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











Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119783, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38871226

ABSTRACT

Kinases are known to have kinase activity independent functions. To gain further insights into potential kinase-independent functions of SLK/STK2, we have developed a kinase-dead allele, SLKK63R using in vivo CRISPR/Cas technology. Our studies show that blastocysts homozygote for SLKK63R do not develop into viable mice. However, heterozygotes are viable and fertile with no overt phenotypes. Analyses of mouse embryonic fibroblasts show that expression of SLKK63R results in a 50% decrease in kinase activity in heterozygotes. In contrast to previous studies, our data show that SLK does not form homodimers and that the kinase defective allele does not act in a dominant negative fashion. Expression of SLKK63R leads to altered Rac1 and RhoA activity, increased stress fiber formation and delayed focal adhesion turnover. Our data support a previously observed role for SLK in cell migration and suggest that at least 50% kinase activity is sufficient for embryonic development.


Subject(s)
Cell Movement , Fibroblasts , Protein Serine-Threonine Kinases , rac1 GTP-Binding Protein , Animals , Fibroblasts/metabolism , Fibroblasts/cytology , Cell Movement/genetics , Mice , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , Focal Adhesions/metabolism , Focal Adhesions/genetics , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Embryo Loss/genetics , Embryo Loss/pathology , CRISPR-Cas Systems , Female , Neuropeptides
2.
Cell Rep ; 42(8): 112936, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37552602

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) plays a crucial role in metastasis, which is the leading cause of death in breast cancer patients. Here, we show that Cdc42 GTPase-activating protein (CdGAP) promotes tumor formation and metastasis to lungs in the HER2-positive (HER2+) murine breast cancer model. CdGAP facilitates intravasation, extravasation, and growth at metastatic sites. CdGAP depletion in HER2+ murine primary tumors mediates crosstalk with a Dlc1-RhoA pathway and is associated with a transforming growth factor ß (TGF-ß)-induced EMT transcriptional signature. CdGAP is positively regulated by TGF-ß signaling during EMT and interacts with the adaptor talin to modulate focal adhesion dynamics and integrin activation. Moreover, HER2+ breast cancer patients with high CdGAP mRNA expression combined with a high TGF-ß-EMT signature are more likely to present lymph node invasion. Our results suggest CdGAP as a candidate therapeutic target for HER2+ metastatic breast cancer by inhibiting TGF-ß and integrin/talin signaling pathways.


Subject(s)
Breast Neoplasms , Transforming Growth Factor beta , Humans , Animals , Mice , Female , Transforming Growth Factor beta/metabolism , Breast Neoplasms/pathology , Talin/metabolism , Carrier Proteins , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Integrins/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Neoplasm Metastasis , Cell Movement
3.
Cell Stem Cell ; 30(2): 188-206.e6, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36640764

ABSTRACT

A central factor in the maintenance of tissue integrity is the response of stem cells to variations in the levels of niche signals. In the gut, intestinal stem cells (ISCs) depend on Wnt ligands for self-renewal and proliferation. Transient increases in Wnt signaling promote regeneration after injury or in inflammatory bowel diseases, whereas constitutive activation of this pathway leads to colorectal cancer. Here, we report that Discs large 1 (Dlg1), although dispensable for polarity and cellular turnover during intestinal homeostasis, is required for ISC survival in the context of increased Wnt signaling. RNA sequencing (RNA-seq) and genetic mouse models demonstrated that DLG1 regulates the cellular response to increased canonical Wnt ligands. This occurs via the transcriptional regulation of Arhgap31, a GTPase-activating protein that deactivates CDC42, an effector of the non-canonical Wnt pathway. These findings reveal a DLG1-ARHGAP31-CDC42 axis that is essential for the ISC response to increased niche Wnt signaling.


