Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nature ; 609(7925): 174-182, 2022 09.
Article in English | MEDLINE | ID: mdl-36002574

ABSTRACT

The efficacy of adoptive T cell therapies for cancer treatment can be limited by suppressive signals from both extrinsic factors and intrinsic inhibitory checkpoints1,2. Targeted gene editing has the potential to overcome these limitations and enhance T cell therapeutic function3-10. Here we performed multiple genome-wide CRISPR knock-out screens under different immunosuppressive conditions to identify genes that can be targeted to prevent T cell dysfunction. These screens converged on RASA2, a RAS GTPase-activating protein (RasGAP) that we identify as a signalling checkpoint in human T cells, which is downregulated upon acute T cell receptor stimulation and can increase gradually with chronic antigen exposure. RASA2 ablation enhanced MAPK signalling and chimeric antigen receptor (CAR) T cell cytolytic activity in response to target antigen. Repeated tumour antigen stimulations in vitro revealed that RASA2-deficient T cells show increased activation, cytokine production and metabolic activity compared with control cells, and show a marked advantage in persistent cancer cell killing. RASA2-knockout CAR T cells had a competitive fitness advantage over control cells in the bone marrow in a mouse model of leukaemia. Ablation of RASA2 in multiple preclinical models of T cell receptor and CAR T cell therapies prolonged survival in mice xenografted with either liquid or solid tumours. Together, our findings highlight RASA2 as a promising target to enhance both persistence and effector function in T cell therapies for cancer treatment.


Subject(s)
Antigens, Neoplasm , Neoplasms , T-Lymphocytes , ras GTPase-Activating Proteins , Animals , Antigens, Neoplasm/immunology , Bone Marrow , CRISPR-Cas Systems , Disease Models, Animal , Gene Knockdown Techniques , Humans , Immunotherapy, Adoptive , Leukemia/immunology , Leukemia/pathology , Leukemia/therapy , Mice , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Xenograft Model Antitumor Assays , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34675073

ABSTRACT

Neutrophils sense and migrate through an enormous range of chemoattractant gradients through adaptation. Here, we reveal that in human neutrophils, calcium-promoted Ras inactivator (CAPRI) locally controls the GPCR-stimulated Ras adaptation. Human neutrophils lacking CAPRI (caprikd ) exhibit chemoattractant-induced, nonadaptive Ras activation; significantly increased phosphorylation of AKT, GSK-3α/3ß, and cofilin; and excessive actin polymerization. caprikd cells display defective chemotaxis in response to high-concentration gradients but exhibit improved chemotaxis in low- or subsensitive-concentration gradients of various chemoattractants, as a result of their enhanced sensitivity. Taken together, our data reveal that CAPRI controls GPCR activation-mediated Ras adaptation and lowers the sensitivity of human neutrophils so that they are able to chemotax through a higher-concentration range of chemoattractant gradients.


Subject(s)
Chemotaxis, Leukocyte/immunology , Neutrophils/immunology , ras GTPase-Activating Proteins/immunology , ras Proteins/antagonists & inhibitors , Actins/immunology , Cell Movement , Cell Polarity , Gene Knockdown Techniques , HL-60 Cells , Humans , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/drug effects , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/drug effects , Neutrophils/physiology , Receptors, G-Protein-Coupled/immunology , Shelterin Complex/immunology , Signal Transduction , Telomere-Binding Proteins/immunology , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics , ras Proteins/immunology
3.
DNA Cell Biol ; 39(7): 1127-1140, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32364766

ABSTRACT

Pulmonary microvascular endothelium barrier plays a critical role in protecting the pulmonary tissue from inflammatory injury in acute respiratory distress syndrome and acute lung injury (ARDS/ALI). The dysregulation of IQ-GTPase-activating protein 1 (IQGAP1) was an important etiology of endothelium barrier injury. However, significant differentially expressed genes (DEGs) and signaling pathways directly regulated by IQGAP1 are too complicated to fully understand. In this research, we identified a total of 1216 DEGs regulated by knockdown of IQGAP1 in rat pulmonary microvascular endothelial cells on the basis of transcriptomic RNA sequencing (RNA-Seq). Among them, 665 were upregulated DEGs and 551 were downregulated DEGs. Gene ontology analysis has revealed that upregulated DEGs were mainly enriched in DNA replication, cell cycle, and chromosome formation, while downregulated DEGs were mainly involved in the regulation of many cellular bioprocesses including cell proliferation, cell adhesion, and cell migration. Kyoto Encyclopedia of Genes and Genomes pathways analysis toward DEGs showed that upregulated pathways were mainly about DNA replication, while the significantly downregulated pathways were about TNF signaling pathway and some inflammatory- and proliferation-related pathways. Furthermore, we choose 30 DEGs for validation by qRT-PCR, the results were quite consistent with the RNA-Seq. In addition, we also found that knockdown of IQGAP1 caused a significant impact on many cytokines and inflammatory factors, which play a vital role in ARDS/ALI. In summary, in this study on the basis of RNA-Seq, we found IQGAP1 not only exerts a crucial role in microvascular endothelium barrier but also plays an important role in inflammation, which might provide a new insight for future study on IQGAP1 in the related diseases such as ARDS/ALI.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Lung/blood supply , Microvessels/cytology , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics , Animals , Gene Ontology , Rats
4.
Int Immunol ; 32(4): 233-241, 2020 04 12.
Article in English | MEDLINE | ID: mdl-31819988

