39 results on '"Mukhopadhyay D"'
Search Results
2. Purification and functional characterization of MerD. A coregulator of the mercury resistance operon in gram-negative bacteria.
- Author
-
Mukhopadhyay, D., primary, Yu, H.R., additional, Nucifora, G., additional, and Misra, T.K., additional
- Published
- 1991
- Full Text
- View/download PDF
3. Placental protein 14 induces apoptosis in T cells but not in monocytes.
- Author
-
Mukhopadhyay, D, Sundereshan, S, Rao, C, and Karande, A A
- Abstract
Substantial evidence exists in literature to suggest that placental protein 14 (PP14) (recently renamed glycodelin A), exhibits immunosuppressive properties and is an indispensable macromolecule in the maternal system for the establishment, maintenance, and progression of pregnancy. Though there are several reports substantiating the above, the mechanism of its action at the molecular level has not been elucidated as yet. In this paper we provide data that suggest that amniotic fluid PP14 and recombinant PP14 expressed in Pichia pastoris induce apoptosis in human peripheral blood lymphocytes upon activation, independent of monocytes. That PP14 has a direct apoptotic action on T cells but not on monocytes was also demonstrated by utilizing human cell lines. PP14 was shown to induce apoptosis in the human T cell lines, Jurkat and MOLT-4 cells, but not in the human monocytic cell line, U937.
- Published
- 2001
- Full Text
- View/download PDF
4. Role of protein kinase C isoforms in phorbol ester-induced vascular endothelial growth factor expression in human glioblastoma cells.
- Author
-
Shih, S C, Mullen, A, Abrams, K, Mukhopadhyay, D, and Claffey, K P
- Abstract
Aberrant expression of the potent angiogenic cytokine, vascular endothelial growth factor (VEGF), has been demonstrated to be associated with most human solid tumors. Both transcriptional and post-transcriptional mechanisms have been shown to modulate VEGF expression in a multitude of cell types. Here we report that when protein kinase C (PKC) pathways were activated in human glioblastoma U373 cells by phorbol 12-myristate 13-acetate (PMA), VEGF mRNA expression was up-regulated via a post-transcriptional mRNA stabilization mechanism. PMA treatment exhibited no increase in VEGF-specific transcriptional activation as determined by run-off transcription assays and VEGF promoter-luciferase reporter assays. However, PMA increased VEGF mRNA half-life from 0.8 to 3.6 h which was blocked by PKC inhibitors but not by protein kinase A or cyclic nucleotide-dependent protein kinase inhibitors. When U373 cells were transfected with antisense oligonucleotide sequences to the translation start sites of PKC-alpha, -beta, -gamma, -delta, -epsilon, or -zeta isoforms, both PKC-alpha and -zeta antisense oligonucleotides showed substantial inhibition of PMA-induced VEGF mRNA. In addition, overexpression of PKC-zeta resulted in a strong constitutive up-regulation of VEGF mRNA expression. This study demonstrates for the first time that specific PKC isoforms regulate VEGF mRNA expression through post-transcriptional mechanisms.
- Published
- 1999
5. Activation of Sp1-mediated vascular permeability factor/vascular endothelial growth factor transcription requires specific interaction with protein kinase C zeta.
- Author
-
Pal, S, Claffey, K P, Cohen, H T, and Mukhopadhyay, D
- Abstract
The transcription factor Sp1 is ubiquitously expressed and plays a significant role in the constitutive and induced expression of a variety of mammalian genes and may even contribute to tumorigenesis. Here, we describe a novel pathway whereby Sp1 promotes the transcription of vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), a potent angiogenic factor, by interacting directly and specifically with protein kinase C zeta (PKC zeta) isoform in renal cell carcinoma. PKC zeta binds and phosphorylates the zinc finger region of Sp1. Moreover, in the presence of the wild type von Hippel-Lindau gene product, the interaction of Sp1 with PKC zeta is inhibited, and in this manner steady state levels of Sp1 phosphorylation are decreased significantly. Co-transfection of renal cell carcinoma cells and human fibrosarcoma cells with a plasmid overexpressing PKC zeta and VPF/VEGF promoter luciferase constructs results in activation of Sp1-mediated transcription, whereas expression of a dominant-negative mutant of PKC zeta repressed this activation. Taken together, our results suggest a new pathway of cell signaling through PKC zeta and provide an insight into PKC zeta and Sp1-dependent transcriptional regulation of VPF/VEGF expression and thus tumor angiogenesis.
- Published
- 1998
6. The von Hippel-Lindau gene product inhibits vascular permeability factor/vascular endothelial growth factor expression in renal cell carcinoma by blocking protein kinase C pathways.
- Author
-
Pal, S, Claffey, K P, Dvorak, H F, and Mukhopadhyay, D
- Abstract
Mutation or loss of function of the von Hippel-Lindau (VHL) tumor suppressor gene is regularly found in sporadic renal cell carcinomas (RCC), well vascularized malignant tumors that characteristically overexpress vascular permeability factor/vascular endothelial growth factor (VPF/VEGF). The wild-type VHL (wt-VHL) gene product acts to suppress VPF/VEGF expression, which is overexpressed when wt-VHL is inactive. The present study investigated the pathways by which VHL regulates VPF/VEGF expression. We found that inhibition of protein kinase C (PKC) represses VPF/VEGF expression in RCC cells that regularly overexpress VPF/VEGF. The wt-VHL expressed by stably transfected RCC cells forms cytoplasmic complexes with two specific PKC isoforms, zeta and delta, and prevents their translocation to the cell membrane where they otherwise would engage in signaling steps that lead to VPF/VEGF overexpression. Other experiments implicated mitogen-activated protein kinase (MAPK) phosphorylation as a downstream step in PKC regulation of VPF/VEGF expression. Taken together, these data demonstrate that wt-VHL, by neutralizing PKC isoforms zeta and delta and thereby inhibiting MAPK activation, plays an important role in preventing aberrant VPF/VEGF overexpression and the angiogenesis that results from such overexpression.
- Published
- 1997
7. Modulation of voltage-dependent Ca2+ channels in rabbit colonic smooth muscle cells by c-Src and focal adhesion kinase.
- Author
-
Hu, X Q, Singh, N, Mukhopadhyay, D, and Akbarali, H I
- Abstract
There is emerging evidence indicating that smooth muscle contraction and Ca2+ influx through voltage-dependent L-type Ca2+ channels are regulated by tyrosine kinases; however, the specific kinases involved are largely unknown. In rabbit colonic muscularis mucosae cells, tyrosine-phosphorylated proteins of approximately 60 and 125 kDa were observed in immunoblots using an anti-phosphotyrosine antibody and were identified as c-Src and focal adhesion kinase (FAK) by immunoblotting with specific antibodies. FAK co-immunoprecipitated with c-Src, and the phosphorylation of the c-Src.FAK complex was markedly enhanced by platelet-derived growth factor (PDGF) BB. The presence of activated c-Src in unstimulated cells was identified in cell lysates by immunoblotting with an antibody recognizing the autophosphorylated site (P416Y). In whole-cell patch-clamp studies, intracellular dialysis of a Src substrate peptide and anti-c-Src and anti-FAK antibodies suppressed Ca2+ currents by 60, 62, and 43%, respectively. In contrast, intracellular dialysis of an anti-mouse IgG or anti-Kv1.5 antibody did not inhibit Ca2+ currents. Co-dialysis of anti-c-Src and anti-FAK antibodies inhibited Ca2+ currents (63%) equivalent to dialysis with the anti-c-Src antibody alone. PDGF-BB enhanced Ca2+ currents by 43%, which was abolished by the anti-c-Src and anti-FAK antibodies. Neither the MEK inhibitor PD 098059 nor an anti-Ras antibody inhibited basal Ca2+ currents or PDGF-stimulated Ca2+ currents. The alpha1C subunit of the L-type Ca2+ channel co-immunoprecipitated with anti-c-Src and anti-phosphotyrosine antibodies, indicating direct association of c-Src kinase with the Ca2+ channel. These data suggest that c-Src and FAK, but not the Ras/mitogen-activated protein kinase cascade, modulate basal Ca2+ channel activity and mediate the PDGF-induced enhancement of L-type Ca2+ currents in differentiated smooth muscle cells.
- Published
- 1998
8. YAP/TAZ-mediated regulation of laminin 332 is enabled by β4 integrin repression of ZEB1 to promote ferroptosis resistance.
- Author
-
Goel HL, Karner ER, Kumar A, Mukhopadhyay D, Goel S, and Mercurio AM
- Subjects
- Female, Humans, Breast Neoplasms metabolism, Breast Neoplasms pathology, Breast Neoplasms genetics, Cell Adhesion Molecules metabolism, Cell Adhesion Molecules genetics, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, MicroRNAs metabolism, MicroRNAs genetics, Phosphoproteins metabolism, Phosphoproteins genetics, Trans-Activators metabolism, Trans-Activators genetics, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics, Integrin beta4 metabolism, Integrin beta4 genetics, Kalinin metabolism, Transcriptional Coactivator with PDZ-Binding Motif Proteins metabolism, YAP-Signaling Proteins metabolism, Zinc Finger E-box-Binding Homeobox 1 metabolism, Zinc Finger E-box-Binding Homeobox 1 genetics, Ferroptosis
- Abstract
We are interested in the contribution of integrins and the extracellular matrix to epithelial differentiation in carcinomas. This study was motivated by our finding that the Hippo effectors YAP and TAZ can sustain the expression of laminin 332 (LM332), the predominant ECM ligand for the integrin β4, in breast carcinoma cells with epithelial differentiation. More specifically, we observed that YAP and TAZ regulate the transcription of the LAMC2 subunit of LM332. Given that the β4-LM332 axis is associated with epithelial differentiation and YAP/TAZ have been implicated in carcinoma de-differentiation, we sought to resolve this paradox. Here, we observed that the β4 integrin sustains the expression of miR-200s that target the transcription factor ZEB1 and that ZEB1 has a pivotal role in determining the nature of YAP/TAZ-mediated transcription. In the presence of β4, ZEB1 expression is repressed enabling YAP/TAZ/TEAD-mediated transcription of LAMC2. The absence of β4, however, induces ZEB1, and ZEB1 binds to the LAMC2 promoter to inhibit LAMC2 transcription. YAP/TAZ-mediated regulation of LAMC2 has important functional consequences because we provide evidence that LM332 enables carcinoma cells to resist ferroptosis in concert with the β4 integrin., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Progesterone receptor membrane component 1 (PGRMC1) binds and stabilizes cytochromes P450 through a heme-independent mechanism.