Subject(s)
Intestinal Mucosa , Wnt Signaling Pathway , Animals , Mice , Cell Proliferation , GTPase-Activating Proteins/metabolism , Intestinal Mucosa/metabolism , Intestines , Stem Cell Niche , Stem Cells , Wnt Signaling Pathway/genetics
4.
Sci Rep ; 12(1): 18657, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333327

ABSTRACT

Rho GTPases are regulators of the actin cytoskeleton and their activity is modulated by GTPase-activating proteins (GAPs) and guanine nucleotide exchanging factors (GEFs). Glomerular podocytes have numerous actin-based projections called foot processes and their alteration is characteristic of proteinuric kidney diseases. We reported previously that Rac1 hyperactivation in podocytes causes proteinuria and glomerulosclerosis in mice. However, which GAP and GEF modulate Rac1 activity in podocytes remains unknown. Here, using a proximity-based ligation assay, we identified CdGAP (ARHGAP31) and ß-PIX (ARHGEF7) as the major regulatory proteins interacting with Rac1 in human podocytes. CdGAP interacted with ß-PIX through its basic region, and upon EGF stimulation, they both translocated to the plasma membrane in podocytes. CdGAP-depleted podocytes had altered cell motility and increased basal Rac1 and Cdc42 activities. When stimulated with EGF, CdGAP-depleted podocytes showed impaired ß-PIX membrane-translocation and tyrosine phosphorylation, and reduced activities of Src kinase, focal adhesion kinase, and paxillin. Systemic and podocyte-specific CdGAP-knockout mice developed mild but significant proteinuria, which was exacerbated by Adriamycin. Collectively, these findings show that CdGAP contributes to maintain podocyte function and protect them from injury.


Subject(s)
Podocytes , Humans , Mice , Animals , Podocytes/metabolism , Focal Adhesions , src-Family Kinases/metabolism , Epidermal Growth Factor/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Proteinuria/metabolism , Mice, Knockout
5.
Genet Med ; 24(12): 2501-2515, 2022 12.
Article in English | MEDLINE | ID: mdl-36178483

ABSTRACT

PURPOSE: The study aimed to identify novel genes for idiopathic hypogonadotropic hypogonadism (IHH). METHODS: A cohort of 1387 probands with IHH underwent exome sequencing and de novo, familial, and cohort-wide investigations. Functional studies were performed on 2 p190 Rho GTPase-activating proteins (p190 RhoGAP), ARHGAP35 and ARHGAP5, which involved in vivo modeling in larval zebrafish and an in vitro p190A-GAP activity assay. RESULTS: Rare protein-truncating variants (PTVs; n = 5) and missense variants in the RhoGAP domain (n = 7) in ARHGAP35 were identified in IHH cases (rare variant enrichment: PTV [unadjusted P = 3.1E-06] and missense [adjusted P = 4.9E-03] vs controls). Zebrafish modeling using gnrh3:egfp phenotype assessment showed that mutant larvae with deficient arhgap35a, the predominant ARHGAP35 paralog in the zebrafish brain, display decreased GnRH3-GFP+ neuronal area, a readout for IHH. In vitro GAP activity studies showed that 1 rare missense variant [ARHGAP35 p.(Arg1284Trp)] had decreased GAP activity. Rare PTVs (n = 2) also were discovered in ARHGAP5, a paralog of ARHGAP35; however, arhgap5 zebrafish mutants did not display significant GnRH3-GFP+ abnormalities. CONCLUSION: This study identified ARHGAP35 as a new autosomal dominant genetic driver for IHH and ARHGAP5 as a candidate gene for IHH. These observations suggest a novel role for the p190 RhoGAP proteins in GnRH neuronal development and integrity.


Subject(s)
Hypogonadism , Zebrafish , Animals , Humans , Zebrafish/genetics , Hypogonadism/genetics , Gonadotropin-Releasing Hormone/genetics , Repressor Proteins , Guanine Nucleotide Exchange Factors , GTPase-Activating Proteins/genetics
6.
Wiley Interdiscip Rev RNA ; 13(2): e1678, 2022 03.
Article in English | MEDLINE | ID: mdl-34155820