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) play critical roles in type 2 immunity and are crucial for pathogenesis of various types of inflammatory disease. IQ motif-containing GTPase-activating protein 1 (IQGAP1) is a ubiquitously expressed scaffold protein that is involved in multiple cellular functions such as cell survival and trafficking. While the roles for IQGAP1 in T and B lymphocytes have been uncovered, the physiological significance of IQGAP1 in innate lymphocytes remains to be elucidated. In the current study, we demonstrate that using bone marrow chimeras, the deficiency of IQGAP1 caused an impaired survival of lung ILC2s in a cell-intrinsic manner and that Iqgap1-/- mice displayed decreased accumulation of ILC2s after administration of papain and thereby reduced the pathology of the disease. Moreover, Iqgap1-/- ILC2s showed a significantly enhanced apoptosis as compared to wild-type ILC2s under both steady-state and inflammatory conditions. Together these results identify for the first time that IQGAP1 is essential for homeostasis of ILC2s in the lung.


Subject(s)
Lung/immunology , Lymphocytes/immunology , ras GTPase-Activating Proteins/immunology , Animals , Homeostasis/immunology , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , ras GTPase-Activating Proteins/deficiency
5.
Pharmacol Rep ; 70(4): 777-783, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29940508

ABSTRACT

BACKGROUND: Synaptic Ras-GTPase-activating protein 1 (SYNGAP1) is an abundant brain-specific protein localized at the postsynaptic density of mammalian excitatory synapses. SYNGAP1 functions as a crucial regulator of downstream intracellular signaling triggered by N-methyl-d-aspartate receptor activation. One of the most important signaling pathways regulated by SYNGAP1 is the Ras-Raf-MEK-ERK pathway. SYNGAP1 deficiency is associated with hyperphosphorylation of MEK and ERK kinases and with altered synaptic function in Syngap1+/- mice. Loss-of-function mutations in the SYNGAP1 gene have been documented in many human cognitive and neurological disorders. However, there are currently no approaches that reverse the phenotypes of SYNGAP1 deficiency. METHODS: Using electrophysiological recordings of field responses in hippocampal slices, we examined if disturbances of synaptic physiology in the hippocampus of 7-8-month old Syngap1+/- mice were sensitive to the effect of the MEK inhibitor PD-0325901 given orally for 6days. RESULTS: We found that in hippocampal slices from vehicle-treated Syngap1+/- mice, basal synaptic responses were higher and their long-term potentiation (LTP) was lower than in slices from wild-type littermates. Chronic administration of PD-0325901 normalized basal synaptic responses, but did not reverse LTP deficit. CONCLUSIONS: The differential sensitivity of basal synaptic transmission and LTP to MEK inhibition indicates that the effects of SYNGAP1 deficiency on these synaptic parameters are mediated by distinct pathways. Our findings also suggest that at least some physiological phenotypes of the germline Syngap1 mutation can be ameliorated by pharmacological treatment of adult animals.


Subject(s)
Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Hippocampus/physiopathology , Membrane Potentials/drug effects , ras GTPase-Activating Proteins/deficiency , Animals , Diphenylamine/pharmacology , Female , Long-Term Potentiation/drug effects , Male , Mice , Mutation , ras GTPase-Activating Proteins/genetics
6.
Oncotarget ; 7(49): 80688-80699, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27729612

