Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Leukemia ; 36(3): 637-647, 2022 03.
Article in English | MEDLINE | ID: mdl-34711926

ABSTRACT

Aberrant RHO guanine nucleotide exchange factor (RhoGEF) activation is chief mechanism driving abnormal activation of their GTPase targets in transformation and tumorigenesis. Consequently, a small-molecule inhibitor of RhoGEF can make an anti-cancer drug. We used cellular, mouse, and humanized models of RAC-dependent BCR-ABL1-driven and Ph-like acute lymphoblastic leukemia to identify VAV3, a tyrosine phosphorylation-dependent RacGEF, as the target of the small molecule IODVA1. We show that through binding to VAV3, IODVA1 inhibits RAC activation and signaling and increases pro-apoptotic activity in BCR-ABL1-transformed cells. Consistent with this mechanism of action, cellular and animal models of BCR-ABL1-induced leukemia in Vav3-null background do not respond to IODVA1. By durably decreasing in vivo RAC signaling, IODVA1 eradicates leukemic propagating activity of TKI-resistant BCR-ABL1(T315I) B-ALL cells after treatment withdrawal. Importantly, IODVA1 suppresses the leukemic burden in the treatment refractory pediatric Ph+ and TKI-resistant Ph+ B-ALL patient-derived xenograft models better than standard-of-care dasatinib or ponatinib and provides a more durable response after treatment withdrawal. Pediatric leukemia samples with diverse genetic lesions show high sensitivity to IODVA1 ex vivo and this sensitivity is VAV3 dependent. IODVA1 thus spearheads a novel class of drugs that inhibits a RacGEF and holds promise as an anti-tumor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Female , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-vav/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Tumor Cells, Cultured
2.
Oncol Rep ; 45(5)2021 05.
Article in English | MEDLINE | ID: mdl-33846812

ABSTRACT

MicroRNA (miR)­29b has been reported to play a controversial role in breast cancer, particularly triple­negative breast cancer (TNBC). Based on our previous data revealing that the PU.1­mediated expression of miR­29b in cells from acute myeloid leukemia is sustained by Vav1, the potential role of this multidomain protein in modulating miR­29b levels in breast tumor cells, in which Vav1 is ecstopically expressed and shows a nuclear accumulation, was investigated. Breast cancer cell lines with various phenotypes and patient­derived xenograft­derived TNBC cells were subjected to Vav1 modulation and reverse transcription quantitative PCR of miR­29b levels. The recruitment of CCAAT enhancer binding protein α (CEBPα) to miR­29b promoters was investigated by quantitative chromatin immunoprecipitation assays. It was found that Vav1 was essential for the recovery of mature miR­29b in breast cancer cell lines, and that it promoted the expression of the miRNA in TNBC cells of the mesenchymal molecular subtype by sustaining the transcription of the miR­29b1/a cluster mediated by CEBPα. The present results suggest that Vav1 is a crucial modulator of miR­29b expression in breast tumor cells, and this finding may help identify strategies that may be useful in the management of TNBC by targeting the Vav1/miR­29b axis, as there is a lack of molecular­based treatments for TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Ductal, Breast/genetics , Proto-Oncogene Proteins c-vav/metabolism , Triple Negative Breast Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Breast/pathology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , MicroRNAs/genetics , Molecular Targeted Therapy/methods , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
mBio ; 11(2)2020 04 21.
Article in English | MEDLINE | ID: mdl-32317318

ABSTRACT

The obligatory intracellular pathogen Ehrlichia chaffeensis lacks most factors that could respond to oxidative stress (a host cell defense mechanism). We previously found that the C terminus of Ehrlichia surface invasin, entry-triggering protein of Ehrlichia (EtpE; EtpE-C) directly binds mammalian DNase X, a glycosylphosphatidylinositol-anchored cell surface receptor and that binding is required to induce bacterial entry and simultaneously to block the generation of reactive oxygen species (ROS) by host monocytes and macrophages. However, how the EtpE-C-DNase X complex mediates the ROS blockade was unknown. A mammalian transmembrane glycoprotein CD147 (basigin) binds to the EtpE-DNase X complex and is required for Ehrlichia entry and infection of host cells. Here, we found that bone marrow-derived macrophages (BMDM) from myeloid cell lineage-selective CD147-null mice had significantly reduced Ehrlichia-induced or EtpE-C-induced blockade of ROS generation in response to phorbol myristate acetate. In BMDM from CD147-null mice, nucleofection with CD147 partially restored the Ehrlichia-mediated inhibition of ROS generation. Indeed, CD147-null mice as well as their BMDM were resistant to Ehrlichia infection. Moreover, in human monocytes, anti-CD147 partially abrogated EtpE-C-induced blockade of ROS generation. Both Ehrlichia and EtpE-C could block activation of the small GTPase Rac1 (which in turn activates phagocyte NADPH oxidase) and suppress activation of Vav1, a hematopoietic-specific Rho/Rac guanine nucleotide exchange factor by phorbol myristate acetate. Vav1 suppression by Ehrlichia was CD147 dependent. E. chaffeensis is the first example of pathogens that block Rac1 activation to colonize macrophages. Furthermore, Ehrlichia uses EtpE to hijack the unique host DNase X-CD147-Vav1 signaling to block Rac1 activation.IMPORTANCEEhrlichia chaffeensis is an obligatory intracellular bacterium with the capability of causing an emerging infectious disease called human monocytic ehrlichiosis. E. chaffeensis preferentially infects monocytes and macrophages, professional phagocytes, equipped with an arsenal of antimicrobial mechanisms, including rapid reactive oxygen species (ROS) generation upon encountering bacteria. As Ehrlichia isolated from host cells are readily killed upon exposure to ROS, Ehrlichia must have evolved a unique mechanism to safely enter phagocytes. We discovered that binding of the Ehrlichia surface invasin to the host cell surface receptor not only triggers Ehrlichia entry but also blocks ROS generation by the host cells by mobilizing a novel intracellular signaling pathway. Knowledge of the mechanisms by which ROS production is inhibited may lead to the development of therapeutics for ehrlichiosis as well as other ROS-related pathologies.