- Author
-
McGuire MR, Mukhopadhyay D, Myers SL, Mosher EP, Brookheart RT, Kammers K, Sehgal A, Selen ES, Wolfgang MJ, Bumpus NN, and Espenshade PJ
- Subjects
- Amino Acid Substitution, Animals, Cytochrome P-450 Enzyme System genetics, Enzyme Stability, HeLa Cells, Heme genetics, Humans, Membrane Proteins genetics, Mice, Mice, Knockout, Mutation, Missense, Receptors, Progesterone genetics, Cytochrome P-450 Enzyme System metabolism, Heme metabolism, Membrane Proteins metabolism, Receptors, Progesterone metabolism
- Abstract
Progesterone receptor membrane component 1 (PGRMC1) is a heme-binding protein implicated in a wide range of cellular functions. We previously showed that PGRMC1 binds to cytochromes P450 in yeast and mammalian cells and supports their activity. Recently, the paralog PGRMC2 was shown to function as a heme chaperone. The extent of PGRMC1 function in cytochrome P450 biology and whether PGRMC1 is also a heme chaperone are unknown. Here, we examined the function of Pgrmc1 in mouse liver using a knockout model and found that Pgrmc1 binds and stabilizes a broad range of cytochromes P450 in a heme-independent manner. Proteomic and transcriptomic studies demonstrated that Pgrmc1 binds more than 13 cytochromes P450 and supports maintenance of cytochrome P450 protein levels posttranscriptionally. In vitro assays confirmed that Pgrmc1 KO livers exhibit reduced cytochrome P450 activity consistent with reduced enzyme levels. Mechanistic studies in cultured cells demonstrated that PGRMC1 stabilizes cytochromes P450 and that binding and stabilization do not require PGRMC1 binding to heme. Importantly, Pgrmc1-dependent stabilization of cytochromes P450 is physiologically relevant, as Pgrmc1 deletion protected mice from acetaminophen-induced liver injury. Finally, evaluation of Y113F mutant Pgrmc1, which lacks the axial heme iron-coordinating hydroxyl group, revealed that proper iron coordination is not required for heme binding, but is required for binding to ferrochelatase, the final enzyme in heme biosynthesis. PGRMC1 was recently identified as the causative mutation in X-linked isolated pediatric cataract formation. Together, these results demonstrate a heme-independent function for PGRMC1 in cytochrome P450 stability that may underlie clinical phenotypes., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
10. Serum lipoprotein-derived fatty acids regulate hypoxia-inducible factor.
- Author
-
Shao W, Hwang J, Liu C, Mukhopadhyay D, Zhao S, Shen MC, Selen ES, Wolfgang MJ, Farber SA, and Espenshade PJ
- Subjects
- Animals, Gene Expression Profiling, Humans, Hydroxylation, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Lipoproteins blood, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction, Zebrafish, Fatty Acids pharmacology, Gene Expression Regulation drug effects, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Lipoproteins chemistry, Oxygen metabolism
- Abstract
Oxygen regulates hypoxia-inducible factor (HIF) transcription factors to control cell metabolism, erythrogenesis, and angiogenesis. Whereas much has been elucidated about how oxygen regulates HIF, whether lipids affect HIF activity is un-known. Here, using cultured cells and two animal models, we demonstrate that lipoprotein-derived fatty acids are an independent regulator of HIF. Decreasing extracellular lipid supply inhibited HIF prolyl hydroxylation, leading to accumulation of the HIFα subunit of these heterodimeric transcription factors comparable with hypoxia with activation of downstream target genes. The addition of fatty acids to culture medium suppressed this signal, which required an intact mitochondrial respiratory chain. Mechanistically, fatty acids and oxygen are distinct signals integrated to control HIF activity. Finally, we observed lipid signaling to HIF and changes in target gene expression in developing zebrafish and adult mice, and this pathway operates in cancer cells from a range of tissues. This study identifies fatty acids as a physiological modulator of HIF, defining a mechanism for lipoprotein regulation that functions in parallel to oxygen., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Shao et al.)
- Published
- 2020
- Full Text
- View/download PDF
11. Helicobacter pylori infection downregulates the DNA glycosylase NEIL2, resulting in increased genome damage and inflammation in gastric epithelial cells.
- Author
-
Sayed IM, Sahan AZ, Venkova T, Chakraborty A, Mukhopadhyay D, Bimczok D, Beswick EJ, Reyes VE, Pinchuk I, Sahoo D, Ghosh P, Hazra TK, and Das S
- Subjects
- Animals, Antigens, Bacterial metabolism, Bacterial Proteins metabolism, DNA Glycosylases genetics, DNA-(Apurinic or Apyrimidinic Site) Lyase genetics, Disease Progression, Gastric Mucosa pathology, Helicobacter Infections microbiology, Helicobacter Infections pathology, Helicobacter pylori metabolism, Humans, Mice, RNA, Messenger genetics, DNA Glycosylases metabolism, DNA-(Apurinic or Apyrimidinic Site) Lyase metabolism, Down-Regulation, Gastric Mucosa metabolism, Genome, Helicobacter Infections metabolism, Helicobacter pylori isolation & purification, Inflammation metabolism
- Abstract
Infection with the Gram-negative, microaerophilic bacterium Helicobacter pylori induces an inflammatory response and oxidative DNA damage in gastric epithelial cells that can lead to gastric cancer (GC). However, the underlying pathogenic mechanism is largely unclear. Here, we report that the suppression of Nei-like DNA glycosylase 2 (NEIL2), a mammalian DNA glycosylase that specifically removes oxidized bases, is one mechanism through which H. pylori infection may fuel the accumulation of DNA damage leading to GC. Using cultured cell lines, gastric biopsy specimens, primary cells, and human enteroid-derived monolayers from healthy human stomach, we show that H. pylori infection greatly reduces NEIL2 expression. The H. pylori infection-induced downregulation of NEIL2 was specific, as Campylobacter jejuni had no such effect. Using gastric organoids isolated from the murine stomach in coculture experiments with live bacteria mimicking the infected stomach lining, we found that H. pylori infection is associated with the production of various inflammatory cytokines. This response was more pronounced in Neil2 knockout (KO) mouse cells than in WT cells, suggesting that NEIL2 suppresses inflammation under physiological conditions. Notably, the H. pylori -infected Neil2- KO murine stomach exhibited more DNA damage than the WT. Furthermore, H. pylori -infected Neil2- KO mice had greater inflammation and more epithelial cell damage. Computational analysis of gene expression profiles of DNA glycosylases in gastric specimens linked the reduced Neil2 level to GC progression. Our results suggest that NEIL2 downregulation is a plausible mechanism by which H. pylori infection impairs DNA damage repair, amplifies the inflammatory response, and initiates GC., Competing Interests: Conflict of interest—All authors declare that they have no conflict of interest., (© 2020 Sayed et al.)
- Published
- 2020
- Full Text
- View/download PDF
12. Protein kinase D up-regulates transcription of VEGF receptor-2 in endothelial cells by suppressing nuclear localization of the transcription factor AP2β.
- Author
-
Wang Y, Hoeppner LH, Angom RS, Wang E, Dutta S, Doeppler HR, Wang F, Shen T, Scarisbrick IA, Guha S, Storz P, Bhattacharya R, and Mukhopadhyay D
- Subjects
- Cell Movement, Cell Proliferation, Gene Knockdown Techniques, HEK293 Cells, Humans, Neovascularization, Physiologic, Promoter Regions, Genetic genetics, Protein Binding, Serine metabolism, Cell Nucleus metabolism, Human Umbilical Vein Endothelial Cells metabolism, Protein Kinase C metabolism, Transcription Factor AP-2 metabolism, Transcription, Genetic, Up-Regulation, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Vascular endothelial growth factor A (VEGF) signals primarily through its cognate receptor VEGF receptor-2 (VEGFR-2) to control vasculogenesis and angiogenesis, key physiological processes in cardiovascular disease and cancer. In human umbilical vein endothelial cells (HUVECs), knockdown of protein kinase D-1 (PKD1) or PKD2 down-regulates VEGFR-2 expression and inhibits VEGF-induced cell proliferation and migration. However, how PKD regulates VEGF signaling is unclear. Previous bioinformatics analyses have identified binding sites for the transcription factor activating enhancer-binding protein 2 (AP2) in the VEGFR-2 promoter. Using ChIP analyses, here we found that PKD knockdown in HUVECs increases binding of AP2β to the VEGFR-2 promoter. Luciferase reporter assays with serial deletions of AP2-binding sites within the VEGFR-2 promoter revealed that its transcriptional activity negatively correlates with the number of these sites. Next we demonstrated that AP2β up-regulation decreases VEGFR-2 expression and that loss of AP2β enhances VEGFR-2 expression in HUVECs. In vivo experiments confirmed increased VEGFR-2 immunostaining in the spinal cord of AP2β knockout mouse embryos. Mechanistically, we observed that PKD phosphorylates AP2β at Ser
258 and Ser277 and suppresses its nuclear accumulation. Inhibition of PKD activity with a pan-PKD inhibitor increased AP2β nuclear localization, and overexpression of both WT and constitutively active PKD1 or PKD2 reduced AP2β nuclear localization through a Ser258 - and Ser277 -dependent mechanism. Furthermore, substitution of Ser277 in AP2β increased its binding to the VEGFR-2 promoter. Our findings uncover evidence of a molecular pathway that regulates VEGFR-2 expression, insights that may shed light on the etiology of diseases associated with aberrant VEGF/VEGFR signaling., (© 2019 Wang et al.)- Published
- 2019
- Full Text
- View/download PDF
13. Exosome Adherence and Internalization by Hepatic Stellate Cells Triggers Sphingosine 1-Phosphate-dependent Migration.