ABSTRACT

Helicobacter pylori infection is responsible for gastric carcinogenesis but host factors are also implicated. IQGAP1, a scaffolding protein of the adherens junctions interacting with E-cadherin, regulates cellular plasticity and proliferation. In mice, IQGAP1 deficiency leads to gastric hyperplasia. The aim of this study was to elucidate the consequences of IQGAP1 deletion on H. pylori-induced gastric carcinogenesis.Transgenic mice deleted for iqgap1 and WT littermates were infected with Helicobacter sp., and histopathological analyses of the gastric mucosa were performed. IQGAP1 and E-cadherin expression was evaluated in gastric tissues and in gastric epithelial cell lines in response to H. pylori infection. The consequences of IQGAP1 deletion on gastric epithelial cell behaviour and on the acquisition of cancer stem cell (CSC)-like properties were evaluated. After one year of infection, iqgap1+/- mice developed more preneoplastic lesions and up to 8 times more gastro-intestinal neoplasia (GIN) than WT littermates. H. pylori infection induced IQGAP1 and E-cadherin delocalization from cell-cell junctions. In vitro, knock-down of IQGAP1 favoured the acquisition of a mesenchymal phenotype and CSC-like properties induced by H. pylori infection.Our results indicate that alterations in IQGAP1 signalling promote the emergence of CSCs and gastric adenocarcinoma development in the context of an H. pylori infection.


Subject(s)
Adenocarcinoma/microbiology , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Neoplastic Stem Cells/microbiology , Precancerous Conditions/microbiology , Stomach Neoplasms/microbiology , ras GTPase-Activating Proteins/deficiency , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Genetic Predisposition to Disease , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Host-Pathogen Interactions , Hyaluronan Receptors/metabolism , Hyperplasia , Mice, 129 Strain , Mice, Knockout , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Time Factors , ras GTPase-Activating Proteins/genetics
7.
Dev Biol ; 418(2): 258-67, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27565025

ABSTRACT

Plexins (Plexs) comprise a large family of cell surface receptors for semaphorins (Semas) that function as evolutionarily conserved guidance molecules. GTPase activating protein (GAP) activity for Ras family small GTPases has been implicated in plexin signaling cascades through its RasGAP domain. However, little is known about how Ras family GTPases are controlled in vivo by plexin signaling. Here, we found that Drosophila Rap1, a member of the Ras family of GTPases, plays an important role controlling intersegmental nerve b motor axon guidance during neural development. Gain-of-function studies using dominant-negative and constitutively active forms of Rap1 indicate that Rap1 contributes to axonal growth and guidance. Genetic interaction analyses demonstrate that the Sema-1a/PlexA-mediated repulsive guidance function is regulated positively by Rap1. Furthermore, neuronal expression of mutant PlexA robustly restored defasciculation defects in PlexA null mutants when the catalytic arginine fingers of the PlexA RasGAP domain critical for GAP activity were disrupted. However, deleting the RasGAP domain abolished the ability of PlexA to rescue the PlexA guidance phenotypes. These findings suggest that PlexA-mediated motor axon guidance is dependent on the presence of the PlexA RasGAP domain, but not on its GAP activity toward Ras family small GTPases.


Subject(s)
Axon Guidance/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Monomeric GTP-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Telomere-Binding Proteins/physiology , ras GTPase-Activating Proteins/physiology , Animals , Animals, Genetically Modified , Axon Guidance/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genes, Insect , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Motor Neurons/physiology , Mutagenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Shelterin Complex , Telomere-Binding Proteins/deficiency , Telomere-Binding Proteins/genetics , Up-Regulation , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
8.
Cell Signal ; 28(11): 1623-30, 2016 11.
Article in English | MEDLINE | ID: mdl-27476001

ABSTRACT

Prospero-related homeobox 1 (PROX1) is an essential regulator in lymphangiogenesis and has been implicated in both oncogenic and tumor-suppressive functions in many types of human cancers. However, the role of PROX1 in prostate cancer (PCa) remains poorly understood. In this study, based on different PCa cell lines and knockout mice, we showed that PROX1 could be suppressed by DAB2IP, a novel member of the Ras GTPase-activating protein family and a critical player in control of epithelial-mesenchymal transition (EMT) and PCa metastasis. Mechanistically, PROX1 overexpression in DAB2IP-deficient PCa cells could enhance the accumulation of HIF1α protein by inhibiting ubiquitin pathway and then consequently induce an EMT response, which is characterized by repression of E-cadherin, up-regulation of vimentin and matrix metallopeptidases (MMPs) and enhancement of cell migration. Together, our data provides a new insight into mechanism that DAB2IP regulates EMT and PCa metastasis, especially points out the potential roles of its downstream PROX1/HIF1α signaling in a unique non-skeletal metastasis of PCa.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription, Genetic , Tumor Suppressor Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Animals , Cell Line, Tumor , Epithelium/metabolism , Epithelium/pathology , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , Models, Biological , Neoplasm Metastasis , Protein Stability , ras GTPase-Activating Proteins/deficiency
9.
Clin Cancer Res ; 22(3): 670-9, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26490309