Subject(s)
Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Basigin/metabolism , Ehrlichia chaffeensis/physiology , Macrophages/immunology , Macrophages/metabolism , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Basigin/genetics , Humans , Macrophages/microbiology , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/metabolism
4.
Cancer Lett ; 433: 199-209, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29964204

ABSTRACT

Triple negative breast cancer (TNBC) usually displays more aggressive metastasis, the underlying mechanism is unclear. Previous studies showed that microRNA-205 (miR-205) has controversial roles in cancer, however, its role in TNBC metastasis and the underlying mechanism have not been well-understood. In this study we found that miR-205 expression level is extremely low in basal mesenchymal-like highly migratory and invasive TNBC cells. Stably re-expressing miR-205 in TNBC cells significantly reduced their migration, invasion capability and cancer stem cell (CSC)-like property. Nude mouse orthotopic mammary xenograft tumor model study revealed that miR-205 re-expression greatly decreases TNBC tumor growth and abolishes spontaneous lung metastasis. Mechanistic studies demonstrated that miR-205 inhibits TNBC cell metastatic traits and tumor metastasis by down-regulating integrin α5 (ITGA5). Moreover, ITGA5 knockout using the CRISPR/Cas9 technique achieved the same strong inhibitory effect on TNBC cell CSC-like property and tumor metastasis as re-expressing miR-205 did. Further mechanistic studies indicated that ITGA5 down-regulation by miR-205 re-expression impairs TNBC cell metastatic traits by inhibiting the Src/Vav2/Rac1 pathway. Together, our findings suggest that miR-205 and ITGA5 may serve as potential targets for developing effective therapies for metastatic TNBC.


Subject(s)
Integrins/biosynthesis , MicroRNAs/genetics , Neoplasm Invasiveness/prevention & control , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/pathology , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Movement , Female , Gene Knockout Techniques , Humans , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Transplantation, Heterologous , Triple Negative Breast Neoplasms/genetics , rac1 GTP-Binding Protein/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors
5.
J Cell Biochem ; 119(7): 5437-5448, 2018 07.
Article in English | MEDLINE | ID: mdl-29369410

ABSTRACT

This study aims to explore the effect of miR-330 targeting VAV1 on amyloid ß-protein (Aß) production, oxidative stress (OS), and mitochondrial dysfunction in Alzheimer's disease (AD) mice through the MAPK signaling pathway. Putative targeted gene of miR-330 was performed by a miRNA target prediction website and dual-luciferase reporter gene assay. AD mouse model was successfully established. Fourteen C57 mice were randomized into AD and control groups. The positive protein expression rate of VAV1 was measured by immunohistochemistry. Neuron cells were assigned into control, blank, negative control (NC), miR-330 mimics, miR-330 inhibitors, siRNA-VAV1, and miR-330 inhibitors + siRNA-VAV1 groups. Expression of miR-330, VAV1, ERK1, JNK1, P38MAPK, Aß, COX, and lipoprotein receptor-related protein-1 (LRP-1) were determined using RT-qPCR and Western blotting. Colorimetry was applied to measure the levels of OS parameters of superoxide dismutase (SOD) and malondialdehyde (MDA). Aß production in brain tissue was detected using ELISA, while that in neuron cell was measured by radioimmunoassay. MiR-330 was down-regulated in neuron cells of AD mice and VAV1 was negatively regulated by miR-330. Compared with the control group, the positive protein expression rate of VAV1 was significantly elevated in the AD group. Overexpression of miR-330 decreased the expression of VAV1, ERK1, JNK1, P38MAPK, and Aß, but increased the expression of COX and LRP-1. AD mice revealed elevated Aß production and MDA with decreased SOD level. The result indicates that overexpressed miR-330 targeting VAV1 through the MAPK signaling pathway reduces Aß production and alleviates OS and mitochondrial dysfunction in AD.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , MAP Kinase Signaling System , MicroRNAs/pharmacology , Mitochondria/genetics , Oxidative Stress/genetics , Protective Agents/pharmacology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction
6.
Tumour Biol ; 39(5): 1010428317698392, 2017 May.
Article in English | MEDLINE | ID: mdl-28459214