- Author
-
Wang R, Ding Q, Yaqoob U, de Assuncao TM, Verma VK, Hirsova P, Cao S, Mukhopadhyay D, Huebert RC, and Shah VH
- Subjects
- Animals, Cell Movement, Hepatic Stellate Cells metabolism, Humans, Integrins genetics, Integrins metabolism, Liver Cirrhosis genetics, Liver Cirrhosis physiopathology, Mice, Mice, Inbred C57BL, Receptors, Lysosphingolipid genetics, Receptors, Lysosphingolipid metabolism, Sphingosine metabolism, Sphingosine-1-Phosphate Receptors, Exosomes metabolism, Hepatic Stellate Cells cytology, Liver Cirrhosis metabolism, Lysophospholipids metabolism, Sphingosine analogs & derivatives
- Abstract
Exosomes are cell-derived extracellular vesicles thought to promote intercellular communication by delivering specific content to target cells. The aim of this study was to determine whether endothelial cell (EC)-derived exosomes could regulate the phenotype of hepatic stellate cells (HSCs). Initial microarray studies showed that fibroblast growth factor 2 induced a 2.4-fold increase in mRNA levels of sphingosine kinase 1 (SK1). Exosomes derived from an SK1-overexpressing EC line increased HSC migration 3.2-fold. Migration was not conferred by the dominant negative SK1 exosome. Incubation of HSCs with exosomes was also associated with an 8.3-fold increase in phosphorylation of AKT and 2.5-fold increase in migration. Exosomes were found to express the matrix protein and integrin ligand fibronectin (FN) by Western blot analysis and transmission electron microscopy. Blockade of the FN-integrin interaction with a CD29 neutralizing antibody or the RGD peptide attenuated exosome-induced HSC AKT phosphorylation and migration. Inhibition of endocytosis with transfection of dynamin siRNA, the dominant negative dynamin GTPase construct Dyn2K44A, or the pharmacological inhibitor Dynasore significantly attenuated exosome-induced AKT phosphorylation. SK1 levels were increased in serum exosomes derived from mice with experimental liver fibrosis, and SK1 mRNA levels were up-regulated 2.5-fold in human liver cirrhosis patient samples. Finally, S1PR2 inhibition protected mice from CCl4-induced liver fibrosis. Therefore, EC-derived SK1-containing exosomes regulate HSC signaling and migration through FN-integrin-dependent exosome adherence and dynamin-dependent exosome internalization. These findings advance our understanding of EC/HSC cross-talk and identify exosomes as a potential target to attenuate pathobiology signals., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
14. Identification of biochemically distinct properties of the small ubiquitin-related modifier (SUMO) conjugation pathway in Plasmodium falciparum.
- Author
-
Reiter K, Mukhopadhyay D, Zhang H, Boucher LE, Kumar N, Bosch J, and Matunis MJ
- Subjects
- Amino Acid Sequence, Erythrocytes metabolism, Gene Expression Regulation, Green Fluorescent Proteins metabolism, Humans, Microscopy, Fluorescence, Molecular Conformation, Molecular Sequence Data, Oxidative Stress, Protein Binding, Protein Processing, Post-Translational, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Sumoylation, Ubiquitin metabolism, Plasmodium falciparum metabolism, Protozoan Proteins metabolism, Small Ubiquitin-Related Modifier Proteins metabolism, Ubiquitin-Activating Enzymes metabolism, Ubiquitin-Conjugating Enzymes metabolism
- Abstract
Small ubiquitin-related modifiers (SUMOs) are post-translationally conjugated to other proteins and are thereby essential regulators of a wide range of cellular processes. Sumoylation, and enzymes of the sumoylation pathway, are conserved in the malaria causing parasite, Plasmodium falciparum. However, the specific functions of sumoylation in P. falciparum, and the degree of functional conservation between enzymes of the human and P. falciparum sumoylation pathways, have not been characterized. Here, we demonstrate that sumoylation levels peak during midstages of the intra-erythrocyte developmental cycle, concomitant with hemoglobin consumption and elevated oxidative stress. In vitro studies revealed that P. falciparum E1- and E2-conjugating enzymes interact effectively to recognize and modify RanGAP1, a model mammalian SUMO substrate. However, in heterologous reactions, P. falciparum E1 and E2 enzymes failed to interact with cognate human E2 and E1 partners, respectively, to modify RanGAP1. Structural analysis, binding studies, and functional assays revealed divergent amino acid residues within the E1-E2 binding interface that define organism-specific enzyme interactions. Our studies identify sumoylation as a potentially important regulator of oxidative stress response during the P. falciparum intra-erythrocyte developmental cycle, and define E1 and E2 interactions as a promising target for development of parasite-specific inhibitors of sumoylation and parasite replication.
- Published
- 2013
- Full Text
- View/download PDF
15. Endogenous vascular endothelial growth factor-A (VEGF-A) maintains endothelial cell homeostasis by regulating VEGF receptor-2 transcription.
- Author
-
E G, Cao Y, Bhattacharya S, Dutta S, Wang E, and Mukhopadhyay D
- Subjects
- Cells, Cultured, Endothelial Cells cytology, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Humans, Response Elements physiology, Signal Transduction physiology, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor Receptor-2 genetics, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism, Endothelial Cells metabolism, Gene Expression Regulation physiology, Homeostasis physiology, Transcription, Genetic physiology, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor Receptor-2 biosynthesis
- Abstract
Vascular endothelial growth factor A (VEGF-A) is one of the most important factors controlling angiogenesis. Although the functions of exogenous VEGF-A have been widely studied, the roles of endogenous VEGF-A remain unclear. Here we focused on the mechanistic functions of endogenous VEGF-A in endothelial cells. We found that it is complexed with VEGF receptor 2 (VEGFR-2) and maintains a basal expression level for VEGFR-2 and its downstream signaling activation. Endogenous VEGF-A also controls expression of key endothelial specific genes including VEGFR-2, Tie-2, and vascular endothelial cadherin. Of importance, endogenous VEGF-A differs from exogenous VEGF-A by regulating VEGFR-2 transcription through mediation of FoxC2 binding to the FOX:ETS motif, and the complex formed by endogenous VEGF-A with VEGFR-2 is localized within the EEA1 (early endosome antigen 1) endosomal compartment. Taken together, our results emphasize the importance of endogenous VEGF-A in endothelial cells by regulating key vascular proteins and maintaining the endothelial homeostasis.
- Published
- 2012
- Full Text
- View/download PDF
16. Neuropilin-1 mediates divergent R-Smad signaling and the myofibroblast phenotype.
- Author
-
Cao Y, Szabolcs A, Dutta SK, Yaqoob U, Jagavelu K, Wang L, Leof EB, Urrutia RA, Shah VH, and Mukhopadhyay D
- Subjects
- Actins genetics, Actins metabolism, Animals, Cell Line, Transformed, Fibroblasts cytology, Gene Expression Regulation physiology, Hepatic Stellate Cells cytology, Hepatic Stellate Cells metabolism, Humans, Liver metabolism, Mice, Mice, Knockout, Myoblasts cytology, Neuropilin-1 genetics, Phosphorylation physiology, Smad Proteins, Receptor-Regulated genetics, Transforming Growth Factor beta1 metabolism, Fibroblasts metabolism, Myoblasts metabolism, Neuropilin-1 metabolism, Signal Transduction physiology, Smad Proteins, Receptor-Regulated metabolism
- Abstract
The transforming growth factor-beta (TGF-β) superfamily is one of the most diversified cell signaling pathways and regulates many physiological and pathological processes. Recently, neuropilin-1 (NRP-1) was reported to bind and activate the latent form of TGF-β1 (LAP-TGF-β1). We investigated the role of NRP-1 on Smad signaling in stromal fibroblasts upon TGF-β stimulation. Elimination of NRP-1 in stromal fibroblast cell lines increases Smad1/5 phosphorylation and downstream responses as evidenced by up-regulation of inhibitor of differentiation (Id-1). Conversely, NRP-1 loss decreases Smad2/3 phosphorylation and its responses as shown by down-regulation of α-smooth muscle actin (α-SMA) and also cells exhibit more quiescent phenotypes and growth arrest. Moreover, we also observed that NRP-1 expression is increased during the culture activation of hepatic stellate cells (HSCs), a liver resident fibroblast. Taken together, our data suggest that NRP-1 functions as a key determinant of the diverse responses downstream of TGF-β1 that are mediated by distinct Smad proteins and promotes myofibroblast phenotype.
- Published
- 2010
- Full Text
- View/download PDF
17. Von Hippel-Lindau gene product modulates TIS11B expression in renal cell carcinoma: impact on vascular endothelial growth factor expression in hypoxia.
- Author
-
Sinha S, Dutta S, Datta K, Ghosh AK, and Mukhopadhyay D
- Subjects
- Cell Line, Tumor, Down-Regulation, Gene Silencing, Humans, Models, Biological, Oxygen chemistry, RNA Interference, RNA, Messenger metabolism, Butyrate Response Factor 1 physiology, Carcinoma, Renal Cell metabolism, Gene Expression Regulation, Neoplastic, Hypoxia, Liver Neoplasms metabolism, Vascular Endothelial Growth Factor A biosynthesis, Von Hippel-Lindau Tumor Suppressor Protein biosynthesis
- Abstract
TIS11B belongs to a group of RNA-binding proteins (including TIS11/tristetraprolin and TIS11D) that share characteristic tandem CCCH-type zinc-finger domains and can be rapidly induced by multiple stimuli. TIS11B has been shown to regulate vascular endothelial growth factor (VEGF) mRNA stability in adrenocorticotropic hormone-stimulated primary adrenocortical cells. TIS11B has also been documented as a negative regulator of VEGF during development, but nothing has yet been reported in the context of human cancers. The Von Hippel-Lindau (VHL) tumor suppressor protein regulates VEGF gene expression at both the transcriptional and post-transcriptional levels in normoxia. However, whether it can do so in hypoxia is still unclear. Here, we report a unique regulatory function of VHL in VEGF expression in hypoxia that is mediated through modulation of TIS11B protein levels in renal cancer cells. In normoxia, we detected increased expression of the microRNA hsa-miR-29b in the VHL-overexpressing renal cancer cell line 786-O. We also show that this increased expression of hsa-miR-29b decreased TIS11B protein expression by post-transcriptional regulation in normoxia. In contrast, in hypoxia, increased TIS11B expression paralleled an increased TIS11B mRNA stability in VHL-overexpressing 786-O cells. This VHL-mediated TIS11B up-regulation in hypoxia may be important for TIS11B-regulated gene expression: we observed a down-regulation of VEGF mRNA in hypoxia in VHL-overexpressing cells compared with parental 786-O cells, and this effect was reversible by silencing TIS11B expression.