ABSTRACT

PURPOSE: Clinical evidence suggests increased cancer stem cells (CSCs) in a tumor mass may contribute to the failure of conventional therapies because CSCs seem to be more resistant than differentiated tumor cells. Thus, unveiling the mechanism regulating CSCs and candidate target molecules will provide new strategy to cure the patients. EXPERIMENTAL DESIGN: The stem-like cell properties were determined by a prostasphere assay and dye exclusion assay. To find critical stem cell marker and reveal regulation mechanism, basic biochemical and molecular biologic methods, such as quantitative real-time PCR, Western blot, reporter gene assay, and chromatin immunoprecipitation assay, were used. In addition, to determine the effect of combination therapy targeting both CSCs and its progeny, in vitro MTT assay and in vivo xenograft model was used. RESULTS: We demonstrate immortalized normal human prostate epithelial cells, appeared nontumorigenic in vivo, become tumorigenic, and acquire stem cell phenotype after knocking down a tumor suppressor gene. Also, those stem-like cells increase chemoresistance to conventional anticancer reagent. Mechanistically, we unveil that Wnt signaling is a key pathway regulating well-known stem cell marker CD44 by directly interacting to the promoter. Thus, by targeting CSCs using Wnt inhibitors synergistically enhances the efficacy of conventional drugs. Furthermore, the in vivo mouse model bearing xenografts showed a robust inhibition of tumor growth after combination therapy. CONCLUSIONS: Overall, this study provides strong evidence of CSC in castration-resistant prostate cancer. This new combination therapy strategy targeting CSC could significantly enhance therapeutic efficacy of current chemotherapy regimen only targeting non-CSC cells.


Subject(s)
Neoplastic Stem Cells/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Docetaxel , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Male , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Binding , Taxoids/pharmacology , Tumor Stem Cell Assay , Wnt Signaling Pathway , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
10.
Eur J Immunol ; 45(5): 1512-23, 2015 May.
Article in English | MEDLINE | ID: mdl-25652366

ABSTRACT

Ras GTPase-activating proteins negatively regulate the Ras/Erk signaling pathway, thereby playing crucial roles in the proliferation, function, and development of various types of cells. In this study, we identified a novel Ras GTPase-activating proteins protein, RASAL3, which is predominantly expressed in cells of hematopoietic lineages, including NKT, B, and T cells. We established systemic RASAL3-deficient mice, and the mice exhibited a severe decrease in NKT cells in the liver at 8 weeks of age. The treatment of RASAL3-deficient mice with α-GalCer, a specific agonist for NKT cells, induced liver damage, but the level was less severe than that in RASAL3-competent mice, and the attenuated liver damage was accompanied by a reduced production of interleukin-4 and interferon-γ from NKT cells. RASAL3-deficient NKT cells treated with α-GalCer in vitro presented augmented Erk phosphorylation, suggesting that there is dysregulated Ras signaling in the NKT cells of RASAL3-deficient mice. Taken together, these results suggest that RASAL3 plays an important role in the expansion and functions of NKT cells in the liver by negatively regulating Ras/Erk signaling, and might be a therapeutic target for NKT-associated diseases.


Subject(s)
Natural Killer T-Cells/immunology , ras GTPase-Activating Proteins/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Galactosylceramides/administration & dosage , Galactosylceramides/immunology , Gene Knockdown Techniques , Humans , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Jurkat Cells , Liver/immunology , Liver/injuries , Liver/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CXCR/metabolism , Receptors, CXCR6 , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
11.
Biol Psychiatry ; 77(9): 805-15, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25444158

ABSTRACT

BACKGROUND: Genetic haploinsufficiency of SYNGAP1/Syngap1 commonly occurs in developmental brain disorders, such as intellectual disability, epilepsy, schizophrenia, and autism spectrum disorder. Thus, studying mouse models of Syngap1 haploinsufficiency may uncover pathologic developmental processes common among distinct brain disorders. METHODS: A Syngap1 haploinsufficiency model was used to explore the relationship between critical period dendritic spine abnormalities, cortical circuit assembly, and the window for genetic rescue to understand how damaging mutations disrupt key substrates of mouse brain development. RESULTS: Syngap1 mutations broadly disrupted a developmentally sensitive period that corresponded to the period of heightened postnatal cortical synaptogenesis. Pathogenic Syngap1 mutations caused a coordinated acceleration of dendrite elongation and spine morphogenesis and pruning of these structures in neonatal cortical pyramidal neurons. These mutations also prevented a form of developmental structural plasticity associated with experience-dependent reorganization of brain circuits. Consistent with these findings, Syngap1 mutant mice displayed an altered pattern of long-distance synaptic inputs into a cortical area important for cognition. Interestingly, the ability to genetically improve the behavioral endophenotype of Syngap1 mice decreased slowly over postnatal development and mapped onto the developmental period of coordinated dendritic insults. CONCLUSIONS: Pathogenic Syngap1 mutations have a profound impact on the dynamics and structural integrity of pyramidal cell postsynaptic structures known to guide the de novo wiring of nascent cortical circuits. These findings support the idea that disrupted critical periods of dendritic growth and spine plasticity may be a common pathologic process in developmental brain disorders.