ABSTRACT

Several studies have proved that Vav2 gene is associated with the carcinogenesis of some tumors, but the relationship between Vav2 gene and gastric cancer remains unclear. Purpose of this study is to detect the expression of Vav2 protein in gastric cancer tissues and to evaluate the clinical value of Vav2. Furthermore, both effect of Vav2 gene on invasion and metastasis of gastric cancer cells and its mechanism are investigated in vitro. Results showed that positive rate of Vav2 protein was significantly higher in gastric cancer tissues than in adjacent tissues and notably higher in metastatic lymph nodes than in gastric cancer tissues. Results of western blot were consistent with immunohistochemistry. Expression of Vav2 protein in gastric cancer tissues was related to degree of tumor differentiation, lymph node metastasis, and clinical stages. Inhibition of endogenous Vav2 in BGC823 cells led to significantly decreased cell activity, migration, and invasion ability in vitro, and expression of Rac1, MMP-2, and MMP-9 decreased, whereas expression of TIMP-1 increased. We concluded that Vav2 might promote invasion and metastasis of gastric cancer by regulating some invasion and metastasis-related genes.


Subject(s)
Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-vav/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Middle Aged , Neoplasm Staging , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Stomach Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , rac1 GTP-Binding Protein/biosynthesis
7.
PLoS One ; 10(12): e0144126, 2015.
Article in English | MEDLINE | ID: mdl-26637171

ABSTRACT

Molecular targeted therapies have been the focus of recent clinical trials for the treatment of patients with recurrent epithelial ovarian cancer (EOC). The majority have not fared well as monotherapies for improving survival of these patients. Poor bioavailability, lack of predictive biomarkers, and the presence of multiple survival pathways can all diminish the success of a targeted agent. Dasatinib is a tyrosine kinase inhibitor of the Src-family kinases (SFK) and in preclinical studies shown to have substantial activity in EOC. However, when evaluated in a phase 2 clinical trial for patients with recurrent or persistent EOC, it was found to have minimal activity. We hypothesized that synthetic lethality screens performed using a cogently designed siRNA library would identify second-site molecular targets that could synergize with SFK inhibition and improve dasatinib efficacy. Using a systematic approach, we performed primary siRNA screening using a library focused on 638 genes corresponding to a network centered on EGFR, HER2, and the SFK-scaffolding proteins BCAR1, NEDD9, and EFS to screen EOC cells in combination with dasatinib. We followed up with validation studies including deconvolution screening, quantitative PCR to confirm effective gene silencing, correlation of gene expression with dasatinib sensitivity, and assessment of the clinical relevance of hits using TCGA ovarian cancer data. A refined list of five candidates (CSNK2A1, DAG1, GRB2, PRKCE, and VAV1) was identified as showing the greatest potential for improving sensitivity to dasatinib in EOC. Of these, CSNK2A1, which codes for the catalytic alpha subunit of protein kinase CK2, was selected for additional evaluation. Synergistic activity of the clinically relevant inhibitor of CK2, CX-4945, with dasatinib in reducing cell proliferation and increasing apoptosis was observed across multiple EOC cell lines. This overall approach to improving drug efficacy can be applied to other targeted agents that have similarly shown poor clinical activity.


Subject(s)
Antineoplastic Agents/pharmacology , Dasatinib/pharmacology , Naphthyridines/pharmacology , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , RNA, Small Interfering/metabolism , Antineoplastic Combined Chemotherapy Protocols , Apoptosis , Carcinoma, Ovarian Epithelial , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Dystroglycans/antagonists & inhibitors , Female , GRB2 Adaptor Protein/antagonists & inhibitors , Gene Library , Humans , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Phenazines , Protein Kinase C-epsilon/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/antagonists & inhibitors
8.
Cancer Res ; 75(14): 2907-15, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25977335