- Published
- 2009
- Full Text
- View/download PDF
18. Regulation of heparan sulfate and chondroitin sulfate glycosaminoglycan biosynthesis by 4-fluoro-glucosamine in murine airway smooth muscle cells.
- Author
-
Nigro J, Wang A, Mukhopadhyay D, Lauer M, Midura RJ, Sackstein R, and Hascall VC
- Subjects
- Acetylglucosamine pharmacology, Animals, Cells, Cultured, Hymecromone analogs & derivatives, Hymecromone metabolism, Hymecromone pharmacology, Kinetics, Mice, Respiratory Muscles cytology, Respiratory Muscles drug effects, Respiratory Muscles metabolism, Trachea cytology, Trachea drug effects, Trachea metabolism, Acetylglucosamine analogs & derivatives, Chondroitin Sulfates biosynthesis, Heparitin Sulfate biosynthesis, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle metabolism
- Abstract
The importance of the pathological changes in proteoglycans has driven the need to study and design novel chemical tools to control proteoglycan synthesis. Accordingly, we tested the fluorinated analogue of glucosamine (4-fluoro-N-acetyl-glucosamine (4-F-GlcNAc)) on the synthesis of heparan sulfate (HS) and chondroitin sulfate (CS) by murine airway smooth muscle (ASM) cells in the presence of radiolabeled metabolic precursors. Secreted and cell-associated CS and HS were assessed for changes in size by Superose 6 chromatography. Treatment of ASM cells with 4-F-GlcNAc (100 microM) reduced the quantity (by 64.1-76.6%) and decreased the size of HS/CS glycosaminoglycans associated with the cell layer (K(av) shifted from 0.30 to 0.45). The quantity of CS secreted into the medium decreased by 65.7-73.0%, and the size showed a K(av) shift from 0.30 to 0.50. Treatment of ASM cells with 45 microM and 179 microM 4-F-GlcNAc in the presence of a stimulator of CS synthesis, 4-methylumbelliferyl-beta-D-xyloside, reduced the amount of the xyloside-CS chains by 65.4 and 87.0%, respectively. The size of xyloside-CS chains synthesized in the presence of 4-F-GlcNAc were only slightly larger than those with xyloside treatment alone (K(av) of 0.55 compared with that of 0.6). The effects of 4-F-GlcNAc to inhibit CS synthesis were not observed with equimolar concentrations of glucosamine. We propose that 4-F-GlcNAc inhibits CS synthesis by inhibiting 4-epimerization of UDP-GlcNAc to UDP-GalNAc, thereby depleting one of the substrates required, whereas HS elongation is inhibited by truncation when the nonreducing terminus of the growing chain is capped with 4-F-GlcNAc.
- Published
- 2009
- Full Text
- View/download PDF
19. Airway smooth muscle cells synthesize hyaluronan cable structures independent of inter-alpha-inhibitor heavy chain attachment.
- Author
-
Lauer ME, Fulop C, Mukhopadhyay D, Comhair S, Erzurum SC, and Hascall VC
- Subjects
- Alpha-Globulins metabolism, Animals, Antiviral Agents pharmacology, Cell Adhesion Molecules metabolism, Humans, Mice, Mice, Inbred BALB C, Poly I-C pharmacology, U937 Cells, Hyaluronic Acid biosynthesis, Myocytes, Smooth Muscle metabolism, Respiratory System metabolism
- Abstract
The covalent association of inter-alpha-inhibitor-derived heavy chains (HCs) with hyaluronan was first described in synovial fluid from arthritic patients and later described as a structural and functional component of hyaluronan "cable" structures produced by many different cells and stimuli. HC transfer has been shown to be mediated by the protein product of TSG-6 (tumor necrosis factor-stimulated gene 6). Considering the accumulation of hyaluronan in airways following asthmatic attacks and the subsequent infiltration of leukocytes, we sought to characterize HC substitution of hyaluronan "cables" in primary mouse airway smooth muscle cells (MASM) and primary human airway smooth muscle cells (HASM). We found that cells derived from mice lacking TSG-6 had no defect in hyaluronan production or hyaluronan-mediated leukocyte adhesion when treated with the viral mimic poly(I,C). Functional hyaluronan cables were induced by cycloheximide in the confirmed absence of protein synthesis, with or without simultaneous treatment with poly(I,C). We characterized the species specificity of the antibody other investigators used to describe the HC-hyaluronan complex of hyaluronan cables and found minimal affinity to bovine-derived HCs in contrast to HCs from mouse and human sera. Thus, we cultured MASM and HASM cells in serum from these three sources and analyzed hyaluronan extracts for HCs and other hyaluronan-binding proteins, using parallel cumulus cell-oocyte complex (COC) extracts as positive controls. We conclude that, if hyaluronan cables derived from MASM and HASM cells are substituted with HCs, the amount of substitution is significantly below the limit of detection when compared with COC extracts of similar hyaluronan mass.
- Published
- 2009
- Full Text
- View/download PDF
20. Primary murine airway smooth muscle cells exposed to poly(I,C) or tunicamycin synthesize a leukocyte-adhesive hyaluronan matrix.
- Author
-
Lauer ME, Mukhopadhyay D, Fulop C, de la Motte CA, Majors AK, and Hascall VC
- Subjects
- Animals, Asthma immunology, Asthma metabolism, Cell Adhesion drug effects, Endoplasmic Reticulum immunology, Endoplasmic Reticulum metabolism, Female, Humans, Hyaluronic Acid immunology, Leukocytes immunology, Mice, Mice, Inbred BALB C, Myocytes, Smooth Muscle immunology, RNA, Double-Stranded pharmacology, Respiratory System immunology, U937 Cells, Antiviral Agents pharmacology, Extracellular Matrix metabolism, Hyaluronic Acid biosynthesis, Leukocytes metabolism, Myocytes, Smooth Muscle metabolism, Poly I-C pharmacology, Respiratory System metabolism, Tunicamycin pharmacology
- Abstract
Asthmatic attacks often follow viral infections with subsequent airway smooth muscle cell proliferation and the formation of an abnormal hyaluronan extracellular matrix with infiltrated leukocytes. In this study, we show that murine airway smooth muscle cells (MASM) treated with polyinosinic acid-polycytidylic acid (poly(I,C)), a double-stranded RNA that simulates a viral infection, synthesize an abnormal hyaluronan matrix that binds leukocytes (U937 cells). Synthesis of this matrix is initiated rapidly and accumulates linearly for approximately 10 h, reaching a plateau level approximately 7-fold higher than control cultures. MASM cells treated with tunicamycin, to induce endoplasmic reticulum stress, also rapidly initiate synthesis of the abnormal hyaluronan matrix with linear accumulation for approximately 10 h, but only reach a plateau level approximately 2-fold higher than control cultures. In contrast to poly(I,C), the response to tunicamycin depends on cell density, with pre-confluent cells producing more abnormal matrix per cell. Furthermore, U937 cell adhesion per hyaluronan content is higher in the sparse matrix produced in response to tunicamycin, suggesting that the structure in the poly(I,C)-induced matrix masks potential binding sites. When MASM cells were exposed to tunicamycin and poly(I,C) at the same time, U937 cell adhesion was partially additive, implying that these two toxins stimulate hyaluronan synthesis through two different pathways. We also characterized the size of hyaluronan produced by MASM cells, in response to poly(I,C) and tunicamycin, and we found that it ranges from 1500 to 4000 kDa, the majority of which was approximately 4000 kDa and not different in size than hyaluronan made by untreated cells.
- Published
- 2009
- Full Text
- View/download PDF
21. Differentiated murine airway epithelial cells synthesize a leukocyte-adhesive hyaluronan matrix in response to endoplasmic reticulum stress.
- Author
-
Lauer ME, Erzurum SC, Mukhopadhyay D, Vasanji A, Drazba J, Wang A, Fulop C, and Hascall VC
- Subjects
- Animals, Basement Membrane metabolism, Cell Adhesion, Cell Differentiation, Cell Membrane metabolism, Dimerization, Female, Humans, Mice, Mice, Inbred BALB C, U937 Cells, Vascular Cell Adhesion Molecule-1 metabolism, Endoplasmic Reticulum metabolism, Epithelial Cells metabolism, Hyaluronic Acid chemistry, Leukocytes cytology
- Abstract
In this report, we describe a novel method for culturing murine trachea epithelial cells on a native basement membrane at an air-liquid interface to produce a pseudostratified, differentiated airway epithelium composed of ciliated and nonciliated cells. This model was used to examine hyaluronan synthesis by the airway epithelial cells (AECs) in response to poly(I,C) and tunicamycin. The former induces a response similar to viral infection, and the latter is a bacterial toxin known to induce endoplasmic reticulum (ER) stress. We found significant accumulation of hyaluronan on the apical surface of the AECs in response to ER stress, but, unlike previously reported results with smooth muscle cells, no increase in hyaluronan was observed in response to poly(I,C). Monocytic U937 cells adhered at 4 degrees C to the apical surface of the AECs subjected to ER stress by a mechanism almost entirely mediated by hyaluronan. The U937 cells spontaneously released themselves from the abnormal hyaluronan matrix when their metabolism was restored by shifting the temperature from 4 to 37 degrees C in a custom-made flow chamber. Time lapse confocal microscopy permitted live imaging of this interaction between the U937 cells and the hyaluronan matrix and their subsequent response at 37 degrees C. Within 45 min, we observed dynamic protrusions of the U937 cell plasma membrane into nearby hyaluronan matrix, resulting in the degradation of this matrix. Simultaneously, we observed some reorganization of the hyaluronan matrix, from a generalized, apical distribution to localized regions around the AEC tight junctions. We discuss the implications these results might have for the airway epithelium and its relation to airway inflammation and hyperresponsiveness associated with asthma and other airway diseases.
