC contractile phenotype-specific genes. Although myocardin and STAT3 are negatively correlated, interestingly, both of them can enhance the expression of VEGF, suggesting a feedback loop to regulate the VSMC phenotypic switch. Taken together, these results indicate that the JAK-STAT3 signaling pathway plays a key role in controlling the phenotypic switch of VSMCs through the interactions between STAT3 and myocardin by various coordinated gene regulation pathways and feedback loops.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Phenotype , STAT3 Transcription Factor/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , Cell Differentiation , Cell Line , Cell Proliferation , Feedback, Physiological , Gene Expression Regulation , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Contraction/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Nuclear Proteins/genetics , STAT3 Transcription Factor/genetics , Serum Response Factor/genetics , Signal Transduction , Trans-Activators/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
14.
PLoS One ; 7(11): e48539, 2012.
Article in English | MEDLINE | ID: mdl-23139789

ABSTRACT

Vascular endothelial growth factor A (VEGF-A) binds to the VEGFR2 receptor tyrosine kinase, regulating endothelial function, vascular physiology and angiogenesis. However, the mechanism underlying VEGFR2 turnover and degradation in this response is unclear. Here, we tested a role for heat-shock proteins in regulating the presentation of VEGFR2 to a degradative pathway. Pharmacological inhibition of HSP90 stimulated VEGFR2 degradation in primary endothelial cells and blocked VEGF-A-stimulated intracellular signaling via VEGFR2. HSP90 inhibition stimulated the formation of a VEGFR2-HSP70 complex. Clathrin-mediated VEGFR2 endocytosis is required for this HSP-linked degradative pathway for targeting VEGFR2 to the endosome-lysosome system. HSP90 perturbation selectively inhibited VEGF-A-stimulated human endothelial cell migration in vitro. A mouse femoral artery model showed that HSP90 inhibition also blocked blood vessel repair in vivo consistent with decreased endothelial regeneration. Depletion of either HSP70 or HSP90 caused defects in blood vessel formation in a transgenic zebrafish model. We conclude that perturbation of the HSP70-HSP90 heat-shock protein axis stimulates degradation of endothelial VEGFR2 and modulates VEGF-A-stimulated intracellular signaling, endothelial cell migration, blood vessel development and repair.


Subject(s)
Blood Vessels/growth & development , Heat-Shock Proteins/metabolism , Neovascularization, Physiologic , Proteolysis , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing , Animals , Arteries/drug effects , Arteries/physiology , Benzoquinones/pharmacology , Blood Vessels/drug effects , Blood Vessels/metabolism , Cell Movement/drug effects , Clathrin/metabolism , Endocytosis/drug effects , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Lactams, Macrocyclic/pharmacology , Mice , Mice, Inbred C57BL , Models, Biological , Neovascularization, Physiologic/drug effects , Protein Stability/drug effects , Protein Transport/drug effects , Proteolysis/drug effects , Regeneration/drug effects , Signal Transduction/drug effects , Wound Healing/drug effects , Zebrafish
15.
FEBS J ; 279(24): 4576-88, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23095053

ABSTRACT

Endothelial cells regulate many aspects of vascular physiology, including vasculogenesis and angiogenesis. The S100 family of calcium-binding proteins regulates many aspects of cell function but their roles in vascular physiology are less well understood. Herein, we investigated the expression and function of S100-related family members in endothelial cells. Analysis of total endothelial mRNAs using a human gene chip array revealed significant gene expression of the S100 calcium-binding protein family members S100A6, S100A10, S100A11 and S100A13. We then examined the expression and functional properties of the major S100 family member, S100A6, in vascular endothelial cells. Comparison of primary and transformed human cells revealed significant differences in S100A6 protein levels in these cells. In primary human endothelial cells, S100A6 was present in both the nucleus and the cytoplasm. To assess the function of endothelial S100A6, we depleted protein levels using RNA interference and this caused increased cell-cycle arrest in the G2/M phase under different conditions. S100A6 depletion caused a decrease in both cyclin-dependent kinase 1 (CDK1) and phospho-CDK1 levels, which are essential for eukaryote cell-cycle progression. S100A6 depletion also decreased expression of CDK1, cyclin A1 (CCNA1) and cyclin B (CCNB1) genes with effects on cell-cycle progression. Depletion of endothelial S100A6 levels also elevated ß-galactosidase expression, which is an important hallmark of cellular senescence and exit from the mammalian cell cycle. We thus propose that S100A6 has an important role in regulating endothelial commitment to, and progression through, the cell cycle.