ABSTRACT

Pancreatic cancer, one of the most lethal forms of human cancer, is largely resistant to many conventional chemotherapeutic agents. Although many therapeutic approaches focus on tumor growth, metastasis is a primary factor contributing to lethality. Therefore, novel therapies to target metastatic invasion could prevent tumor spread and recurrence resulting from local and distant metastasis. The protein Vav1 is aberrantly expressed in more than half of pancreatic cancers. Its expression promotes activation of Rac and Cdc42 and leads to enhanced invasion and migration, as well as increased tumor cell survival and proliferation, suggesting that Vav1 could be a potent therapeutic target for pancreatic cancer. The purine analogue azathioprine, well known for its function as an anti-inflammatory compound, was recently shown to function by inhibiting Vav1 signaling in immune cells. We therefore hypothesized that azathioprine could also inhibit Vav1 in pancreatic tumor cells to reduce its proinvasive functions. Indeed, we have found that treatment of cultured pancreatic tumor cells with azathioprine inhibited Vav1-dependent invasive cell migration and matrix degradation, through inhibition of Rac and Cdc42 signaling. Furthermore, azathioprine treatment decreased metastasis in both xenograft and genetic mouse models of pancreatic cancer. Strikingly, metastasis was dramatically reduced in Vav1-expressing tumors arising from p48(Cre/+), Kras(G12D/+), p53(F/+) mice. These inhibitory effects were mediated through Vav1, as Vav1-negative cell lines and tumors were largely resistant to azathioprine treatment. These findings demonstrate that azathioprine and related compounds could be potent antimetastatic agents for Vav1-positive pancreatic tumors.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents/therapeutic use , Azathioprine/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Adenocarcinoma/genetics , Animals , Female , Humans , Mice , Mice, Nude , Mice, Transgenic , Molecular Targeted Therapy/methods , Neoplasm Invasiveness , Neoplasm Metastasis , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-vav/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cell Signal ; 26(10): 2202-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24880064

ABSTRACT

Vav proteins are guanine nucleotide exchange factors (GEFs) that activate a group of small G proteins (GTPases). Vav1 is predominantly expressed in hematopoietic cells, whereas Vav2 and Vav3 are ubiquitously distributed in almost all human tissues. All three Vav proteins contain conserved structural motifs and associate with a variety of cellular activities including proliferation, migration, and survival. Previous observation with Jurkat leukemia T cells showed that Vav1 possessed anti-apoptotic activity by enhancing Bcl-2 transcription. However the mechanism has not been unveiled. Here, we explored the effectors of Vav1 in promoting Bcl-2 expression in Jurkat cells and revealed that Rac2-Akt was specifically evoked by the expression of Vav1, but not Vav2 or Vav3. Although all three Vav isoforms existed in Jurkat cells, Rac2 was distinguishably activated by Vav1 and that led to enhanced Bcl-2 expression and cell survival. Akt was modulated downstream of Vav1-Rac2, and the activation of Akt was indispensable in the enhanced transcription of Bcl-2. Intriguingly, neither Vav2 nor Vav3 was able to activate Rac2-Akt pathway as determined by gene silencing approach. Our data illustrated a unique role of Vav1 in T leukemia survival by selectively triggering Rac2-Akt axis and elevating the expression of anti-apoptotic Bcl-2.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-vav/metabolism , rac GTP-Binding Proteins/metabolism , Apoptosis , HEK293 Cells , Humans , Jurkat Cells , Leukemia/metabolism , Leukemia/pathology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/genetics , RNA Interference , RNA, Small Interfering/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics , RAC2 GTP-Binding Protein
10.
Tumour Biol ; 35(2): 1481-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24072493

ABSTRACT

Vav3, a Rho GTPase guanine nucleotide exchange factor, is associated with tumor growth, apoptosis, invasion and metastasis, and angiogenesis. However, the role of Vav3 in gastric cancer remains unclear. In this study, Vav3 expression was blocked by specific siRNA in gastric cancer cell line MGC803. MTT was used to assay cell proliferation activity; wound healing assay and transwell assay were applied to detect cell migration and invasion ability; and qRT-PCR and Western blot were employed to detect expression levels of Vav3 as well as proliferation, migration, and invasion-related genes. The results showed that Vav3 expression in gastric cancer tissues and cell lines was significantly upregulated and was higher than that in adjacent tissues of cancer and normal gastric mucosal cell lines. Vav3 knockdown inhibited proliferation, migration, and invasion of MGC803 gastric cancer cells. The expression of P21, P27, TIMP-1, and TIMP-2 was upregulated, while proliferating cell nuclear antigen, cyclin E1, matrix metalloproteinase (MMP)-2, and MMP-7 were downregulated by Vav3 knockdown in MGC803 gastric cells. In conclusion, Vav3 is involved in the proliferation, migration, and invasion of gastric cancer cell as a tumor oncogene.