- Published
- 2008
- Full Text
- View/download PDF
22. Identifying key residues of sphinganine-1-phosphate lyase for function in vivo and in vitro.
- Author
-
Mukhopadhyay D, Howell KS, Riezman H, and Capitani G
- Subjects
- Aldehyde-Lyases genetics, Amino Acid Sequence, Binding Sites, Cell Membrane metabolism, Endoplasmic Reticulum metabolism, Glycosylation, Models, Molecular, Molecular Sequence Data, Mutation genetics, Proteasome Endopeptidase Complex metabolism, Protein Structure, Quaternary, Protein Structure, Tertiary, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Aldehyde-Lyases chemistry, Aldehyde-Lyases metabolism
- Abstract
Sphinganine-1-phosphate lyase (Dpl1p) is a highly conserved enzyme of sphingolipid metabolism that catalyzes the irreversible degradation of sphingoid base phosphates, which are potent signaling molecules. Sphingoid base phosphates play a vital role in cell survival, proliferation, migration, heat stress, and cell wall integrity pathways. Little is known about the structure and regulation of Dpl1p. In this study, we have undertaken a combined computational modeling and mutagenesis approach for structure-function analysis of Dpl1p to discover possible modes of regulation. Our results identify important residues for catalysis in Dpl1p and confirm it as an integral endoplasmic reticulum-resident protein. Results further indicate that Dpl1p is most likely not regulated spatially. Importantly, we demonstrate that Dpl1p exists as an oligomer and that polar residues in its transmembrane domain are required for its full function in vivo but not for its localization or for its catalytic activity in vitro.
- Published
- 2008
- Full Text
- View/download PDF
23. TEDS site phosphorylation of the yeast myosins I is required for ligand-induced but not for constitutive endocytosis of the G protein-coupled receptor Ste2p.
- Author
-
Grosshans BL, Grötsch H, Mukhopadhyay D, Fernández IM, Pfannstiel J, Idrissi FZ, Lechner J, Riezman H, and Geli MI
- Subjects
- Actins chemistry, Binding Sites, Cathepsin A metabolism, Cytoskeleton metabolism, DNA metabolism, Endocytosis, Genotype, Glucocorticoids metabolism, Immunoblotting, Immunoprecipitation, Ligands, Mass Spectrometry, Microscopy, Fluorescence, Models, Biological, Phenotype, Phosphorylation, Plasmids metabolism, Protein Binding, Protein Kinases metabolism, Protein Structure, Tertiary, Receptors, G-Protein-Coupled metabolism, Saccharomyces cerevisiae Proteins metabolism, Serine chemistry, Signal Transduction, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Temperature, Time Factors, cdc42 GTP-Binding Protein metabolism, Myosins chemistry, Receptors, Mating Factor physiology, Saccharomyces cerevisiae Proteins physiology
- Abstract
The yeast myosins I Myo3p and Myo5p have well established functions in the polarization of the actin cytoskeleton and in the endocytic uptake of the G protein-coupled receptor Ste2p. A number of results suggest that phosphorylation of the conserved TEDS serine of the myosin I motor head by the Cdc42p activated p21-activated kinases Ste20p and Cla4p is required for the organization of the actin cytoskeleton. However, the role of this signaling cascade in the endocytic uptake has not been investigated. Interestingly, we find that Myo5p TEDS site phosphorylation is not required for slow, constitutive endocytosis of Ste2p, but it is essential for rapid, ligand-induced internalization of the receptor. Our results strongly suggest that a kinase activates the myosins I to sustain fast endocytic uptake. Surprisingly, however, despite the fact that only p21-activated kinases are known to phosphorylate the conserved TEDS site, we find that these kinases are not essential for ligand-induced internalization of Ste2p. Our observations indicate that a different signaling cascade, involving the yeast homologues of the mammalian PDK1 (3-phosphoinositide-dependent-protein kinase-1), Phk1p and Pkh2p, and serum and glucocorticoid-induced kinase, Ypk1p and Ypk2p, activate Myo3p and Myo5p for their endocytic function.
- Published
- 2006
- Full Text
- View/download PDF
24. Inhibition of vascular permeability factor/vascular endothelial growth factor-mediated angiogenesis by the Kruppel-like factor KLF2.
- Author
-
Bhattacharya R, Senbanerjee S, Lin Z, Mir S, Hamik A, Wang P, Mukherjee P, Mukhopadhyay D, and Jain MK
- Subjects
- Adenoviridae genetics, Animals, Blotting, Western, Calcium metabolism, Cell Nucleus metabolism, Cell Proliferation, Cells, Cultured, Endothelium, Vascular cytology, Green Fluorescent Proteins metabolism, Humans, Inflammation, Kruppel-Like Transcription Factors, Male, Mice, Mice, Nude, Thrombosis, Time Factors, Transfection, Zinc Fingers, Neovascularization, Pathologic, Trans-Activators metabolism, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
The Kruppel-like factor KLF2 was recently identified as a novel regulator of endothelial pro-inflammatory and pro-thrombotic function. Here it is shown that overexpression of KLF2 potently inhibits vascular permeability factor/vascular endothelial growth factor (VEGF-A)-mediated angiogenesis and tissue edema in the nude ear mouse model of angiogenesis. In vitro, KLF2 expression retards VEGF-mediated calcium flux, proliferation and induction of pro-inflammatory factors in endothelial cells. This effect is due to a potent inhibition of VEGFR2/KDR expression and promoter activity. These observations identify KLF2 as a regulator of VEGFR2/KDR and provide a foundation for novel approaches to regulate angiogenesis.
- Published
- 2005
- Full Text
- View/download PDF
25. Characterization of complexes formed between TSG-6 and inter-alpha-inhibitor that act as intermediates in the covalent transfer of heavy chains onto hyaluronan.
- Author
-
Rugg MS, Willis AC, Mukhopadhyay D, Hascall VC, Fries E, Fülöp C, Milner CM, and Day AJ
- Subjects
- Animals, Catalysis, Cell Adhesion Molecules genetics, Cell Line, Chondroitinases and Chondroitin Lyases pharmacology, Drosophila, Humans, Hyaluronic Acid chemistry, In Vitro Techniques, Magnesium metabolism, Manganese metabolism, Mice, Molecular Weight, Protein Binding drug effects, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sodium Hydroxide pharmacology, Alpha-Globulins metabolism, Cell Adhesion Molecules metabolism, Hyaluronic Acid metabolism
- Abstract
The high molecular mass glycosaminoglycan hyaluronan (HA) can become modified by the covalent attachment of heavy chains (HCs) derived from the serum protein inter-alpha-inhibitor (IalphaI), which is composed of three subunits (HC1, HC2 and bikunin) linked together via a chondroitin sulfate moiety. The formation of HC.HA is likely to play an important role in the stabilization of HA-rich extracellular matrices in the context of inflammatory disease (e.g. arthritis) and ovulation. Here, we have characterized the complexes formed in vitro between purified human IalphaI and recombinant human TSG-6 (an inflammation-associated protein implicated previously in this process) and show that these complexes (i.e. TSG-6 x HC1 and TSG-6 x HC2) act as intermediates in the formation of HC x HA. This is likely to involve two transesterification reactions in which an ester bond linking an HC to chondroitin sulfate in intact IalphaI is transferred first onto TSG-6 and then onto HA. The formation of TSG-6 x HC1 and TSG-6 x C2 complexes was accompanied by the production of bikunin x HC2 and bikunin x HC1 by-products, respectively, which were observed to break down, releasing free bikunin and HCs. Both TSG-6 x HC formation and the subsequent HC transfer are metal ion-dependent processes; these reactions have a requirement for either Mg2+ or Mn2+ and are inhibited by Co2+. TSG-6, which is released upon the transfer of HCs from TSG-6 onto HA, was shown to combine with IalphaI to form new TSG-6 x HC complexes and thus be recycled. The finding that TSG-6 acts as cofactor and catalyst in the production of HC x HA complexes has important implications for our understanding of inflammatory and inflammation-like processes.
- Published
- 2005
- Full Text
- View/download PDF
26. The peroxisome proliferator-activated receptor alpha (PPARalpha) agonist ciprofibrate inhibits apolipoprotein B mRNA editing in low density lipoprotein receptor-deficient mice: effects on plasma lipoproteins and the development of atherosclerotic lesions.
- Author
-
Fu T, Mukhopadhyay D, Davidson NO, and Borensztajn J
- Subjects
- Animals, Aorta metabolism, Arteriosclerosis metabolism, Blotting, Western, Cholesterol metabolism, Cytoplasm metabolism, Female, Fibric Acids, Intestinal Mucosa metabolism, Lipid Metabolism, Lipoproteins metabolism, Lipoproteins, LDL metabolism, Liver metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Polyethylene Glycols pharmacology, RNA metabolism, RNA, Messenger metabolism, Time Factors, Triglycerides metabolism, Apolipoproteins B metabolism, Clofibric Acid analogs & derivatives, Clofibric Acid pharmacology, Peroxisome Proliferators pharmacology, RNA Editing, Receptors, Cytoplasmic and Nuclear agonists, Receptors, LDL genetics, Transcription Factors agonists
- Abstract
Low density lipoprotein receptor (LDLR)-deficient mice fed a chow diet have a mild hypercholesterolemia caused by the abnormal accumulation in the plasma of apolipoprotein B (apoB)-100- and apoB-48-carrying intermediate density lipoproteins (IDL) and low density lipoproteins (LDL). Treatment of LDLR-deficient mice with ciprofibrate caused a marked decrease in plasma apoB-48-carrying IDL and LDL but at the same time caused a large accumulation of triglyceride-depleted apoB-100-carrying IDL and LDL, resulting in a significant increase in plasma cholesterol levels. These plasma lipoprotein changes were associated with an increase in the hepatic secretion of apoB-100-carrying very low density lipoproteins (VLDL) and a decrease in the secretion of apoB-48-carrying VLDL, accompanied by a significant decrease in hepatic apoB mRNA editing. Hepatic apobec-1 complementation factor mRNA and protein abundance were significantly decreased, whereas apobec-1 mRNA and protein abundance remained unchanged. No changes in apoB mRNA editing occurred in the intestine of the treated animals. After 150 days of treatment with ciprofibrate, consistent with the increased plasma accumulation of apoB-100-carrying IDL and LDL, the LDLR-deficient mice displayed severe atherosclerotic lesions in the aorta. These findings demonstrate that ciprofibrate treatment decreases hepatic apoB mRNA editing and alters the pattern of hepatic lipoprotein secretion toward apoB-100-associated VLDL, changes that in turn lead to increased atherosclerosis.