ABSTRACT

Microexons are small sized (≤51 bp) exons which undergo extensive alternative splicing in neurons, microglia, embryonic stem cells, and cancer cells, giving rise to cell type specific protein isoforms. Due to their small sizes, microexons provide a unique challenge for the splicing machinery. They frequently lack exon splicer enhancers/repressors and require specialized neighboring trans-regulatory and cis-regulatory elements bound by RNA binding proteins (RBPs) for their inclusion. The functional consequences of including microexons within mRNAs have been extensively documented in the central nervous system (CNS) and aberrations in their inclusion have been observed to lead to abnormal processes. Despite the increasing evidence for microexons impacting cellular physiology within CNS, mechanistic details illustrating their functional importance in diseases of the CNS is still limited. In this review, we discuss the unique characteristics of microexons, and how RBPs participate in regulating their inclusion and exclusion during splicing. We consider recent findings of microexon alternative splicing and their implication for regulating the function of small GTPases in the context of the microglia, and we extrapolate these findings to what is known in neurons. We further discuss the emerging evidence for dysregulation of the Rho GTPase pathway in CNS diseases and the consequences contributed by the mis-splicing of microexons. This article is categorized under: RNA Processing > Splicing Mechanisms RNA Processing > Splicing Regulation/Alternative Splicing RNA in Disease and Development > RNA in Disease.


Subject(s)
Central Nervous System Diseases , Monomeric GTP-Binding Proteins , Alternative Splicing , Central Nervous System Diseases/genetics , Humans , RNA , RNA Splicing , RNA-Binding Proteins
7.
Commun Biol ; 4(1): 1042, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493786

ABSTRACT

High mortality of prostate cancer patients is primarily due to metastasis. Understanding the mechanisms controlling metastatic processes remains essential to develop novel therapies designed to prevent the progression from localized disease to metastasis. CdGAP plays important roles in the control of cell adhesion, migration, and proliferation, which are central to cancer progression. Here we show that elevated CdGAP expression is associated with early biochemical recurrence and bone metastasis in prostate cancer patients. Knockdown of CdGAP in metastatic castration-resistant prostate cancer (CRPC) PC-3 and 22Rv1 cells reduces cell motility, invasion, and proliferation while inducing apoptosis in CdGAP-depleted PC-3 cells. Conversely, overexpression of CdGAP in DU-145, 22Rv1, and LNCaP cells increases cell migration and invasion. Using global gene expression approaches, we found that CdGAP regulates the expression of genes involved in epithelial-to-mesenchymal transition, apoptosis and cell cycle progression. Subcutaneous injection of CdGAP-depleted PC-3 cells into mice shows a delayed tumor initiation and attenuated tumor growth. Orthotopic injection of CdGAP-depleted PC-3 cells reduces distant metastasic burden. Collectively, these findings support a pro-oncogenic role of CdGAP in prostate tumorigenesis and unveil CdGAP as a potential biomarker and target for prostate cancer treatments.


Subject(s)
Apoptosis , Cell Cycle , Epithelial-Mesenchymal Transition , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Animals , Male , Mice , Mice, Nude
8.
Nat Commun ; 12(1): 4688, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344896

ABSTRACT

Internalization and intracellular trafficking of G protein-coupled receptors (GPCRs) play pivotal roles in cell responsiveness. Dysregulation in receptor trafficking can lead to aberrant signaling and cell behavior. Here, using an endosomal BRET-based assay in a high-throughput screen with the prototypical GPCR angiotensin II type 1 receptor (AT1R), we sought to identify receptor trafficking inhibitors from a library of ~115,000 small molecules. We identified a novel dual Ras and ARF6 inhibitor, which we named Rasarfin, that blocks agonist-mediated internalization of AT1R and other GPCRs. Rasarfin also potently inhibits agonist-induced ERK1/2 signaling by GPCRs, and MAPK and Akt signaling by EGFR, as well as prevents cancer cell proliferation. In silico modeling and in vitro studies reveal a unique binding modality of Rasarfin within the SOS-binding domain of Ras. Our findings unveil a class of dual small G protein inhibitors for receptor trafficking and signaling, useful for the inhibition of oncogenic cellular responses.


Subject(s)
ADP-Ribosylation Factors/antagonists & inhibitors , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Receptors, G-Protein-Coupled/metabolism , ras Proteins/antagonists & inhibitors , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Binding Sites , Bioluminescence Resonance Energy Transfer Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Molecular Dynamics Simulation , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , ras Proteins/chemistry , ras Proteins/metabolism
9.
Cell Rep ; 33(13): 108560, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378678