Subject(s)
Cell Movement/genetics , Cell Proliferation , Proto-Oncogene Proteins c-vav/genetics , Stomach Neoplasms/genetics , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , RNA, Small Interfering , Stomach Neoplasms/pathology
11.
Hepatology ; 58(2): 642-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23468064

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is characterized by active angiogenesis and metastasis, which account for rapid recurrence and poor survival. There is frequent down-regulation of miR-195 expression in HCC tissues. In this study, the role of miR-195 in HCC angiogenesis and metastasis was investigated with in vitro capillary tube formation and transwell assays, in vivo orthotopic xenograft mouse models, and human HCC specimens. Reduction of miR-195 in HCC tissues was significantly associated with increased angiogenesis, metastasis, and worse recurrence-free survival. Both gain-of-function and loss-of-function studies of in vitro models revealed that miR-195 not only suppressed the ability of HCC cells to promote the migration and capillary tube formation of endothelial cells but also directly repressed the abilities of HCC cells to migrate and invade extracellular matrix gel. Based on mouse models, we found that the induced expression of miR-195 dramatically reduced microvessel densities in xenograft tumors and repressed both intrahepatic and pulmonary metastasis. Subsequent investigations disclosed that miR-195 directly inhibited the expression of the proangiogenic factor vascular endothelial growth factor (VEGF) and the prometastatic factors VAV2 and CDC42. Knockdown of these target molecules of miR-195 phenocopied the effects of miR-195 restoration, whereas overexpression of these targets antagonized the function of miR-195. Furthermore, we revealed that miR-195 down-regulation resulted in enhanced VEGF levels in the tumor microenvironment, which subsequently activated VEGF receptor 2 signaling in endothelial cells and thereby promoted angiogenesis. Additionally, miR-195 down-regulation led to increases in VAV2 and CDC42 expression, which stimulated VAV2/Rac1/CDC42 signaling and lamellipodia formation and thereby facilitated the metastasis of HCC cells. CONCLUSION: miR-195 deregulation contributes to angiogenesis and metastasis in HCC. The restoration of miR-195 expression may be a promising strategy for HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , MicroRNAs/physiology , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic/physiopathology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , cdc42 GTP-Binding Protein/antagonists & inhibitors , Animals , Cell Line, Tumor , Down-Regulation/physiology , Heterografts , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-vav/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/physiology , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/physiology
13.
Sci Signal ; 5(225): ra38, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22623753

ABSTRACT

Interferon-ß (IFN-ß) is widely used to treat multiple sclerosis (MS), and its efficacy was demonstrated in the setting of experimental autoimmune encephalomyelitis (EAE), an animal model of MS; however, IFN-ß is not effective in treating all cases of MS. Here, we demonstrate that signaling by IFNAR (the shared receptor for IFN-α and IFN-ß) on macrophages inhibits activation of Rac1 and the generation of reactive oxygen species (ROS) through suppressor of cytokine signaling 1 (SOCS1). The inhibition of Rac1 activation and ROS generation suppressed the activity of the Nod-like receptor (NLR) family, pyrin domain-containing 3 (NLRP3) inflammasome, which resulted in attenuated EAE pathogenicity. We further found that two subsets of EAE could be defined on the basis of their dependency on the NLRP3 inflammasome and that IFN-ß was not an effective therapy when EAE was induced in an NLRP3 inflammasome-independent fashion. Thus, our study demonstrates a previously uncharacterized signaling pathway that is involved in the suppression of EAE by IFN-ß and characterizes NLRP3-independent EAE, which cannot be treated with IFN-ß.


Subject(s)
Carrier Proteins/physiology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Inflammasomes/physiology , Interferon-beta/therapeutic use , Receptor, Interferon alpha-beta/physiology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , CD4-Positive T-Lymphocytes/transplantation , Carrier Proteins/genetics , Caspase 1/physiology , Cytoskeletal Proteins/deficiency , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Enzyme Activation , Gene Expression Regulation/drug effects , Interferon beta-1b , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Neuropeptides/antagonists & inhibitors , Neuropeptides/physiology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/physiology , Reactive Oxygen Species/metabolism , Receptor, Interferon alpha-beta/deficiency , Recombinant Proteins/therapeutic use , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/physiology , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/physiology , rac1 GTP-Binding Protein
14.
Cell Physiol Biochem ; 28(2): 229-38, 2011.
Article in English | MEDLINE | ID: mdl-21865730