- Published
- 2004
- Full Text
- View/download PDF
27. Specificity of the tumor necrosis factor-induced protein 6-mediated heavy chain transfer from inter-alpha-trypsin inhibitor to hyaluronan: implications for the assembly of the cumulus extracellular matrix.
- Author
-
Mukhopadhyay D, Asari A, Rugg MS, Day AJ, and Fülöp C
- Subjects
- Animals, Female, Glycosaminoglycans metabolism, Mice, Alpha-Globulins metabolism, Extracellular Matrix metabolism, Hyaluronic Acid metabolism, Tumor Necrosis Factor-alpha metabolism, Zona Pellucida metabolism
- Abstract
The formation of the hyaluronan-rich cumulus extracellular matrix is crucial for female fertility and accompanied by a transesterification reaction in which the heavy chains (HCs) of inter-alpha-trypsin inhibitor (IalphaI)-related proteins are covalently transferred to hyaluronan. Tumor necrosis factor-induced protein-6 (TNFIP6) is essential for this transfer reaction. Female mice deficient in TNFIP6 are infertile due to the lack of a correctly formed cumulus matrix. In this report, we characterize the specificity of TNFIP6-mediated HC transfer from IalphaI to hyaluronan. Hyaluronan oligosaccharides with eight or more monosaccharide units are potent acceptors in the HC transfer, with longer oligosaccharides being somewhat more efficient. Epimerization of the N-acetyl-glucosamine residues to N-acetyl-galactosamines (i.e. in chondroitin) still allows the HC transfer although at a significantly lower efficiency. Sulfation of the N-acetyl-galactosamines in dermatan-4-sulfate or chondroitin-6-sulfate prevents the HC transfer. Hyaluronan oligosaccharides disperse cumulus cells from expanding cumulus cell-oocyte complexes with the same size specificity as their HC acceptor specificity. This process is accompanied by the loss of hyaluronan-linked HCs from the cumulus matrix and the appearance of oligosaccharide-linked HCs in the culture medium. Chondroitin interferes with the expansion of cumulus cell-oocyte complexes only when added with exogenous TNFIP6 before endogenous hyaluronan synthesis starts, supporting that chondroitin is a weaker HC acceptor than hyaluronan. Our data indicate that TNFIP6-mediated HC transfer to hyaluronan is a prerequisite for the correct cumulus matrix assembly and hyaluronan oligosaccharides and chondroitin interfere with this assembly by capturing the HCs of the IalphaI-related proteins.
- Published
- 2004
- Full Text
- View/download PDF
28. Glycodelin A, not glycodelin S, is apoptotically active. Relevance of sialic acid modification.
- Author
-
Mukhopadhyay D, SundarRaj S, Alok A, and Karande AA
- Subjects
- Adult, Female, Glycodelin, Glycoproteins chemistry, Glycoproteins genetics, Glycosylation, Humans, Immunosuppressive Agents chemistry, Immunosuppressive Agents metabolism, Jurkat Cells, Male, Middle Aged, N-Acetylneuraminic Acid, Pregnancy Proteins chemistry, Pregnancy Proteins genetics, Protein Isoforms metabolism, Semen metabolism, Structure-Activity Relationship, T-Lymphocytes metabolism, T-Lymphocytes pathology, Apoptosis physiology, Glycoproteins metabolism, Pregnancy Proteins metabolism
- Abstract
Glycodelin, previously known as PP14 (placental protein-14), is a kernel lipocalin secreted by the glandular epithelium of the endometrium upon progesterone stimulation and by the seminal vesicles. The isoform of the protein present in female reproductive tissue, glycodelin A (GdA), and the male counterpart, glycodelin S (GdS), have identical amino acid sequences, but strikingly different N-linked glycans. It is well documented in literature that GdA is an immunosuppressive protein, and we have shown that this activity is due to its ability to induce apoptosis in activated T cells. The precise role of GdS in seminal plasma is not known. In this study, we report that GdS is not apoptotically active. We observe that the apoptotic activity requires the presence of sialic acid residues on the complex glycans, as in the case of GdA; however, complex glycans of GdS are non-sialylated. We have expressed the wild-type protein in Pichia pastoris, which does not add sialic acid to the secreted proteins, and confirmed our observations that the protein is apoptotically inactive in the non-sialylated form. Our results indicate that differential glycosylation modulates the function of the different glycodelin isoforms.
- Published
- 2004
- Full Text
- View/download PDF
29. Role of insulin receptor substrates and protein kinase C-zeta in vascular permeability factor/vascular endothelial growth factor expression in pancreatic cancer cells.
- Author
-
Neid M, Datta K, Stephan S, Khanna I, Pal S, Shaw L, White M, and Mukhopadhyay D
- Subjects
- Adenocarcinoma blood supply, Adenocarcinoma genetics, Adenocarcinoma metabolism, Base Sequence, Cell Line, Tumor, DNA, Neoplasm genetics, Feedback, Gene Expression Regulation, Neoplastic, Humans, Insulin Receptor Substrate Proteins, Intracellular Signaling Peptides and Proteins, Neovascularization, Pathologic, Pancreatic Neoplasms blood supply, Signal Transduction, Sp1 Transcription Factor metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Phosphoproteins metabolism, Protein Kinase C metabolism, Vascular Endothelial Growth Factor A genetics
- Abstract
Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), the critical molecule in tumor angiogenesis, is regulated by different stimuli, such as hypoxia and oncogenes, and also by growth factors. Previously we have shown that in AsPC-1 pancreatic adenocarcinoma cells, insulin-like growth factor receptor (IGF-IR) regulates VPF/VEGF expression. Insulin receptor substrate-1 and -2 (IRS-1 and IRS-2), two major downstream molecules of IGF-1R, are known to be important in the genesis of diabetes. In this study, we have defined a new role of IRS in angiogenesis. Both of the IRS proteins modulate VPF/VEGF expression in pancreatic cancer cells by different mechanistic pathways. The Sp1-dependent VPF/VEGF transcription is regulated mainly by IRS-2. Protein kinase C-zeta (PKC-zeta) plays a central role in VPF/VEGF expression and acts as a switching element. Furthermore, we have also demonstrated that the phosphatidylinositol 3-kinase pathway, but not the Ras pathway, is a downstream event of IRS proteins for VPF/VEGF expression in AsPC-1 cells. Interestingly, like renal cancer cells, in AsPC-1 cells PKC-zeta leads to direct Sp1-dependent VPF/VEGF transcription; in addition, it also promotes a negative feedback loop to IRS-2 that decreases the association of IRS-2/IGF-1R and IRS-2/p85. Taken together, our results show that in AsPC-1 pancreatic carcinoma cells, Sp1-dependent VPF/VEGF transcription is controlled by IGF-1R signaling through IRS-2 proteins and modulated by a negative feedback loop of PKC-zeta to IRS-2. Our data also suggest that IRS proteins, which are known to play crucial roles in IGF-1R signaling, are also important mediators for tumor angiogenesis.
- Published
- 2004
- Full Text
- View/download PDF
30. Neuropilin-1-mediated vascular permeability factor/vascular endothelial growth factor-dependent endothelial cell migration.
- Author
-
Wang L, Zeng H, Wang P, Soker S, and Mukhopadhyay D
- Subjects
- Base Sequence, Cell Division, Cells, Cultured, DNA Primers, Humans, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Signal Transduction, Cell Movement physiology, Endothelium, Vascular cytology, Neuropilin-1 physiology, Vascular Endothelial Growth Factor A physiology
- Abstract
Neuropilin-1 (NRP-1) has been found to be expressed by endothelial cells and tumor cells as an isoform-specific receptor for vascular permeability factor/vascular endothelial growth factor (VEGF). Previous studies were mainly focused on the extracellular domain of NRP-1 that can bind to VEGF165 and, thus, enables NRP-1 to act as a co-receptor for VEGF165, which enhances its binding to VEGFR-2 and its bioactivity. However, the exact functional roles and related signaling mechanisms of NRP-1 in angiogenesis are not well understood. In this study we constructed a chimeric receptor, EGNP-1, by fusing the extracellular domain of epidermal growth factor receptor to the transmembrane and intracellular domains of NRP-1 and transduced it into HUVECs with a retroviral expression vector. We observed that NRP-1/EGNP-1 mediates ligand-stimulated migration of human umbilical vein endothelial cells (HUVECs) but not proliferation. Our results show that NRP-1 alone can mediate HUVEC migration through its intracellular domain, and its C-terminal three amino acids (SEA-COOH) are essential for the process. We demonstrate that phosphatidylinositol 3-kinase inhibitor Ly294002 and the p85 dominant negative mutant can block NRP-1-mediated HUVEC migration. NRP-1-mediated migration can be significantly reduced by overexpression of the dominant negative mutant of RhoA (RhoA-19N). In addition, Gq family proteins and Gbetagamma subunits are also required for NRP-1-mediated HUVEC migration. These results show for the first time that NRP-1 can independently promote cell signaling in endothelial cells and also demonstrate the importance of last three amino acids of NRP-1 for its function.
- Published
- 2003
- Full Text
- View/download PDF
31. Heterotrimeric G alpha q/G alpha 11 proteins function upstream of vascular endothelial growth factor (VEGF) receptor-2 (KDR) phosphorylation in vascular permeability factor/VEGF signaling.