Subject(s)
Cerebral Cortex/abnormalities , Cerebral Cortex/growth & development , Haploinsufficiency , Pyramidal Cells/physiology , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics , Animals , Animals, Newborn , Conditioning, Psychological/physiology , Dendritic Spines/pathology , Dendritic Spines/physiology , Endophenotypes , Exploratory Behavior/physiology , Fear/physiology , Hippocampus/abnormalities , Hippocampus/growth & development , Maze Learning/physiology , Mice, Transgenic , Neural Pathways/abnormalities , Neural Pathways/growth & development , Pyramidal Cells/pathology , Sensory Deprivation/physiology , Vibrissae/physiology
12.
Oncotarget ; 5(14): 5523-34, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25015118

ABSTRACT

Cytolethal distending toxin (CDT) produced by Campylobacter jejuni is a genotoxin that induces cell-cycle arrest and apoptosis in mammalian cells. Recent studies have demonstrated that prostate cancer (PCa) cells can acquire radio-resistance when DOC-2/DAB2 interactive protein (DAB2IP) is downregulated. In this study, we showed that CDT could induce cell death in DAB2IP-deficient PCa cells. A combination of CDT and radiotherapy significantly elicited cell death in DAB2IP-deficient PCa cells by inhibiting the repair of ionizing radiation (IR)-induced DNA double-strand break (DSB) during G2/M arrest, which is triggered by ataxia telangiectasia mutated (ATM)-dependent DNA damage checkpoint responses. We also found that CDT administration significantly increased the efficacy of radiotherapy in a xenograft mouse model. These results indicate that CDT can be a potent therapeutic agent for radio-resistant PCa.


Subject(s)
Bacterial Toxins/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Prostatic Neoplasms/pathology , Radiation, Ionizing , Xenograft Model Antitumor Assays , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
13.
J Biol Chem ; 289(13): 9100-12, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24550401

ABSTRACT

The estrogen receptor (ER) is a steroid hormone receptor that acts as a transcription factor, modulating genes that regulate a vast range of cellular functions. IQGAP1 interacts with several signaling proteins, cytoskeletal components, and transmembrane receptors, thereby serving as a scaffold to integrate signaling pathways. Both ERα and IQGAP1 contribute to breast cancer. In this study, we report that IQGAP1 binds ERα and ERß. In vitro analysis with pure proteins revealed a direct interaction between IQGAP1 and ERα. Investigation with multiple short fragments of each protein showed that ERα binds to the IQ domain of IQGAP1, whereas the hinge region of ERα is responsible for binding IQGAP1. In addition, IQGAP1 and ERα co-immunoprecipitated from cells, and the association was modulated by estradiol. The interaction has functional effects. Knockdown of endogenous IQGAP1 attenuated the ability of estradiol to induce transcription of the estrogen-responsive genes pS2, progesterone receptor, and cyclin D1. These data reveal that IQGAP1 binds to ERα and modulates its transcriptional function, suggesting that IQGAP1 might be a target for therapy in patients with breast carcinoma.


Subject(s)
Estrogen Receptor alpha/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , Cell Line , Estradiol/pharmacology , Gene Knockdown Techniques , Humans , Protein Binding , Transcriptional Activation/drug effects , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
14.
Arterioscler Thromb Vasc Biol ; 34(3): 603-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24407031