ABSTRACT

TGFß has been shown to induce short- and long-term actin reorganization controlled by Rho-GTPase signaling. A number of direct Smad target genes, rapidly activated by TGFß, have been previously reported to control the long-term Rho activation and actin reorganization. However, the molecular mechanisms that regulate the prompt stimulation of Rho GTPases by TGFß remain unknown. In the present study we report that TGFß rapidly stimulated RhoA and RhoB activation in JEG3 choriocarcinoma cells that lack endogenous Smad3. Inhibition of Smad2 expression via siRNA-mediated silencing or by blocking its phosphorylation using the TßRI inhibitor SB431542 did not prevent the early RhoA/B activation by TGFß indicating that this effect is Smad2/3-independent. Pre-treatment of the cells with the general tyrosine kinase inhibitor Genistein blocked the TGFß-induced early RhoA activation. In line with this finding, TGFß-stimulation resulted in a quick activation of the non-receptor tyrosine kinase Src, followed by activation of the guanine nucleotide exchange factor (GEF) Vav2. Inhibition of Src kinase by the selective inhibitor of the Src family tyrosine kinases PP2 totally blocked the early TGFß-induced RhoA activation. Similarly, Vav2 silencing via siRNA reduced the TGFß-induced RhoA activation implying that the rapid Src/Vav2 stimulation was effective in regulating RhoA activation. Our present findings provide for the first time a clear evidence for the role of Src and Vav2-GEF in the early Smad2/3-independent Rho activation by TGFß.


Subject(s)
Proto-Oncogene Proteins c-vav/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/pharmacology , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , Benzamides/pharmacology , Cell Line, Tumor , Dioxoles/pharmacology , Enzyme Activation/drug effects , Genistein/pharmacology , Humans , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/genetics , rhoB GTP-Binding Protein/metabolism
15.
Mol Cell Biol ; 30(20): 4786-96, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20696841

ABSTRACT

Although overactivation of RhoA is recognized as a common component of vascular disorders, the molecular mechanisms regulating RhoA activity in vascular smooth muscle cells (VSMC) are still unclear. We have previously shown that in VSMC, RhoA is phosphorylated on Ser188 by nitric oxide (NO)-stimulated cGMP-dependent kinase (PKG), which leads to RhoA-Rho kinase pathway inhibition. In this study, we showed that expression of phosphoresistant RhoA mutants prevented the stimulation of VSMC migration and adhesion induced by NO-PKG pathway activation. In contrast, under basal conditions, phosphomimetic RhoA mutants stimulated VSMC adhesion and migration through a signaling pathway requiring Rac1 and the Rho exchange factor Vav3. RhoA phosphorylation or phosphomimetic RhoA mutants induced Rac1 activation but did not activate Vav3. Indeed, phosphorylated RhoA or phosphomimetic mutants trapped guanine dissociation inhibitor α (GDIα), leading to the release of Rac1 and its translocation to the membrane, where it was then activated by the basal Vav3 nucleotide exchange activity. In vivo, RhoA phosphorylation induced by PKG activation in the aortas of rats treated with sildenafil induced dissociation of Rac1 from GDIα and activation of the Rac1 signaling pathway. These results suggest that the phosphorylation of RhoA represents a novel potent and physiological GDIα displacement factor that leads to Rac1 activation and regulation of Rac1-dependent VSMC functions.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Myocytes, Smooth Muscle/physiology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Base Sequence , Cell Adhesion/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/metabolism , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , RNA, Small Interfering/genetics , Rats , Signal Transduction , Thionucleotides/pharmacology , Wound Healing/physiology , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
16.
Cancer Biol Ther ; 8(14): 1402-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19597340

ABSTRACT

PURPOSE: Indoleamine 2,3-dioxygenase (IDO), a tryptophan catabolic enzyme, plays an important role in immune escape through suppressing T-cell function. Since Vav1 signaling pathway regulates T cell homeostasis, this study was designed to test the hypothesis that IDO induces T-cell immunosuppression through inhibiting Vav1 signaling. RESULTS: We found that IDO produced by IDO stably expressing CHO cells significantly inhibited interleukin (IL)-2 expression and proliferative response in T cells and increased apoptosis of T cells. IDO suppressed Vav1 mRNA and protein production in T cells. Furthermore, IDO inhibited TCR activation-induced Vav1 phosphorylation, which represents Vav1's activation state in T cells. These effects on T-cells induced by co-culture of CHO/IDO with T cells were attenuated by 1-MT. MATERIALS AND METHODS: Chinese hamster ovary (CHO) cells were stably transfected with human IDO (CHO/IDO). CD3(+) T cells were isolated from human peripheral blood monouclear cells. After co-culture of CHO/IDO cells with T cells in the presence or absence of an anti-CD3 antibody to activate T cell receptor (TCR) and/or 1-methyl-L-tryptophan (1-MT) to inhibit IDO activity, T cell proliferation and apoptosis were determined. T cell total RNA and cellular protein samples were isolated for detecting Vav1 gene and protein expression and activation state. CONCLUSIONS: The inhibitory effects of IDO on T cell immune responses may be through downregulation of Vav1 protein expression and activation. These studies provide insight into understanding the mechanisms of immune escape induced by IDO and therapeutic application of IDO inhibitors for cancer treatment.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , T-Lymphocytes/immunology , Animals , Apoptosis , CHO Cells , Cells, Cultured/cytology , Cells, Cultured/metabolism , Coculture Techniques , Cricetinae , Cricetulus , Humans , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lymphocyte Activation , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-vav/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/physiology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tryptophan/analogs & derivatives , Tryptophan/metabolism , Tryptophan/pharmacology , Tumor Escape/physiology
17.
J Cell Sci ; 121(Pt 16): 2635-42, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18653540