Subject(s)
Cell Cycle Proteins/physiology , Cell Cycle/physiology , Cellular Senescence/physiology , S100 Proteins/physiology , Cell Cycle Proteins/genetics , DNA Replication/physiology , Humans , RNA, Messenger/genetics , S100 Calcium Binding Protein A6 , S100 Proteins/genetics
16.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 27(2): 395-9, 2010 Apr.
Article in Chinese | MEDLINE | ID: mdl-20481326

ABSTRACT

The epithelial growth factor receptor interference (EGFRi) was obtained by synthetic primers. Overlapping PCR was used to produce EGFRi-IL-24 fusion gene, which is linked by Gly4Ser3. After sequence analysis, EGFRi-IL-24 was cloned into expression vector pPIC9k; EGFRi-IL-24/pPIC9k was linearized with SacI,and then transformed to electroporated pastoris GS115. Subsequently, positive clone was selected by G418 and PCR, and its phenotype was determined by SDS-PAGE and MTT assay. The results demonstrated that EGFRi-IL-24 protein was expressed and shown to have the potential for use in researches of its biological function and in clinical application.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/biosynthesis , Genetic Vectors/genetics , Interleukins/biosynthesis , Antineoplastic Agents/pharmacology , ErbB Receptors/genetics , Humans , Interleukins/genetics , Pichia/genetics , Pichia/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
17.
Biochem Soc Trans ; 37(Pt 6): 1193-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19909245

ABSTRACT

The mammalian endothelium expresses two related but distinct receptor tyrosine kinases, VEGFR1 and VEGFR2 [VEGF (vascular endothelial growth factor) receptor 1 and 2], that regulate the vascular response to a key cytokine, VEGF-A. In the present review, we suggest a model for integrating the signals from these receptor tyrosine kinases by co-ordinating the spatial and temporal segregation of these membrane proteins linked to distinct signalling outputs associated with each intracellular location. Activation of pro-angiogenic VEGFR2 stimulates a programme of tyrosine phosphorylation, ubiquitination and proteolysis. This is linked to ESCRT (endosomal sorting complex required for transport)-mediated recognition of activated VEGFR2 and sorting in endosomes before arrival in lysosomes for terminal degradation. In addition, Rab GTPases regulate key events in VEGFR2 trafficking between the plasma membrane, early and late endosomes, with distinct roles for Rab4a, Rab5a and Rab7a. Manipulation of GTPase levels affects not only VEGFR2 activation and intracellular signalling, but also functional outputs such as VEGF-A-stimulated endothelial cell migration. In contrast, VEGFR1 displays stable Golgi localization that can be perturbed by cell stimuli that elevate cytosolic Ca(2+) ion levels. One model is that VEGFR1 translocates from the trans-Golgi network to the plasma membrane via a calcium-sensitive trafficking step. This allows rapid and preferential sequestration of VEGF-A by the higher-affinity VEGFR1, thus blocking further VEGFR2 activation. Recycling or degradation of VEGFR1 allows resensitization of the VEGFR2-dependent signalling pathway. Thus a dual VEGFR system with a built-in negative-feedback loop is utilized by endothelial cells to sense a key cytokine in vascular tissues.


Subject(s)
Endothelial Cells/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Golgi Apparatus/metabolism , Hydrolysis , Phosphorylation , Protein Transport/physiology , Ubiquitin/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , rab GTP-Binding Proteins/metabolism
18.
Mol Membr Biol ; 26(3): 127-35, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19115141

ABSTRACT

Membrane protein-protein interactions are important for regulation, targeting, and activity of proteins in membranes but are difficult to detect and analyse. This review covers current approaches to studying membrane protein interactions. In addition to standard biochemical and genetic techniques, the classic yeast nuclear two-hybrid system has been highly successful in identification and characterization of soluble protein-protein interactions. However, classic yeast two-hybrid assays do not work for membrane proteins because such yeast-based interactions must occur in the nucleus. Here, we highlight recent advances in yeast systems for the detection and characterization of eukaryote membrane protein-protein interactions. We discuss these implications for drug screening and discovery.