ABSTRACT

The role of RNA binding proteins in regulating the phagocytic and cytokine-releasing functions of microglia is unknown. Here, we show that microglia deficient for the QUAKING (QKI) RNA binding protein have increased proinflammatory cytokine release and defects in processing phagocytosed cargo. Splicing analysis reveals a role for QKI in regulating microexon networks of the Rho GTPase pathway. We show an increase in RhoA activation and proinflammatory cytokines in QKI-deficient microglia that are repressed by treating with a Rock kinase inhibitor. During the cuprizone diet, mice with QKI-deficient microglia are inefficient at supporting central nervous system (CNS) remyelination and cause the recruited oligodendrocyte precursor cells to undergo apoptosis. Furthermore, the expression of QKI in microglia is downregulated in preactive, chronic active, and remyelinating white matter lesions of multiple sclerosis (MS) patients. Overall, our findings identify QKI as an alternative splicing regulator governing a network of Rho GTPase microexons with implications for CNS remyelination and MS patients.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Microglia/physiology , RNA-Binding Proteins/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Central Nervous System/metabolism , Cytokines/metabolism , Female , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Multiple Sclerosis/genetics , Phagocytosis , RNA/metabolism , RNA-Seq , Remyelination , Signal Transduction/drug effects , rho-Associated Kinases/metabolism
10.
Cancer Lett ; 483: 98-113, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32217106

ABSTRACT

Liver metastases remain a major cause of death from gastrointestinal tract cancers and other malignancies, such as breast and lung carcinomas. Understanding the underlying biology is essential for the design of effective therapies. We previously identified the chemokine CCL7 and its receptor CCR3 as critical mediators of invasion and metastasis in lung and colon carcinoma cells. Here we show that the CCL7/CCR3 axis regulates a late stage in invadopodia genesis namely, the targeting of MMP-9 to the invadopodia complex, thereby promoting invadopodia maturation and collagen degradation. We show that this process could be blocked by overexpression of a dominant negative RhoA in highly invasive cells, while a constitutively active RhoA upregulated invadopodia maturation in CCL7-silenced and poorly invasive and metastatic cells and also enhanced their metastatic potential in vivo, collectively, implicating RhoA activation in signaling downstream of CCL7. Blockade of the ERK or PI3K pathways by chemical inhibitors also inhibited invadopodia formation, but affected the initiation stage of invadopodia genesis. Our data implicate CCL7/CCR3 signaling in invadopodia maturation and suggest that chemokine signaling acts in concert with extracellular matrix-initiated signals to promote invasion and liver metastasis.


Subject(s)
Carcinoma, Lewis Lung/enzymology , Cell Movement , Chemokine CCL7/metabolism , Collagen/metabolism , Colonic Neoplasms/enzymology , Liver Neoplasms/enzymology , Matrix Metalloproteinase 9/metabolism , Podosomes/enzymology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Chemokine CCL7/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Matrix Metalloproteinase 9/genetics , Mice , Phosphatidylinositol 3-Kinase/metabolism , Podosomes/genetics , Podosomes/pathology , Protein Transport , Proteolysis , Receptors, CCR3/genetics , Receptors, CCR3/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
11.
J Neurochem ; 152(3): 315-332, 2020 02.
Article in English | MEDLINE | ID: mdl-31344270

ABSTRACT

During development, neurons extend axons toward their appropriate synaptic targets to establish functional neuronal connections. The growth cone, a highly motile structure at the tip of the axon, is capable of recognizing extracellular guidance cues and translating them into directed axon outgrowth through modulation of the actin cytoskeleton. Netrin-1 mediates its attractive function through the receptor deleted in colorectal cancer (DCC) to promote axon outgrowth and guidance. The calcium-activated protease calpain is involved in the cleavage of cytoskeletal proteins, which plays an important role during adhesion turnover and cell migration. However, its function during neuronal development is less understood. Here we demonstrate that netrin-1 activated calpain in embryonic rat cortical neurons in an extracellular-regulated kinase 1/2-dependent manner. In addition, we found that netrin-1 stimulation led to an increase in calpain-1 localization in the axon, whereas its endogenous inhibitor calpastatin was decreased in the growth cones of cortical neurons by indirect immunofluorescence. Interestingly, calpain-1 was able to cleave DCC in vitro. Furthermore, netrin-1 induced the cleavage of the cytoskeletal proteins spectrin and focal adhesion kinase concomitantly with the intracellular domain of DCC in a calpain-dependent manner in embryonic rat cortical neurons. Cortical neurons over-expressing calpastatin or calpain-depleted neurons displayed increased basal axon length and were unresponsive to netrin-1 stimulation. Altogether, we propose a novel model whereby netrin-1/DCC-mediated axon outgrowth is modulated by calpain-mediated proteolysis of DCC and cytoskeletal targets in embryonic cortical neurons. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.