ABSTRACT

Rac1 has a crucial role in epidermal growth factor (EGF)-induced membrane ruffling, lamellipodial protrusion, and cell migration. Several guanine nucleotide exchange factors (GEFs) including Sos1, Sos2, Tiam1 and Vav2 have been shown to transduce the growth signal from the EGF receptor to Rac1. To clarify the role of each GEF, we time-lapse imaged the EGF-induced activity change of Rac1 in A431 cells transfected with siRNA targeting each Rac1 GEF. Because knockdown of these GEFs suppressed EGF-induced Rac1 activation only partially, we looked for another Rac1 GEF downstream of the EGF receptor and found that Asef, a Rac1-Cdc42 GEF bound to the tumor suppressor APC, also contributed to EGF-induced Rac1 activation. Intriguingly, EGF stimulation induced phosphorylation of Tyr94 within the APC-binding region of Asef in a manner dependent on Src-family tyrosine kinases. The suppression of EGF-induced Rac1 activation in siRNA-treated cells was restored by wild-type Asef, but not by the Tyr94Phe mutant of Asef. This observation strongly argues for the positive role of Tyr94 phosphorylation in EGF-induced Asef activation following the activation of Rac1.


Subject(s)
Epidermal Growth Factor/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , rac1 GTP-Binding Protein/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cells, Cultured , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/physiology , HeLa Cells , Humans , Models, Biological , Molecular Sequence Data , Phosphorylation/drug effects , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/physiology , RNA, Small Interfering/pharmacology , Rho Guanine Nucleotide Exchange Factors , Sequence Homology, Amino Acid , Son of Sevenless Protein, Drosophila/antagonists & inhibitors , Son of Sevenless Protein, Drosophila/genetics , Son of Sevenless Protein, Drosophila/physiology , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Tyrosine/metabolism
18.
Oncogene ; 27(19): 2673-85, 2008 Apr 24.
Article in English | MEDLINE | ID: mdl-18059343

ABSTRACT

The chimeric oncogene Bcr-Abl is known to induce autonomous motility of leukemic cells. We show here that p210(bcr-abl) responsible for chronic myelogenous leukemia induces an amoeboid type of motility while p190(bcr-abl), associated with acute lymphoid leukemia, induces a rolling type of motility. We previously reported that p210(bcr-abl) activates RhoA and Rac1, while p190(bcr-abl) although devoid of a Dbl-homology (DH) domain activates Rac1, but not RhoA. We investigated the regulation of GDP/GTP exchange factor (GEF) activities in the Bcr-Abl complex. For that purpose, different GEF activity mutants of Vav and of Bcr-Abl were constructed and stably transfected in Ba/F3 cells. Using these mutants, we demonstrate that RhoA is exclusively activated by the DH domain of p210(bcr-abl), while Rac1 activation is mostly due to Vav. Inhibition of Rac1 by Vav GEF mutant leads to immobilization of cells. Vav depletion using shRNA also induces immobilization of cells and suppression of GTP-bound Rac1. RhoA inactivation induces the specific loss of amoeboid movements. These results suggest that Rac1 activation by Vav triggers the motility of Bcr-Abl-expressing Ba/F3 cells, while the specific amoeboid mode of motility induced by p210(bcr-abl) is a consequence of RhoA activation.


Subject(s)
Cell Movement , DNA-Binding Proteins/physiology , Fusion Proteins, bcr-abl/physiology , GTPase-Activating Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , Proto-Oncogene Proteins c-vav/genetics , Repressor Proteins/physiology , Animals , Cell Line , Cell Migration Inhibition/genetics , Cell Movement/genetics , Cell Movement/physiology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Genes, abl , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Mice , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/physiology , RNA Interference , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/physiology
19.
J Immunol ; 179(1): 409-20, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579062