ABSTRACT

OBJECTIVE: To investigate the novel function of ASK1-interacting protein-1 (AIP1) in vascular endothelial cell growth factor receptor (VEGFR)-3 signaling, and VEGFR-3-dependent angiogenesis and lymphangiogenesis. APPROACH AND RESULTS: AIP1, a signaling scaffold protein, is highly expressed in the vascular endothelium. We have previously reported that AIP1 functions as an endogenous inhibitor in pathological angiogenesis by blocking VEGFR-2 activity. Surprisingly, here we observe that mice with a global deletion of AIP1-knockout mice (AIP1-KO) exhibit reduced retinal angiogenesis with less sprouting and fewer branches. Vascular endothelial cell (but not neuronal)-specific deletion of AIP1 causes similar defects in retinal angiogenesis. The reduced retinal angiogenesis correlates with reduced expression in VEGFR-3 despite increased VEGFR-2 levels in AIP1-KO retinas. Consistent with the reduced expression of VEGFR-3, AIP1-KO show delayed developmental lymphangiogenesis in neonatal skin and mesentery, and mount weaker VEGF-C-induced cornea lymphangiogenesis. In vitro, human lymphatic endothelial cells with AIP1 small interfering RNA knockdown, retinal endothelial cells, and lymphatic endothelial cells isolated from AIP1-KO all show attenuated VEGF-C-induced VEGFR-3 signaling. Mechanistically, we demonstrate that AIP1 via vegfr-3-specific miR-1236 increases VEGFR-3 protein expression and that, by directly binding to VEGFR-3, it enhances VEGFR-3 endocytosis and stability. CONCLUSION: Our in vivo and in vitro results provide the first insight into the mechanism by which AIP1 mediates VEGFR-3-dependent angiogenic and lymphangiogenic signaling.


Subject(s)
Carrier Proteins/physiology , Lymphangiogenesis/physiology , Retinal Neovascularization/physiopathology , Vascular Endothelial Growth Factor Receptor-3/physiology , ras GTPase-Activating Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Cornea , Endocytosis , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Eye Proteins/physiology , Guanylate Kinases , Humans , Mice , Mice, Knockout , MicroRNAs/physiology , Neurons/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Notch/physiology , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/physiology , Vascular Endothelial Growth Factor Receptor-3/biosynthesis , Vascular Endothelial Growth Factor Receptor-3/genetics , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
15.
PLoS One ; 8(8): e71826, 2013.
Article in English | MEDLINE | ID: mdl-23951254

ABSTRACT

It is broadly accepted that genetically engineered animal models do not always recapitulate human pathobiology. Therefore identifying best-fit mouse models of human cancers that truly reflect the corresponding human disease is of vital importance in elucidating molecular mechanisms of tumorigenesis and developing preventive and therapeutic approaches. A new hepatocellular carcinoma (HCC) mouse model lacking a novel putative tumor suppressor IQGAP2 has been generated by our laboratory. The aim of this study was to obtain the molecular signature of Iqgap2(-/-) HCC tumors and establish the relevance of this model to human disease. Here we report a comprehensive transcriptome analysis of Iqgap2(-/-) livers and a cross-species comparison of human and Iqgap2(-/-) HCC tumors using Significance Analysis of Microarray (SAM) and unsupervised hierarchical clustering analysis. We identified the Wnt/ß-catenin signaling pathway as the top canonical pathway dysregulated in Iqgap2(-/-) livers. We also demonstrated that Iqgap2(-/-) hepatic tumors shared genetic signatures with HCC tumors from patients with advanced disease as evidenced by a 78% mouse-to-human microarray data set concordance rate with 117 out of 151 identified ortholog genes having similar expression profiles across the two species. Collectively, these results indicate that the Iqgap2 knockout mouse model closely recapitulates human HCC at the molecular level and supports its further application for the study of this disease.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Gene Expression Profiling , Liver Neoplasms/genetics , Liver Neoplasms/pathology , ras GTPase-Activating Proteins/deficiency , Animals , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Disease Models, Animal , Female , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Knockout , Neoplasm Staging , Reproducibility of Results , Signal Transduction , Transcriptome , Wnt Signaling Pathway , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
16.
Arterioscler Thromb Vasc Biol ; 33(4): 795-804, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23413429