- Author
-
Zeng H, Zhao D, Yang S, Datta K, and Mukhopadhyay D
- Subjects
- Calcium metabolism, Cell Division physiology, Cells, Cultured, DNA biosynthesis, Endothelial Growth Factors metabolism, Endothelial Growth Factors pharmacology, Endothelium, Vascular cytology, Extracellular Matrix Proteins metabolism, GTP-Binding Protein alpha Subunits, Gq-G11, Humans, Intercellular Signaling Peptides and Proteins metabolism, Intercellular Signaling Peptides and Proteins pharmacology, Lymphokines metabolism, Lymphokines pharmacology, Mitogen-Activated Protein Kinases metabolism, Phosphorylation, Signal Transduction drug effects, Signal Transduction physiology, Tyrosine metabolism, Umbilical Veins cytology, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factor Receptor-1, Vascular Endothelial Growth Factors, Endothelium, Vascular metabolism, Heterotrimeric GTP-Binding Proteins metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) functions by activating two receptor-tyrosine kinases, Flt-1 (VEGF receptor (VEGFR)-1) and KDR (VEGFR-2), both of which are selectively expressed on primary vascular endothelium. KDR is responsible for VPF/VEGF-stimulated endothelial cell proliferation and migration, whereas Flt-1 down-modulates KDR-mediated endothelial cell proliferation. Our most recent works show that pertussis toxin-sensitive G proteins and Gbetagamma subunits are required for Flt-1-mediated down-regulation of human umbilical vein endothelial cell (HUVEC) proliferation and that Gq/11 proteins are required for KDR-mediated RhoA activation and HUVEC migration. In this study, we demonstrate that Gq/11 proteins are also required for VPF/VEGF-stimulated HUVEC proliferation. Our results further indicate that Gq/11 proteins specifically mediate KDR signaling such as intracellular Ca2+ mobilization rather than Flt-1-induced CDC42 activation and that a Gq/11 antisense oligonucleotide completely inhibits MAPK phosphorylation induced by KDR but has no effect on Flt-1-induced MAPK activation. More importantly, we demonstrate that Gq/11 proteins interact with KDR in vivo, and the interaction of Gq/11 proteins with KDR does not require KDR tyrosine phosphorylation. Surprisingly, the Gq/11 antisense oligonucleotide completely inhibits VPF/VEGF-stimulated KDR phosphorylation. Expression of a constitutively active mutant of G11 but not Gq can cause phosphorylation of KDR and MAPK. In addition, a Gbetagamma minigene, hbetaARK1(495), inhibits VPF/VEGF-stimulated HUVEC proliferation, MAPK phosphorylation, and intracellular Ca2+ mobilization but has no effect on KDR phosphorylation. Taken together, this study demonstrates that Gq/11 proteins mediate KDR tyrosine phosphorylation and KDR-mediated HUVEC proliferation through interaction with KDR.
- Published
- 2003
- Full Text
- View/download PDF
32. KDR stimulates endothelial cell migration through heterotrimeric G protein Gq/11-mediated activation of a small GTPase RhoA.
- Author
-
Zeng H, Zhao D, and Mukhopadhyay D
- Subjects
- Base Sequence, Cells, Cultured, GTP-Binding Protein alpha Subunits, Gq-G11, Genes, Dominant, Humans, Oligonucleotides, Antisense, Recombinant Proteins genetics, Recombinant Proteins metabolism, Tyrosine physiology, Vascular Endothelial Growth Factor Receptor-2 chemistry, rac1 GTP-Binding Protein metabolism, rhoA GTP-Binding Protein genetics, Cell Movement physiology, Endothelium, Vascular cytology, Heterotrimeric GTP-Binding Proteins physiology, Vascular Endothelial Growth Factor Receptor-2 physiology, rhoA GTP-Binding Protein metabolism
- Abstract
Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) functions by activating two receptor tyrosine kinases, Flt-1 (VEGFR-1) and KDR (VEGFR-2), both of which are selectively expressed on the primary vascular endothelium. KDR is responsible for VPF/VEGF-stimulated endothelial cell (EC) proliferation and migration, whereas Flt-1 down-modulates KDR-mediated EC proliferation. Flt-1 mediates down-regulation of EC proliferation through pertussis toxin-sensitive G proteins, betagamma subunits, small GTPase CDC42, and partly by Rac-1. However, the molecular mechanism by which KDR mediates EC migration is not clear yet. Here we show for the first time that activation of RhoA and Rac1 is fully and partially required for KDR-mediated human umbilical vein endothelial cell (HUVEC) migration, respectively, and that CDC42, however, is not involved. Furthermore, overexpression of the RhoA dominant negative mutant RhoA-19N does not affect VPF/VEGF-stimulated KDR phosphorylation, intracellular Ca(2+) mobilization, and mitogen-activated protein kinase phosphorylation. Utilizing the receptor chimeras (EGDR and EGLT) in which the extracellular domain of the epidermal growth factor receptor (EGFR) was fused to the transmembrane domain and the intracellular domains of KDR and Flt-1, respectively, we demonstrate that RhoA activation is mediated by EGDR, not by EGLT, and that EGDR mediates activation of Rac1, not CDC42. Furthermore, the EGDR-mediated RhoA and Rac1 activation is regulated by G proteins Gq/11, Gbetagamma, and phospholipase C independent of phosphatidylinositol 3-kinase and intracellular Ca(2+) mobilization. Interestingly, the RhoA activation can be partially inhibited by overexpression of Rac1-17N, but overexpression of RhoA-19N has no effect on Rac1 activation. Finally, Gq/11 and Gbetagamma subunits are also required for VPF/VEGF-stimulated HUVEC migration. Taken together, our results indicate that KDR stimulates endothelial cell migration through a heterotrimeric G protein Gq/11 and Gbetagamma-mediated RhoA pathway.
- Published
- 2002
- Full Text
- View/download PDF
33. Flt-1-mediated down-regulation of endothelial cell proliferation through pertussis toxin-sensitive G proteins, beta gamma subunits, small GTPase CDC42, and partly by Rac-1.
- Author
-
Zeng H, Zhao D, and Mukhopadhyay D
- Subjects
- Calcium metabolism, Cell Line, Endothelial Growth Factors physiology, Humans, Lymphokines physiology, Mutation, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factor Receptor-1, Vascular Endothelial Growth Factors, cdc42 GTP-Binding Protein genetics, rac1 GTP-Binding Protein genetics, Cell Division physiology, Down-Regulation physiology, Endothelium, Vascular cytology, Extracellular Matrix Proteins physiology, Pertussis Toxin, Virulence Factors, Bordetella pharmacology, cdc42 GTP-Binding Protein physiology, rac1 GTP-Binding Protein physiology
- Abstract
Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) promotes its function primarily by activating two receptor tyrosine kinases, Flt-1 (VEGFR-1) and KDR (VEGFR-2). Recently, it has been shown that KDR is responsible for VPF/VEGF-stimulated endothelial cell (EC) proliferation and migration, whereas Flt-1 activation down-modulates KDR-mediated EC proliferation. Although KDR-mediated EC proliferation and migration have been extensively studied, much less is known about Flt-1-mediated antiproliferation. Here, we demonstrate that Flt-1-mediated antiproliferative activity can be blocked completely by the dominant negative mutant of CDC42 (CDC42-17N) and partially by a Rac1 dominant negative mutant (Rac1-17N) but is not affected by a RhoA dominant negative mutant (RhoA-19N). Both CDC42-17N and Rac1-17N increase the intracellular Ca(2+) mobilization in response to VPF/VEGF but have no effect on KDR and MAPK phosphorylation. Using the chimeric-receptor EGLT in which the extracellular domain of epidermal growth factor receptor was fused to the transmembrane and intracellular domains of Flt-1, we also demonstrate that CDC42 and Rac1 are activated by EGLT. Previously, we showed that phosphatidylinositol 3-kinase is required for Flt-1-mediated antiproliferative activity, but phospholipase C is not required. As expected, CDC42 and Rac1 activation mediated by EGLT can be completely inhibited by PI3K inhibitors, wortmannin and LY294002, and the p85 dominant negative mutant but not by either the phospholipase C inhibitor, or an intracellular Ca(2+) chilator BAPTA/AM. Surprisingly, pertussis toxin and overexpression of the free Gbetagamma-specific sequestering minigene hbetaARK1(495) also inhibit EGLT-mediated CDC42 and Rac1 activation completely. Moreover, pertussis toxin treatment also increases the intracellular Ca(2+) mobilization and inhibits the antiproliferation activity, thus suggesting that pertussis toxin-sensitive G proteins and the Gbetagamma subunits are involved in the signaling pathway of Flt-1 that down-regulates EC proliferation. Taken together, these results further expand our understanding of Flt-1-mediated antiproliferative activity in VPF/VEGF-stimulated endothelium.
- Published
- 2002
- Full Text
- View/download PDF
34. Novel role for RNA-binding protein CUGBP2 in mammalian RNA editing. CUGBP2 modulates C to U editing of apolipoprotein B mRNA by interacting with apobec-1 and ACF, the apobec-1 complementation factor.
- Author
-
Anant S, Henderson JO, Mukhopadhyay D, Navaratnam N, Kennedy S, Min J, and Davidson NO
- Subjects
- APOBEC-1 Deaminase, Active Transport, Cell Nucleus, Animals, Base Sequence, Blotting, Western, CELF1 Protein, Cattle, Cell Line, Cell Nucleus metabolism, Cloning, Molecular, Cytoplasm metabolism, DNA, Complementary metabolism, Dose-Response Relationship, Drug, Glutathione Transferase metabolism, Holoenzymes chemistry, Liver metabolism, Microscopy, Fluorescence, Molecular Sequence Data, Oligonucleotides, Antisense pharmacology, Precipitin Tests, Protein Binding, RNA metabolism, RNA Splicing, Rats, Recombinant Fusion Proteins metabolism, Recombinant Proteins metabolism, S100 Proteins metabolism, Subcellular Fractions metabolism, Transfection, Transgenes, Tumor Cells, Cultured, Ultraviolet Rays, Apolipoproteins B metabolism, Cytidine Deaminase, RNA Editing, RNA, Messenger metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Ribonucleoproteins genetics, Ribonucleoproteins metabolism
- Abstract
Mammalian apolipoprotein B (apoB) mRNA editing is mediated by a multicomponent holoenzyme containing apobec-1 and ACF. We have now identified CUGBP2, a 54-kDa RNA-binding protein, as a component of this holoenzyme. CUGBP2 and ACF co-fractionate in bovine liver S-100 extracts, and addition of recombinant apobec-1 leads to assembly of a holoenzyme. Immunodepletion of CUGBP2 co-precipitates ACF, and these proteins co-localize the nucleus of transfected cells, suggesting that CUGBP2 and ACF are bound in vivo. CUGBP2 binds apoB RNA, specifically an AU-rich sequence located immediately upstream of the edited cytidine. ApoB RNA from McA cells, bound to CUGBP2, was more extensively edited than the unbound fraction. However, addition of recombinant CUGBP2 to a reconstituted system demonstrated a dose-dependent inhibition of C to U RNA editing, which was rescued with either apobec-1 or ACF. Antisense CUGBP2 knockout increased endogenous apoB RNA editing, whereas antisense knockout of either apobec-1 or ACF expression eliminated apoB RNA editing, establishing the absolute requirement of these components of the core enzyme. These data suggest that CUGBP2 plays a role in apoB mRNA editing by forming a regulatory complex with the three components of the minimal editing enzyme, apobec-1, ACF, and apoB RNA.