Subject(s)
Calpain/metabolism , DCC Receptor/metabolism , Neurogenesis/physiology , Neuronal Outgrowth/physiology , Neurons , Animals , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Growth Cones/metabolism , Netrin-1/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
12.
Int J Mol Sci ; 20(6)2019 Mar 25.
Article in English | MEDLINE | ID: mdl-30934641

ABSTRACT

One of the fundamental steps during development of the nervous system is the formation of proper connections between neurons and their target cells-a process called neural wiring, failure of which causes neurological disorders ranging from autism to Down's syndrome. Axons navigate through the complex environment of a developing embryo toward their targets, which can be far away from their cell bodies. Successful implementation of neuronal wiring, which is crucial for fulfillment of all behavioral functions, is achieved through an intimate interplay between axon guidance and neural activity. In this review, our focus will be on axon pathfinding and the implication of some of its downstream molecular components in neurological disorders. More precisely, we will talk about axon guidance and the molecules implicated in this process. After, we will briefly review the Rho family of small GTPases, their regulators, and their involvement in downstream signaling pathways of the axon guidance cues/receptor complexes. We will then proceed to the final and main part of this review, where we will thoroughly comment on the implication of the regulators for Rho GTPases-GEFs (Guanine nucleotide Exchange Factors) and GAPs (GTPase-activating Proteins)-in neurological diseases and disorders.


Subject(s)
Axon Guidance , Nervous System Diseases/enzymology , Nervous System/enzymology , rho GTP-Binding Proteins/metabolism , Animals , Humans , Models, Biological
13.
Oncogene ; 37(28): 3790-3805, 2018 07.
Article in English | MEDLINE | ID: mdl-29651051

ABSTRACT

Liver metastases remain a major cause of death from gastrointestinal tract cancers as well as from other malignancies such as breast and lung carcinomas and melanoma. Understanding the underlying biology is essential for the design of effective targeted therapies. We previously reported that collagen IV α1/α2 overexpression in non-metastatic lung carcinoma (M27colIV) cells increased their metastatic ability, specifically to the liver and documented high collagen IV levels in surgical resections of liver metastases from diverse tumor types. Here, we aimed to elucidate the functional relevance of collagen IV to metastatic outgrowth in the liver. Gene expression profiling revealed in M27colIVcells significant increases in the expression of chemokines CCL5 (5.7-fold) and CCL7 (2.6-fold) relative to wild-type cells, and this was validated by qPCR and western blotting. Similarly, in human colon carcinoma KM12C and KM12SM cells with divergent liver-colonizing potentials, CCL7 and CCL5 production correlated with type IV collagen expression and the metastatic phenotype. CCL7 silencing by short hairpin RNA (shRNA) reduced experimental liver metastasis in both cell types, whereas CCL5 silencing reduced metastasis of M27colIV cells, implicating these cytokines in metastatic expansion in the liver. Subsequent functional analyses implicated both MEK/ERK and PI3K signaling upstream of CCL7 upregulation and identified CCL7 (but not CCL5) as a critical migration/invasion factor, acting via the chemokine receptor CCR3. Chemokine CCL5 was identified as a regulator of the T-cell immune response in the liver. Loss of CCL7 in KM12SM cells was also associated with altered E-cadherin and reduced vimentin and Snail expression, implicating it in epithelial-to-mesenchymal transition in these cells. Moreover, in clinical specimens of colon cancer liver metastases analyzed by immunohistochemistry, CCL5 and CCL7 levels paralleled those of collagen IV. The results identify the chemokines CCL5 and CCL7 as type IV collagen-regulated genes that promote liver metastasis by distinct and complementary mechanisms.