ABSTRACT

OBJECTIVE: Apoptosis signal-regulating kinase 1-interacting protein-1 (AIP1) is a signaling adaptor molecule implicated in stress and apoptotic signaling induced by proinflammatory mediators. However, its function in atherosclerosis has not been established. In the present study, we use AIP1-null (AIP1(-/-)) mice to examine its effect on atherosclerotic lesions in an apolipoprotein E-null (ApoE(-/-)) mouse model of atherosclerosis. APPROACH AND RESULTS: ApoE(-/-) control mice developed atherosclerosis in the aortic roots and descending aortas on Western-type diet for 10 weeks, whereas the atherosclerotic lesions are significantly augmented in ApoE(-/-)AIP1(-/-) double knockout (DKO) mice. DKO mice show increases in plasma inflammatory cytokines with no significant alterations in body weight, total cholesterol levels, or lipoprotein profiles. Aortas in DKO mice show increased inflammation and endothelial cell (EC) dysfunction with nuclear factor-κB activity, correlating with increased accumulation of macrophages in the lesion area. Importantly, macrophages from DKO donors are not sufficient to augment inflammatory responses and atherogenesis when transferred to ApoE-KO recipients. Mechanistic studies suggest that AIP1 is highly expressed in aortic EC, but not in macrophages, and AIP1 deletion in EC significantly enhance oxidized low-density lipoprotein-induced nuclear factor-κB signaling, gene expression of inflammatory molecules, and monocyte adhesion, suggesting that vascular EC are responsible for the increased inflammatory responses observed in DKO mice. CONCLUSIONS: Our data demonstrate that loss of AIP1 in aortic EC primarily contributes to the exacerbated lesion expansion in the ApoE(-/-)AIP1(-/-) mice, revealing an important role of AIP1 in limiting inflammation, EC dysfunction, and atherosclerosis.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Endothelium, Vascular/metabolism , Hyperlipidemias/complications , Inflammation/prevention & control , Vasoconstriction , Vasodilation , ras GTPase-Activating Proteins/metabolism , Animals , Aortic Diseases/blood , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Biomarkers/blood , Bone Marrow Transplantation , Cholesterol/blood , Cytokines/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Gene Expression Regulation , Hyperlipidemias/blood , Hyperlipidemias/genetics , Hyperlipidemias/physiopathology , Inflammation/blood , Inflammation/etiology , Inflammation/genetics , Inflammation/physiopathology , Inflammation Mediators/blood , Lipoproteins/blood , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Macrophages/transplantation , Mice , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction , Triglycerides/blood , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics
17.
PLoS One ; 8(1): e53635, 2013.
Article in English | MEDLINE | ID: mdl-23326475

ABSTRACT

DOC-2/DAB-2 interacting protein (Dab2IP) is a GTPase activating protein that binds to Disabled-1, a cytosolic adapter protein involved in Reelin signaling and brain development. Dab2IP regulates PI3K-AKT signaling and is associated with metastatic prostate cancer, abdominal aortic aneurysms and coronary heart disease. To date, the physiological function of Dab2IP in the nervous system, where it is highly expressed, is relatively unknown. In this study, we generated a mouse model with a targeted disruption of Dab2IP using a retrovirus gene trap strategy. Unlike reeler mice, Dab2IP knock-down mice did not exhibit severe ataxia or cerebellar hypoplasia. However, Dab2IP deficiency produced a number of cerebellar abnormalities such as a delay in the development of Purkinje cell (PC) dendrites, a decrease in the parallel fiber synaptic marker VGluT1, and an increase in the climbing fiber synaptic marker VGluT2. These findings demonstrate for the first time that Dab2IP plays an important role in dendrite development and regulates the number of synapses in the cerebellum.


Subject(s)
Cerebellum/cytology , Cerebellum/enzymology , Dendrites/enzymology , Neurogenesis , Synapses/enzymology , ras GTPase-Activating Proteins/metabolism , Animals , Biomarkers/metabolism , Female , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mossy Fibers, Hippocampal/enzymology , Protein Transport , Purkinje Cells/cytology , Purkinje Cells/enzymology , Reelin Protein , Reproducibility of Results , ras GTPase-Activating Proteins/deficiency
18.
Learn Mem ; 19(7): 268-81, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22700469

ABSTRACT

The synaptic Ras/Rap-GTPase-activating protein (SynGAP1) plays a unique role in regulating specific downstream intracellular events in response to N-methyl-D-aspartate receptor (NMDAR) activation. Constitutive heterozygous loss of SynGAP1 disrupts NMDAR-mediated physiological and behavioral processes, but the disruptions might be of developmental origin. Therefore, the precise role of SynGAP1 in the adult brain, including its relative functional significance within specific brain regions, remains unexplored. The present study constitutes the first attempt in achieving adult hippocampal-specific SynGAP1 knockout using the Cre/loxP approach. Here, we report that this manipulation led to a significant numerical increase in both small and large GluA1 and NR1 immunoreactive clusters, many of which were non-opposed to presynaptic terminals. In parallel, the observed marked decline in the amplitude of spontaneous excitatory currents (sEPSCs) and inter-event intervals supported the impression that SynGAP1 loss might facilitate the accumulation of extrasynaptic glutamatergic receptors. In addition, SynGAP1-mediated signaling appears to be critical for the proper integration and survival of newborn neurons. The manipulation impaired reversal learning in the probe test of the water maze and induced a delay-dependent impairment in spatial recognition memory. It did not significantly affect anxiety or reference memory acquisition but induced a substantial elevation in spontaneous locomotor activity in the open field test. Thus, the present study demonstrates the functional significance of SynGAP1 signaling in the adult brain by capturing several changes that are dependent on NMDAR and hippocampal integrity.