ABSTRACT

The biological effects of rIgG(1) 13B8.2, directed against the CDR3-like loop on the D1 domain of CD4, are partly due to signals that prevent NF-kappaB nuclear translocation, but the precise mechanisms of action, particularly at the level of membrane proximal signaling, remain obscure. We support the hypothesis that rIgG(1) 13B8.2 acts by interfering with the spatiotemporal distribution of signaling or receptor molecules inside membrane rafts. Upon cross-linking of Jurkat T lymphocytes, rIgG(1) 13B8.2 was found to induce an accumulation/retention of the CD4 molecule inside polyoxyethylene-20 ether Brij 98 detergent-resistant membranes at 37 degrees C, together with recruitment of TCR, CD3zeta, p56 Lck, Lyn, and Syk p70 kinases, linker for activation of T cells, and Csk-binding protein/phosphoprotein associated with glycosphingolipid adaptor proteins, and protein kinase Ctheta, but excluded Zap70 and its downstream targets Src homology 2-domain-containing leukocyte protein of 76 kDa, phospholipase Cgamma1, and p95(vav). Analysis of key upstream events such as Zap70 phosphorylation showed that modulation of Tyr(292) and Tyr(319) phosphorylation occurred concomitantly with 13B8.2-induced Zap70 exclusion from the membrane rafts. 13B8.2-induced differential raft partitioning was epitope, cholesterol, and actin dependent but did not require Ab hyper-cross-linking. Fluorescence confocal imaging confirmed the spatiotemporal segregation of the CD4 complex inside rafts and concomitant Zap70 exclusion, which occurred within 10-30 s following rIgG(1) 13B8.2 ligation, reached a plateau at 1 min, and persisted until the end of the 1-h experiment. The differential spatiotemporal partitioning between the CD4 receptor and the Zap70-signaling kinase inside membrane rafts interrupts the proximal signal cross-talk leading to subsequent NF-kappaB nuclear translocation and explains how baculovirus-expressed CD4-CDR3-like-specific rIgG(1) 13B8.2 acts to induce its biological effects.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , CD4 Antigens/metabolism , Membrane Microdomains/immunology , Phospholipase C gamma/antagonists & inhibitors , Phosphoproteins/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , ZAP-70 Protein-Tyrosine Kinase/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Antibodies, Blocking/genetics , Antibodies, Monoclonal/genetics , Baculoviridae/genetics , Baculoviridae/immunology , CD4 Antigens/immunology , Cross-Linking Reagents/metabolism , Detergents , Humans , Immunoglobulin G/genetics , Immunoglobulin G/pharmacology , Jurkat Cells , Membrane Microdomains/drug effects , Membrane Microdomains/enzymology , Membrane Microdomains/metabolism , Phospholipase C gamma/metabolism , Phosphoproteins/metabolism , Plant Oils , Polyethylene Glycols , Proto-Oncogene Proteins c-vav/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism
20.
Transpl Immunol ; 18(1): 53-61, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17584603

ABSTRACT

BACKGROUND: Vav1-Rac signaling plays a pivotal role in TCR/antigen and CD28 signals for T cell activation. However, pharmacological interference of this signaling has not been tested in the prevention of alloimmune-mediated allograft rejection. It has been demonstrated that 6-thio-GTP, a metabolite of azathioprine, specifically inhibits Vav1-Rac activity in T lymphocytes. Here we show the immunosuppressive efficacy of 6-thio-GTP in the prevention of cardiac allograft rejection. METHODS: T cell proliferations were measured by (3)H-thymidine uptake. The immunosuppressive activities of 6-thio-GTP were tested in the cardiac allograft model of C57BL/6 (H-2(b)) to Balb/c (H-2(d)) mice. RESULTS: 6-Thio-GTP inhibited TCR/alloantigen stimulated T cell proliferation and CD28-dependent T cell survival. Administration of 6-thio-GTP (0.5 mg/kg) prolonged graft survival to 13.8+/-2.39 days compared to 8.3+/-0.48 days in PBS controls (p<0.0001). Combination of 6-thio-GTP (0.5 mg/kg) with CsA (15 mg/kg) enhanced graft survival from 15.0+/-1.61 days in CsA treated recipients to 36.8+/-2.17 days in those received 20 days of combination therapy of CsA and 6-thio-GTP (p<0.0001), or to 42.7+/-16.63 days in the group treated with 20 days of CsA and 60 days of 6-thio-GTP (p<0.0001). Lymphocytes from 6-thio-GTP treated recipients with long-term surviving grafts (>60 days) displayed reduced proliferative response to alloantigen and higher frequencies of regulatory T cells (Treg). CONCLUSION: Vav1-Rac inhibitor 6-thio-GTP prolongs allograft survival alone or in combination with CsA by suppression of alloreactive T cell activation. Our findings suggest the therapeutic potential of pharmacological interference of Vav1-Rac signaling for transplantation.


Subject(s)
Graft Survival/drug effects , Guanosine Triphosphate/analogs & derivatives , Heart Transplantation , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Signal Transduction/drug effects , Thionucleotides/pharmacology , rac GTP-Binding Proteins/antagonists & inhibitors , Animals , CD28 Antigens/physiology , Cyclosporine/pharmacology , Guanosine Triphosphate/pharmacology , Heart Transplantation/immunology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Proto-Oncogene Proteins c-vav/physiology , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Transplantation, Homologous , rac GTP-Binding Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...