Subject(s)
Chemokine CCL5/metabolism , Chemokine CCL7/metabolism , Collagen Type IV/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Animals , Cadherins/metabolism , Carcinoma, Lewis Lung , Cell Line, Tumor , Cell Movement/physiology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Snail Family Transcription Factors/metabolism , Up-Regulation/physiology , Vimentin/metabolism
14.
Oncotarget ; 9(14): 11646-11664, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29545927

ABSTRACT

Cdc42 GTPase-activating protein (CdGAP, also named ARHGAP31) is a negative regulator of the GTPases Rac1 and Cdc42. Associated with the rare developmental disorder Adams-Oliver Syndrome (AOS), CdGAP is critical for embryonic vascular development and VEGF-mediated angiogenesis. Moreover, CdGAP is an essential component in the synergistic interaction between TGFß and ErbB-2 signaling pathways during breast cancer cell migration and invasion, and is a novel E-cadherin transcriptional co-repressor with Zeb2 in breast cancer. CdGAP is highly phosphorylated on serine and threonine residues in response to growth factors and is a substrate of ERK1/2 and GSK-3. Here, we identified Ser1093 and Ser1163 in the C-terminal region of CdGAP, which are phosphorylated by RSK in response to phorbol ester. These phospho-residues create docking sites for binding to 14-3-3 adaptor proteins. The interaction between CdGAP and 14-3-3 proteins inhibits the GAP activity of CdGAP and sequesters CdGAP into the cytoplasm. Consequently, the nucleocytoplasmic shuttling of CdGAP is inhibited and CdGAP-induced cell rounding is abolished. In addition, 14-3-3ß inhibits the ability of CdGAP to repress the E-cadherin promoter and to induce cell migration. Finally, we show that 14-3-3ß is unable to regulate the activity and subcellular localization of the AOS-related mutant proteins lacking these phospho-residues. Altogether, we provide a novel mechanism of regulation of CdGAP activity and localization, which impacts directly on a better understanding of the role of CdGAP as a promoter of breast cancer and in the molecular causes of AOS.

15.
FASEB J ; 31(8): 3467-3483, 2017 08.
Article in English | MEDLINE | ID: mdl-28442549

ABSTRACT

Gangliosides (sialylated glycolipids) play an essential role in the CNS by regulating recognition and signaling in neurons. Metabolic blocks in processing and catabolism of gangliosides result in the development of severe neurologic disorders, including gangliosidoses manifesting with neurodegeneration and neuroinflammation. We demonstrate that 2 mammalian enzymes, neuraminidases 3 and 4, play important roles in catabolic processing of brain gangliosides by cleaving terminal sialic acid residues in their glycan chains. In neuraminidase 3 and 4 double-knockout mice, GM3 ganglioside is stored in microglia, vascular pericytes, and neurons, causing micro- and astrogliosis, neuroinflammation, accumulation of lipofuscin bodies, and memory loss, whereas their cortical and hippocampal neurons have lower rate of neuritogenesis in vitro Double-knockout mice also have reduced levels of GM1 ganglioside and myelin in neuronal axons. Furthermore, neuraminidase 3 deficiency drastically increased storage of GM2 in the brain tissues of an asymptomatic mouse model of Tay-Sachs disease, a severe human gangliosidosis, indicating that this enzyme is responsible for the metabolic bypass of ß-hexosaminidase A deficiency. Together, our results provide the first in vivo evidence that neuraminidases 3 and 4 have important roles in CNS function by catabolizing gangliosides and preventing their storage in lipofuscin bodies.-Pan, X., De Britto Pará De Aragão, C., Velasco-Martin, J. P., Priestman, D. A., Wu, H. Y., Takahashi, K., Yamaguchi, K., Sturiale, L., Garozzo, D., Platt, F. M., Lamarche-Vane, N., Morales, C. R., Miyagi, T., Pshezhetsky, A. V. Neuraminidases 3 and 4 regulate neuronal function by catabolizing brain gangliosides.