Subject(s)
Membrane Proteins/metabolism , Protein Interaction Mapping/methods , Proteins/metabolism , Protein Binding , Two-Hybrid System Techniques , Yeasts
19.
Cancer Chemother Pharmacol ; 57(2): 248-56, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16028100

ABSTRACT

PURPOSE: To investigate the antitumor effects of tyroserleutide (tyrosyl-seryl-leucine, YSL) on human Bel7402 hepatocarcinoma in vitro and in vivo, with preliminary exploration of its antitumor mechanism. METHODS: MTT was used to observe the anticarcinogenic effects of YSL on human hepatocarcinoma Bel7402 cells in vitro. The ultrastructure of tumor cells was observed by electron microscopy. Nude mice bearing xenografts of human hepatocarcinoma Bel7402 were given daily i.p. injections of YSL or saline and an admixture of amino acids as controls, after tumor implantation. The inhibition of xenografts was determined by calculating the tumor volume and measuring tumor weight. The effects of YSL on the cell cycle and apoptosis of Bel7402 cells were determined by flow cytometry, and the effects on the ultrastructure of the cells by electron microscopy. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and proliferating-cell nuclear antigen (PCNA) immunohistochemical staining were used to investigate apoptosis in tumor tissue in nude mice. RESULTS: In vitro YSL inhibited the proliferation of human Bel7402 tumor cells and changed their ultrastructure, resulting in the necrosis and apoptosis of the tumor cells. YSL at 80, 160, or 320 microg/kg/d inhibited tumor growth in nude mice by 40.26, 64.17, and 59.19%, respectively, which are significantly lower than the inhibition exerted by saline and an admixture of YSL amino acids (P<0.05). The ultrastructure and cell cycle of human hepatocarcinoma Bel7402 cells were changed by treatment with YSL, with a rate of apoptosis higher than that of the control group. TUNEL and PCNA analysis showed that YSL inhibited the proliferation of tumor cells and induced apoptosis at the level of the cell. CONCLUSIONS: YSL significantly inhibited human hepatocarcinoma Bel7402 growth in vitro and in vivo. The growth inhibition of the tumor may involve necrosis and apoptosis of the tumor induced by YSL.


Subject(s)
Carcinoma/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Oligopeptides/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Nude , Necrosis , Transplantation, Heterologous , Tumor Cells, Cultured
20.
Cell Mol Immunol ; 2(5): 365-72, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16368063

ABSTRACT

To determine the regulatory effects of estrogen and cytokine IL-6 and IL-8 on the growth of epithelial ovarian cancer (OVCA), we first examined the status of estrogen receptors (ERalpha and ERbeta ), IL-6 receptor (IL-6Ralpha and gp130), and IL-8 receptor (IL-8RA and IL-8RB) on five epithelial OVCA cell lines by semiquantitative RT-PCR and Western blot analysis. Results showed that the expressions of these receptors were variable on the five cells. Those OVCA cells expressing the receptors were selected to study related molecular mechanism. MTT assay was performed to observe the effects of 17beta-estradiol (E2), IL-6 and IL-8 on cell proliferation. We discovered that E2 markedly promoted the proliferation of CAOV-3 and OVCAR-3 cell in a time- and dose-dependent manner. Tamoxifen (Txf), an ER inhibitor, completely blocked the proliferation of the E2-induced cells, and IL-6- or/and IL-8-neutralizing antibody only showed partially blocking activity. IL-6 and IL-8 were able to significantly stimulate CAOV-3 and OVCAR-3 cell proliferation in a time- and dose-dependent manner, which had a potential synergistic effect on CAOV-3 cells but not on OVCAR-3 cells. The cell proliferation induced by these two cytokines was abolished completely by their specific neutralizing antibodies, partially by Txf, but not by unrelated goat IgG. Taken together, our results suggested that estrogen, IL-6 and IL-8 could modulate OVCA growth by forming a reciprocal cascade with amplifying effect.


Subject(s)
Cell Proliferation/drug effects , Estradiol/pharmacology , Interleukin-6/pharmacology , Interleukin-8/pharmacology , Ovarian Neoplasms/immunology , Cell Line, Tumor , Cytokine Receptor gp130/biosynthesis , Cytokine Receptor gp130/immunology , Dose-Response Relationship, Drug , Epithelial Cells/immunology , Epithelial Cells/pathology , Estradiol/immunology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Interleukin-6/immunology , Interleukin-8/immunology , Ovarian Neoplasms/pathology , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/immunology , Receptors, Interleukin-8A/biosynthesis , Receptors, Interleukin-8A/immunology , Receptors, Interleukin-8B/biosynthesis , Receptors, Interleukin-8B/immunology
SELECTION OF CITATIONS
SEARCH DETAIL