- Published
- 2001
- Full Text
- View/download PDF
35. Tyrosine residues 951 and 1059 of vascular endothelial growth factor receptor-2 (KDR) are essential for vascular permeability factor/vascular endothelial growth factor-induced endothelium migration and proliferation, respectively.
- Author
-
Zeng H, Sanyal S, and Mukhopadhyay D
- Subjects
- Amino Acid Substitution, Calcium metabolism, Calcium Signaling drug effects, Calcium Signaling physiology, Cell Division drug effects, Cell Division physiology, Cell Movement drug effects, Cell Movement physiology, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Humans, Mutagenesis, Site-Directed, Protein Isoforms pharmacology, Receptor Protein-Tyrosine Kinases genetics, Receptors, Growth Factor genetics, Receptors, Vascular Endothelial Growth Factor, Recombinant Proteins pharmacology, Umbilical Veins, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors pharmacology, Endothelium, Vascular physiology, Lymphokines pharmacology, Receptor Protein-Tyrosine Kinases chemistry, Receptor Protein-Tyrosine Kinases physiology, Receptors, Growth Factor chemistry, Receptors, Growth Factor physiology, Tyrosine
- Abstract
Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) exerts its multiple functions by activating two receptor tyrosine kinases, Flt-1 (VEGFR-1) and KDR (VEGFR-2), both of which are selectively expressed on primary vascular endothelium. To dissect the respective signaling pathways and biological functions mediated by these receptors in primary endothelial cells with two receptors intact, we, recently developed chimeric receptors (EGDR and EGLT) in which the extracellular domain of the epidermal growth factor receptor was fused to the transmembrane domain and intracellular domain of KDR and Flt-1, respectively. With these fusion receptors, we have shown that KDR is solely responsible for VPF/VEGF-induced human umbilical vein endothelial cell (HUVEC) proliferation and migration, whereas Flt-1 showed an inhibitory effect on KDR-mediated proliferation but not migration. To further characterize the VPF/VEGF-stimulated HUVEC proliferation and migration here, we have created several EGDR mutants by site-directed mutagenesis. We show that tyrosine residues 1059 and 951 of KDR are essential for VPF/VEGF-induced HUVEC proliferation and migration, respectively. Furthermore, the mutation of tyrosine 1059 to phenylanaline results in the complete loss of KDR/EGDR-mediated intracellular Ca(2+) mobilization and MAPK phosphorylation, but the mutation of tyrosine 951 to phenylanaline did not affect these events. Our results suggest that KDR mediates different signaling pathways for HUVEC proliferation and migration and, moreover, intracellular Ca(2+) mobilization and MAPK phosphorylation are not essential for VPF/VEGF-induced HUVEC migration.
- Published
- 2001
- Full Text
- View/download PDF
36. Vascular permeability factor (VPF)/vascular endothelial growth factor (VEGF) peceptor-1 down-modulates VPF/VEGF receptor-2-mediated endothelial cell proliferation, but not migration, through phosphatidylinositol 3-kinase-dependent pathways.
- Author
-
Zeng H, Dvorak HF, and Mukhopadhyay D
- Subjects
- Base Sequence, Cell Movement physiology, Cells, Cultured, DNA Primers, Endothelium, Vascular enzymology, Humans, Proto-Oncogene Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Recombinant Fusion Proteins metabolism, Recombinant Proteins metabolism, Vascular Endothelial Growth Factor Receptor-1, Cell Division physiology, Down-Regulation, Endothelium, Vascular cytology, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins physiology, Receptor Protein-Tyrosine Kinases physiology, Receptors, Growth Factor physiology, Signal Transduction physiology
- Abstract
Vascular permeability factor (VPF)/vascular endothelial growth factor (VEGF) achieves its multiple functions by activating two receptor tyrosine kinases, Flt-1 (VEGF receptor-1) and KDR (VEGF receptor-2), both of which are selectively expressed on primary vascular endothelium. To dissect the respective signaling pathways and biological functions mediated by these receptors in primary endothelial cells with these two receptors intact, we developed a chimeric receptor system in which the N terminus of the epidermal growth factor receptor was fused to the transmembrane domain and intracellular domain of KDR (EGDR) and Flt-1 (EGLT). We observed that KDR, but not Flt-1, was responsible for VPF/VEGF-induced human umbilical vein endothelial cell (HUVEC) proliferation and migration. Moreover, Flt-1 showed an inhibitory effect on KDR-mediated proliferation, but not migration. We also demonstrated that the inhibitory function of Flt-1 was mediated through the phosphatidylinositol 3-kinase (PI-3K)-dependent pathway because inhibitors of PI-3K as well as a dominant negative mutant of p85 (PI-3K subunit) reversed the inhibition, whereas a constitutively activated mutant of p110 introduced the inhibition to HUVEC-EGDR. We also observed that, in VPF/VEGF-stimulated HUVECs, the Flt-1/EGLT-mediated down-modulation of KDR/EGDR signaling was at or before intracellular Ca(2+) mobilization, but after KDR/EGDR phosphorylation. By mutational analysis, we further identified that the tyrosine 794 residue of Flt-1 was essential for its antiproliferative effect. Taken together, these studies contribute significantly to our understanding of the signaling pathways and biological functions triggered by KDR and Flt-1 and describe a unique mechanism in which PI-3K acts as a mediator of antiproliferation in primary vascular endothelium.
- Published
- 2001
- Full Text
- View/download PDF
37. Role of protein kinase Czeta in Ras-mediated transcriptional activation of vascular permeability factor/vascular endothelial growth factor expression.
- Author
-
Pal S, Datta K, Khosravi-Far R, and Mukhopadhyay D
- Subjects
- 3-Phosphoinositide-Dependent Protein Kinases, Enzyme Activation, Neovascularization, Pathologic, Oligonucleotides, Antisense, Phosphatidylinositol 3-Kinases metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-raf metabolism, Signal Transduction, Sp1 Transcription Factor metabolism, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors genetics, Lymphokines genetics, Protein Kinase C metabolism, Transcriptional Activation, ras Proteins metabolism
- Abstract
Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF), a multifunctional cytokine, is regulated by different factors including degree of cell differentiation, hypoxia, and certain oncogenes namely, ras and src. The up-regulation of VPF/VEGF expression by Ras has been found to be through both transcription and mRNA stability. The present study investigates a novel pathway whereby Ras promotes the transcription of VPF/VEGF by activating protein kinase Czeta (PKCzeta). The Ras-mediated overexpression of VPF/VEGF was also found to be inhibited by using the antisense or the dominant-negative mutant of PKCzeta. In co-transfection assays, by overexpressing oncogenic Ha-Ras (12 V) and PKCzeta, there was an additive effect up to 4-fold in activation of Sp1-mediated VPF/VEGF transcription. It has been shown through electrophoretic mobility shift assay that Ras promoted the PKCzeta-induced binding of Sp1 to the VPF/VEGF promoter. In the presence of PDK-1, a major activating kinase for PKC, the Ras-mediated activation of VPF/VEGF promoter through PKCzeta was further increased, suggesting that PKCzeta can serve as an effector for both Ras and PDK-1. In other experiments, with the use of a dominant-negative mutant of phosphatidylinositol 3-kinase, the activation of VPF/VEGF promoter through Ras, PDK-1, and PKCzeta was completely repressed, indicating phosphatidylinositol 3-kinase as an important component of this pathway. Taken together, these data elucidate the signaling mechanism of Ras-mediated VPF/VEGF transcriptional activation through PKCzeta and also provide insight into PKCzeta and Sp1-dependent transcriptional regulation of VPF/VEGF.
- Published
- 2001
- Full Text
- View/download PDF
38. Role of protein kinase C isoforms in phorbol ester-induced vascular endothelial growth factor expression in human glioblastoma cells
- Author
-
Shih SC, Mullen A, Abrams K, Mukhopadhyay D, and Claffey KP
- Published
- 2000
39. Inhibition of insulin-like growth factor-I-mediated cell signaling by the von Hippel-Lindau gene product in renal cancer.
- Author
-
Datta K, Nambudripad R, Pal S, Zhou M, Cohen HT, and Mukhopadhyay D
- Subjects
- Amino Acid Sequence, Genes, Tumor Suppressor, Humans, Insulin-Like Growth Factor I metabolism, Isoenzymes chemistry, Isoenzymes metabolism, Models, Molecular, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments pharmacology, Protein Binding drug effects, Protein Kinase C chemistry, Protein Kinase C metabolism, Protein Kinase C-delta, Proteins chemistry, Receptor, IGF Type 1 immunology, Receptor, IGF Type 1 metabolism, Sequence Alignment, Tumor Cells, Cultured, Von Hippel-Lindau Tumor Suppressor Protein, Carcinoma, Renal Cell metabolism, Insulin-Like Growth Factor I antagonists & inhibitors, Kidney Neoplasms metabolism, Ligases, Proteins pharmacology, Signal Transduction, Tumor Suppressor Proteins, Ubiquitin-Protein Ligases
- Abstract
Insulin-like growth factor-I (IGF-I)-mediated signaling is thought to be involved in the regulation of multiple cellular functions in different tumors including renal cell carcinoma (RCC). Blocking IGF-I signaling by any of the several strategies abolishes or delays the progression of a variety of tumors in animal models. Herein, we demonstrate that in RCC cell lines, IGF-I-mediated signaling is found to be inhibited in the presence of wild type von Hippel-Lindau (VHL) tumor suppresser gene. Moreover, molecular modeling and biochemical approaches have revealed that beta-domain of the VHL gene product by interacting directly with protein kinase Cdelta inhibits its association with IGF-IR for downstream signaling. We also demonstrated that RCC has IGF-I-mediated invasive activity where protein kinase Cdelta is an important downstream molecule, and this invasiveness can be blocked by wild type VHL. These experiments thus elucidate a novel tumor suppresser function of VHL with its unique kinase inhibitory domain.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.