Subject(s)
Brain/metabolism , Gangliosides/metabolism , Neuraminidase/metabolism , Neurons/physiology , Animals , Brain/pathology , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Enzymologic , Mice , Mice, Knockout , Motor Activity/physiology , Mucolipidoses/metabolism , Neuraminidase/genetics
16.
Nat Commun ; 7: 13340, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27827368

ABSTRACT

Haploinsufficiency of the SYNGAP1 gene, which codes for a Ras GTPase-activating protein, impairs cognition both in humans and in mice. Decrease of Syngap1 in mice has been previously shown to cause cognitive deficits at least in part by inducing alterations in glutamatergic neurotransmission and premature maturation of excitatory connections. Whether Syngap1 plays a role in the development of cortical GABAergic connectivity and function remains unclear. Here, we show that Syngap1 haploinsufficiency significantly reduces the formation of perisomatic innervations by parvalbumin-positive basket cells, a major population of GABAergic neurons, in a cell-autonomous manner. We further show that Syngap1 haploinsufficiency in GABAergic cells derived from the medial ganglionic eminence impairs their connectivity, reduces inhibitory synaptic activity and cortical gamma oscillation power, and causes cognitive deficits. Our results indicate that Syngap1 plays a critical role in GABAergic circuit function and further suggest that Syngap1 haploinsufficiency in GABAergic circuits may contribute to cognitive deficits.


Subject(s)
Cognition Disorders/genetics , Cognition/physiology , GABAergic Neurons/physiology , Synapses/physiology , ras GTPase-Activating Proteins/physiology , Animals , Cells, Cultured , Disease Models, Animal , Female , Gene Knockdown Techniques , Haploinsufficiency , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Synaptic Transmission/physiology , ras GTPase-Activating Proteins/genetics
17.
Sci Rep ; 6: 27485, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27270835

ABSTRACT

Mutations in the CdGAP/ARHGAP31 gene, which encodes a GTPase-activating protein for Rac1 and Cdc42, have been reported causative in the Adams-Oliver developmental syndrome often associated with vascular defects. However, despite its abundant expression in endothelial cells, CdGAP function in the vasculature remains unknown. Here, we show that vascular development is impaired in CdGAP-deficient mouse embryos at E15.5. This is associated with superficial vessel defects and subcutaneous edema, resulting in 44% embryonic/perinatal lethality. VEGF-driven angiogenesis is defective in CdGAP(-/-) mice, showing reduced capillary sprouting from aortic ring explants. Similarly, VEGF-dependent endothelial cell migration and capillary formation are inhibited upon CdGAP knockdown. Mechanistically, CdGAP associates with VEGF receptor-2 and controls VEGF-dependent signaling. Consequently, CdGAP depletion results in impaired VEGF-mediated Rac1 activation and reduced phosphorylation of critical intracellular mediators including Gab1, Akt, PLCγ and SHP2. These findings are the first to demonstrate the importance of CdGAP in embryonic vascular development and VEGF-induced signaling, and highlight CdGAP as a potential therapeutic target to treat pathological angiogenesis and vascular dysfunction.


Subject(s)
Blood Vessels/embryology , GTPase-Activating Proteins/physiology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/physiology , cdc42 GTP-Binding Protein/physiology , Animals , Mice , Mice, Knockout
18.
PLoS Genet ; 12(2): e1005785, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26859289

ABSTRACT

Rho family GTPases act as molecular switches regulating actin cytoskeleton dynamics. Attenuation of their signaling capacity is provided by GTPase-activating proteins (GAPs), including p190A, that promote the intrinsic GTPase activity of Rho proteins. In the current study we have performed a small-scale ENU mutagenesis screen and identified a novel loss of function allele of the p190A gene Arhgap35, which introduces a Leu1396 to Gln substitution in the GAP domain. This results in decreased GAP activity for the prototypical Rho-family members, RhoA and Rac1, likely due to disrupted ordering of the Rho binding surface. Consequently, Arhgap35-deficient animals exhibit hypoplastic and glomerulocystic kidneys. Investigation into the cystic phenotype shows that p190A is required for appropriate primary cilium formation in renal nephrons. P190A specifically localizes to the base of the cilia to permit axoneme elongation, which requires a functional GAP domain. Pharmacological manipulations further reveal that inhibition of either Rho kinase (ROCK) or F-actin polymerization is able to rescue the ciliogenesis defects observed upon loss of p190A activity. We propose a model in which p190A acts as a modulator of Rho GTPases in a localized area around the cilia to permit the dynamic actin rearrangement required for cilia elongation. Together, our results establish an unexpected link between Rho GTPase regulation, ciliogenesis and glomerulocystic kidney disease.