Subject(s)
Hippocampus/cytology , Learning Disabilities/genetics , Neurons/physiology , Synaptic Transmission/genetics , ras GTPase-Activating Proteins/deficiency , Analysis of Variance , Animals , Avoidance Learning/physiology , Doublecortin Domain Proteins , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Hippocampus/metabolism , Integrases/genetics , Integrases/metabolism , Maze Learning/physiology , Membrane Potentials/genetics , Memory Disorders/genetics , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Motor Activity/genetics , Neurons/cytology , Neurons/drug effects , Neuropeptides/metabolism , Patch-Clamp Techniques , Reaction Time/genetics , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Spatial Behavior/physiology , Synaptic Transmission/drug effects , Transduction, Genetic , ras GTPase-Activating Proteins/metabolism
19.
J Immunol ; 188(5): 2057-63, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22345702

ABSTRACT

Activating and inhibiting receptors of lymphocytes collect valuable information about their mikròs kósmos. This information is essential to initiate or to turn off complex signaling pathways. Irrespective of these advances, our knowledge on how these intracellular activation cascades are coordinated in a spatiotemporal manner is far from complete. Among multiple explanations, the scaffolding proteins have emerged as a critical piece of this evolutionary tangram. Among many, IQGAP1 is one of the essential scaffolding proteins that coordinate multiple signaling pathways. IQGAP1 possesses multiple protein interaction motifs to achieve its scaffolding functions. Using these domains, IQGAP1 has been shown to regulate a number of essential cellular events. This includes actin polymerization, tubulin multimerization, microtubule organizing center formation, calcium/calmodulin signaling, Pak/Raf/Mek1/2-mediated Erk1/2 activation, formation of maestrosome, E-cadherin, and CD44-mediated signaling and glycogen synthase kinase-3/adenomatous polyposis coli-mediated ß-catenin activation. In this review, we summarize the recent developments and exciting new findings of cellular functions of IQGAP1.


Subject(s)
Cell Communication/immunology , Intracellular Space/immunology , Intracellular Space/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Protein Multimerization/immunology , ras GTPase-Activating Proteins/physiology , Animals , Cells, Cultured , Intracellular Space/chemistry , Lymphocyte Subsets/chemistry , Mice , Mice, Knockout , Protein Interaction Mapping/methods , Signal Transduction/immunology , ras GTPase-Activating Proteins/chemistry , ras GTPase-Activating Proteins/deficiency
20.
Regul Pept ; 173(1-3): 36-46, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-21968151

ABSTRACT

UNLABELLED: Long-chain fatty acids (LCFA) serve as structural components for membrane biogenesis and as primary energy sources during mitochondrial ß-oxidation reactions. Hepatic LCFA uptake is complex, with characteristics suggestive of a dual-kinetic model manifested by rapid (carrier-assisted/facilitated) and delayed (passive diffusional) phases. Our previous work using mice deficient of the Iqgap2 gene established a highly novel link between IQGAP2, a putative GTPase-activating protein, and hepatocarcinogenesis. Now we report that Iqgap2 deficiency also results in selective loss of the facilitated phase of hepatocyte LCFA uptake with preservation of the diffusional component. This molecular defect was seen in Iqgap2(-/-) hepatocytes of all ages studied (1-, 4-, 8-months). The loss of facilitated LCFA uptake protected against development of hepatic triglyceride accumulation in Iqgap2-deficient mice fed high-fat diet, consistent with a fundamental role in physiological fat partitioning. These phenotypic changes could not be explained by genetic loss of fatty acid processing proteins known to regulate lipid uptake or metabolic processing pathways. Iqgap2-deficient livers also displayed enhanced insulin sensitivity. CONCLUSION: These observations identify a novel property of the putative GTPase-activating protein IQGAP2 in LCFA uptake in vitro and in vivo, and implicate IQGAP2 in an intracellular signaling pathway necessary for functional fatty acid uptake, lipid processing, and, possibly, glucose homeostasis.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified/metabolism , Fatty Liver/etiology , ras GTPase-Activating Proteins/genetics , Animals , Cells, Cultured , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Gene Expression Profiling , Glucose Tolerance Test , Hep G2 Cells , Hepatocytes/metabolism , Humans , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Male , Mice , Mice, 129 Strain , Mice, Knockout , PPAR alpha/genetics , PPAR alpha/metabolism , Transcription, Genetic , Triglycerides/blood , Triglycerides/metabolism , Weight Gain , ras GTPase-Activating Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...