Subject(s)
Cilia/metabolism , GTPase-Activating Proteins/genetics , Kidney Diseases, Cystic/genetics , Kidney Glomerulus/pathology , Organogenesis , Point Mutation/genetics , Repressor Proteins/genetics , Actins/metabolism , Alleles , Amino Acid Sequence , Amino Acid Substitution , Animals , Cytoskeleton/metabolism , Embryo, Mammalian/cytology , Ethylnitrosourea , Female , Fibroblasts/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Kidney Diseases, Cystic/pathology , Kidney Glomerulus/metabolism , Kidney Tubules/abnormalities , Kidney Tubules/pathology , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Neural Tube Defects/pathology , Phenotype , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Reproducibility of Results
19.
Biol Cell ; 108(5): 115-26, 2016 May.
Article in English | MEDLINE | ID: mdl-26787017

ABSTRACT

BACKGROUND INFORMATION: Rho GTPases play an essential role during the development of the nervous system. They induce cytoskeletal rearrangements that are critical for the regulation of axon outgrowth and guidance. It is generally accepted that Rac1 and Cdc42 are positive regulators of axon outgrowth and guidance, whereas RhoA is a negative regulator. However, spatiotemporal control of their activity can modify the function of Rho GTPases during axonal morphogenesis. Signalling downstream of the axon guidance cue netrin-1 and its receptor deleted in colorectal cancer (DCC) triggers the activation of Rac1 and the inhibition of RhoA to promote axon outgrowth. However, our previous work also suggests that netrin-1/DCC signalling can activate RhoA in a time- and region-specific manner. RESULTS: Here, we visualised RhoA activation in response to netrin-1 in live embryonic cortical neurons using fluorescence resonance energy transfer. RhoA activity oscillated in unstimulated neurons and netrin-1 increased the amplitude of the oscillations in growth cones after 5 min of stimulation. Within this period of time, netrin-1 transiently increased RhoA activity and modulated the pattern of RhoA oscillations. We found that the timing of netrin-1-induced RhoA activation was different in whole neurons, cell bodies and growth cones. CONCLUSIONS: We conclude that netrin-1 modulates the spatiotemporal activation of RhoA in embryonic cortical neurons. SIGNIFICANCE: This study demonstrates for the first time the short-term localised activation of RhoA in neuronal growth cones by the axon guidance cue netrin-1.


Subject(s)
Axons/metabolism , Cerebral Cortex/metabolism , Embryo, Mammalian/metabolism , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian/cytology , Fluorescence Resonance Energy Transfer , Netrin-1 , Rats
20.
J Biol Chem ; 291(9): 4589-602, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26710849

ABSTRACT

The receptor deleted in colorectal cancer (DCC) mediates the attraction of growing axons to netrin-1 during brain development. In response to netrin-1 stimulation, DCC becomes a signaling platform to recruit proteins that promote axon outgrowth and guidance. The Ras GTPase-activating protein (GAP) p120RasGAP inhibits Ras activity and mediates neurite retraction and growth cone collapse in response to repulsive guidance cues. Here we show an interaction between p120RasGAP and DCC that positively regulates netrin-1-mediated axon outgrowth and guidance in embryonic cortical neurons. In response to netrin-1, p120RasGAP is recruited to DCC in growth cones and forms a multiprotein complex with focal adhesion kinase and ERK. We found that Ras/ERK activities are elevated aberrantly in p120RasGAP-deficient neurons. Moreover, the expression of p120RasGAP Src homology 2 (SH2)-SH3-SH2 domains, which interact with the C-terminal tail of DCC, is sufficient to restore netrin-1-dependent axon outgrowth in p120RasGAP-deficient neurons. We provide a novel mechanism that exploits the scaffolding properties of the N terminus of p120RasGAP to tightly regulate netrin-1/DCC-dependent axon outgrowth and guidance.


Subject(s)
Axons/metabolism , Cerebral Cortex/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Receptors, Cell Surface/agonists , Signal Transduction , Tumor Suppressor Proteins/agonists , Tumor Suppressor Proteins/metabolism , p120 GTPase Activating Protein/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Cerebral Cortex/cytology , Chickens , DCC Receptor , Embryo, Mammalian/cytology , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Mutant Proteins/agonists , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Netrin-1 , Neurons/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , Rats , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , p120 GTPase Activating Protein/antagonists & inhibitors , p120 GTPase Activating Protein/chemistry , p120 GTPase Activating Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL