Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G627-G643, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33566751

ABSTRACT

Rho guanine nucleotide exchange factors (RhoGEFs) regulate Rho GTPase activity and cytoskeletal and cell adhesion dynamics. ßPix, a CDC42/RAC family RhoGEF encoded by ARHGEF7, is reported to modulate human colon cancer cell proliferation and postwounding restitution of rat intestinal epithelial monolayers. We hypothesized that ßPix plays a role in maintaining intestinal epithelial homeostasis. To test this hypothesis, we examined ßPix distribution in the human and murine intestine and created mice with intestinal epithelial-selective ßPix deletion [ßPixflox/flox/Tg(villin-Cre); Arhgef7 CKO mice]. Using Arhgef7 conditional knockout (CKO) and control mice, we investigated the consequences of ßPix deficiency in vivo on intestinal epithelial and enteroid development, dextran sodium sulfate-induced mucosal injury, and gut permeability. In normal human and murine intestines, we observed diffuse cytoplasmic and moderate nuclear ßPix immunostaining in enterocytes. Arhgef7 CKO mice were viable and fertile, with normal gross intestinal architecture but reduced small intestinal villus height, villus-to-crypt ratio, and goblet cells; small intestinal crypt cells had reduced Ki67 staining, compatible with impaired cell proliferation. Enteroids derived from control mouse small intestine were viable for more than 20 passages, but those from Arhgef7 CKO mice did not survive beyond 24 h despite addition of Wnt proteins or conditioned media from normal enteroids. Adding a Rho kinase (ROCK) inhibitor partially rescued CKO enteroid development. Compared with littermate control mice, dextran sodium sulfate-treated ßPix-deficient mice lost more weight and had greater impairment of intestinal barrier function, and more severe colonic mucosal injury. These findings reveal ßPix expression is important for enterocyte development, intestinal homeostasis, and resistance to toxic injury.NEW & NOTEWORTHY To explore the role of ßPix, a guanine nucleotide exchange factor encoded by ARHGEF7, in intestinal development and physiology, we created mice with intestinal epithelial cell Arhgef7/ßPix deficiency. We found ßPix essential for normal small intestinal epithelial cell proliferation, villus development, and mucosal resistance to injury. Moreover, Rho kinase signaling mediated developmental arrest observed in enteroids derived from ßPix-deficient small intestinal crypts. Our studies provide insights into the role Arhgef7/ßPix plays in intestinal epithelial homeostasis.


Subject(s)
Cell Proliferation , Colitis/metabolism , Colon/metabolism , Enterocytes/metabolism , Intestinal Mucosa/metabolism , Microvilli/metabolism , Rho Guanine Nucleotide Exchange Factors/deficiency , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Enterocytes/pathology , Female , Gene Deletion , Humans , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microvilli/pathology , Organoids , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Tissue Culture Techniques , rho-Associated Kinases/metabolism
2.
Basic Res Cardiol ; 115(6): 60, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32910221

ABSTRACT

Obscurin comprises a family of giant modular proteins that play key structural and regulatory roles in striated muscles. Immunoglobulin domains 58/59 (Ig58/59) of obscurin mediate binding to essential modulators of muscle structure and function, including canonical titin, a smaller splice variant of titin, termed novex-3, and phospholamban (PLN). Importantly, missense mutations localized within the obscurin-Ig58/59 region that affect binding to titins and/or PLN have been linked to the development of myopathy in humans. To elucidate the pathophysiological role of this region, we generated a constitutive deletion mouse model, Obscn-ΔIg58/59, that expresses obscurin lacking Ig58/59, and determined the consequences of this manipulation on cardiac morphology and function under conditions of acute stress and through the physiological process of aging. Our studies show that young Obscn-ΔIg58/59 mice are susceptible to acute ß-adrenergic stress. Moreover, sedentary Obscn-ΔIg58/59 mice develop left ventricular hypertrophy that progresses to dilation, contractile impairment, atrial enlargement, and arrhythmia as a function of aging with males being more affected than females. Experiments in ventricular cardiomyocytes revealed altered Ca2+ cycling associated with changes in the expression and/or phosphorylation levels of major Ca2+ cycling proteins, including PLN, SERCA2, and RyR2. Taken together, our work demonstrates that obscurin-Ig58/59 is an essential regulatory module in the heart and its deletion leads to age- and sex-dependent cardiac remodeling, ventricular dilation, and arrhythmia due to deregulated Ca2+ cycling.


Subject(s)
Arrhythmias, Cardiac/enzymology , Heart Rate , Hypertrophy, Left Ventricular/enzymology , Myocytes, Cardiac/enzymology , Protein Serine-Threonine Kinases/deficiency , Rho Guanine Nucleotide Exchange Factors/deficiency , Ventricular Dysfunction, Left/enzymology , Ventricular Function, Left , Ventricular Remodeling , Action Potentials , Age Factors , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Calcium Signaling , Calcium-Binding Proteins/metabolism , Female , Gene Deletion , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Immunoglobulin Domains , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sedentary Behavior , Sex Factors , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
3.
J Cell Biol ; 219(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32379884

ABSTRACT

Cells navigating through complex tissues face a fundamental challenge: while multiple protrusions explore different paths, the cell needs to avoid entanglement. How a cell surveys and then corrects its own shape is poorly understood. Here, we demonstrate that spatially distinct microtubule dynamics regulate amoeboid cell migration by locally promoting the retraction of protrusions. In migrating dendritic cells, local microtubule depolymerization within protrusions remote from the microtubule organizing center triggers actomyosin contractility controlled by RhoA and its exchange factor Lfc. Depletion of Lfc leads to aberrant myosin localization, thereby causing two effects that rate-limit locomotion: (1) impaired cell edge coordination during path finding and (2) defective adhesion resolution. Compromised shape control is particularly hindering in geometrically complex microenvironments, where it leads to entanglement and ultimately fragmentation of the cell body. We thus demonstrate that microtubules can act as a proprioceptive device: they sense cell shape and control actomyosin retraction to sustain cellular coherence.


Subject(s)
Actomyosin/metabolism , Cell Movement/physiology , Dendritic Cells/cytology , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Adhesion/physiology , Cell Shape/physiology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Organizing Center/drug effects , Microtubules/drug effects , Nocodazole/pharmacology , Protein Binding , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics
4.
J Am Soc Nephrol ; 31(5): 996-1008, 2020 05.
Article in English | MEDLINE | ID: mdl-32188698

ABSTRACT

BACKGROUND: Previous studies showed that Cdc42, a member of the prototypical Rho family of small GTPases and a regulator of the actin cytoskeleton, is critical for the normal development and health of podocytes. However, upstream regulatory mechanisms for Cdc42 activity in podocytes are largely unknown. METHODS: We used a proximity-based ligation assay, BioID, to identify guanine nucleotide exchange factors that activate Cdc42 in immortalized human podocytes. We generated podocyte-specific ARHGEF7 (commonly known as ß-PIX) knockout mice by crossing ß-PIX floxed mice with Podocin-Cre mice. Using shRNA, we established cultured mouse podocytes with ß-PIX knockdown and their controls. RESULTS: We identified ß-PIX as a predominant guanine nucleotide exchange factor that interacts with Cdc42 in human podocytes. Podocyte-specific ß-PIX knockout mice developed progressive proteinuria and kidney failure with global or segmental glomerulosclerosis in adulthood. Glomerular podocyte density gradually decreased in podocyte-specific ß-PIX knockout mice, indicating podocyte loss. Compared with controls, glomeruli from podocyte-specific ß-PIX knockout mice and cultured mouse podocytes with ß-PIX knockdown exhibited significant reduction in Cdc42 activity. Loss of ß-PIX promoted podocyte apoptosis, which was mediated by the reduced activity of the prosurvival transcriptional regulator Yes-associated protein. CONCLUSIONS: These findings indicate that ß-PIX is required for the maintenance of podocyte architecture and glomerular function via Cdc42 and its downstream Yes-associated protein activities. This appears to be the first evidence that a Rho-guanine nucleotide exchange factor plays a critical role in podocytes.


Subject(s)
Podocytes/metabolism , Rho Guanine Nucleotide Exchange Factors/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Crosses, Genetic , Enzyme Activation , Female , Gene Knockdown Techniques , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Lipopolysaccharides/toxicity , Mice , Mice, 129 Strain , Mice, Inbred ICR , Podocytes/physiology , Podocytes/ultrastructure , Proteinuria/etiology , Proteinuria/metabolism , Proteinuria/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rho Guanine Nucleotide Exchange Factors/deficiency , Signal Transduction , YAP-Signaling Proteins , cdc42 GTP-Binding Protein/metabolism
5.
Int J Mol Sci ; 21(4)2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32075101

ABSTRACT

Inflammasomes are intracellular multiple protein complexes that mount innate immune responses to tissue damage and invading pathogens. Their excessive activation is crucial in the development and pathogenesis of inflammatory disorders. Microtubules have been reported to provide the platform for mediating the assembly and activation of NLRP3 inflammasome. Recently, we have identified the microtubule-associated immune molecule guanine nucleotide exchange factor-H1 (GEF-H1) that is crucial in coupling microtubule dynamics to the initiation of microtubule-mediated immune responses. However, whether GEF-H1 also controls the activation of other immune receptors that require microtubules is still undefined. Here we employed GEF-H1-deficient mouse bone marrow-derived macrophages (BMDMs) to interrogate the impact of GEF-H1 on the activation of NLRP3 inflammasome. NLRP3 but not NLRC4 or AIM2 inflammasome-mediated IL-1ß production was dependent on dynamic microtubule network in wild-type (WT) BMDMs. However, GEF-H1 deficiency did not affect NLRP3-driven IL-1ß maturation and secretion in macrophages. Moreover, α-tubulin acetylation and mitochondria aggregations were comparable between WT and GEF-H1-deficient BMDMs in response to NLRP3 inducers. Further, GEF-H1 was not required for NLRP3-mediated immune defense against Salmonella typhimurium infection. Collectively, these findings suggest that the microtubule-associated immune modulator GEF-H1 is dispensable for microtubule-mediated NLRP3 activation and host defense in mouse macrophages.


Subject(s)
Inflammasomes/metabolism , Macrophages/metabolism , Microtubules/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Acetylation , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Immunity, Innate , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Nigericin/pharmacology , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Salmonella Infections/immunology , Salmonella Infections/pathology , Salmonella typhimurium/pathogenicity
6.
Elife ; 82019 09 24.
Article in English | MEDLINE | ID: mdl-31549965

ABSTRACT

Myogenic vasoconstriction is an autoregulatory function of small arteries. Recently, G-protein-coupled receptors have been involved in myogenic vasoconstriction, but the downstream signalling mechanisms and the in-vivo-function of this myogenic autoregulation are poorly understood. Here, we show that small arteries from mice with smooth muscle-specific loss of G12/G13 or the Rho guanine nucleotide exchange factor ARHGEF12 have lost myogenic vasoconstriction. This defect was accompanied by loss of RhoA activation, while vessels showed normal increases in intracellular [Ca2+]. In the absence of myogenic vasoconstriction, perfusion of peripheral organs was increased, systemic vascular resistance was reduced and cardiac output and left ventricular mass were increased. In addition, animals with defective myogenic vasoconstriction showed aggravated hypotension in response to endotoxin. We conclude that G12/G13- and Rho-mediated signaling plays a key role in myogenic vasoconstriction and that myogenic tone is required to maintain local and systemic vascular resistance under physiological and pathological condition.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Vascular Resistance , Vasoconstriction , Animals , GTP-Binding Protein alpha Subunits, G12-G13/deficiency , Mice, Inbred C57BL , Rho Guanine Nucleotide Exchange Factors/deficiency
7.
J Am Heart Assoc ; 8(9): e011712, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30994039

ABSTRACT

Background Platelets are the cellular mediators of hemostasis and thrombosis, and their function is regulated by a number of molecular mediators, such as small GTP ases. These small GTP ases are themselves regulated by guanine nucleotide exchange factors such as Arhgefs, several of which are found in platelets, including the highly expressed Arhgef1. However, the role of Arhgef1 in platelets has not yet been investigated. Methods and Results We employed mice with genetic deletion of Arhgef1 (ie, Arhgef1-/-) and investigated their platelet phenotype by employing a host of in vivo and in vitro platelet assays. Our results indicate that Arhgef1-/- mice had prolonged carotid artery occlusion and tail bleeding times. Moreover, platelets from these mice exhibited defective aggregation, dense and α granule secretion, α II bß3 integrin activation, clot retraction and spreading, in comparison to their wild-type littermates. Finally, we also found that the mechanism by which Arhgef1 regulates platelets is mediated in part by a defect in the activation of the RhoA-Rho-associated kinase axis, but not Rap1b. Conclusions Our data demonstrate, for the first time, that Arhgef1 plays a critical role in platelet function, in vitro and in vivo.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Platelet Activation , Rho Guanine Nucleotide Exchange Factors/blood , Thrombosis/blood , Animals , Blood Coagulation/genetics , Disease Models, Animal , Mice, Knockout , Phenotype , Phosphorylation , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Thrombosis/genetics , rho-Associated Kinases/blood , rhoA GTP-Binding Protein/blood
8.
J Clin Invest ; 129(3): 1047-1060, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30521495

ABSTRACT

ARHGEF1 is a RhoA-specific guanine nucleotide exchange factor expressed in hematopoietic cells. We used whole-exome sequencing to identify compound heterozygous mutations in ARHGEF1, resulting in the loss of ARHGEF1 protein expression in 2 primary antibody-deficient siblings presenting with recurrent severe respiratory tract infections and bronchiectasis. Both ARHGEF1-deficient patients showed an abnormal B cell immunophenotype, with a deficiency in marginal zone and memory B cells and an increased frequency of transitional B cells. Furthermore, the patients' blood contained immature myeloid cells. Analysis of a mediastinal lymph node from one patient highlighted the small size of the germinal centers and an abnormally high plasma cell content. On the molecular level, T and B lymphocytes from both patients displayed low RhoA activity and low steady-state actin polymerization (even after stimulation of lysophospholipid receptors). As a consequence of disturbed regulation of the RhoA downstream target Rho-associated kinase I/II (ROCK), the patients' lymphocytes failed to efficiently restrain AKT phosphorylation. Enforced ARHGEF1 expression or drug-induced activation of RhoA in the patients' cells corrected the impaired actin polymerization and AKT regulation. Our results indicate that ARHGEF1 activity in human lymphocytes is involved in controlling actin cytoskeleton dynamics, restraining PI3K/AKT signaling, and confining B lymphocytes and myelocytes within their dedicated functional environment.


Subject(s)
B-Lymphocytes , Primary Immunodeficiency Diseases , Signal Transduction , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Female , Germinal Center/immunology , Germinal Center/pathology , Humans , Immunologic Memory/genetics , Male , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/immunology , Siblings , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , rho-Associated Kinases/genetics , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/genetics
9.
J Cell Biol ; 218(1): 350-379, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30523041

ABSTRACT

Growth cones are complex, motile structures at the tip of an outgrowing neurite. They often exhibit a high density of filopodia (thin actin bundles), which complicates the unbiased quantification of their morphologies by software. Contemporary image processing methods require extensive tuning of segmentation parameters, require significant manual curation, and are often not sufficiently adaptable to capture morphology changes associated with switches in regulatory signals. To overcome these limitations, we developed Growth Cone Analyzer (GCA). GCA is designed to quantify growth cone morphodynamics from time-lapse sequences imaged both in vitro and in vivo, but is sufficiently generic that it may be applied to nonneuronal cellular structures. We demonstrate the adaptability of GCA through the analysis of growth cone morphological variation and its relation to motility in both an unperturbed system and in the context of modified Rho GTPase signaling. We find that perturbations inducing similar changes in neurite length exhibit underappreciated phenotypic nuance at the scale of the growth cone.


Subject(s)
Growth Cones/ultrastructure , Image Processing, Computer-Assisted/statistics & numerical data , Molecular Imaging/standards , Neurons/ultrastructure , Software , Time-Lapse Imaging/standards , rho GTP-Binding Proteins/genetics , Animals , Cell Line, Tumor , Cell Movement , Cell Shape/genetics , Gene Expression Regulation , Genetic Heterogeneity , Growth Cones/metabolism , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Mice , Molecular Imaging/methods , Neurons/metabolism , Neuropeptides/deficiency , Neuropeptides/genetics , Phosphoproteins/deficiency , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , Time-Lapse Imaging/methods , cdc42 GTP-Binding Protein/deficiency , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/deficiency , rac1 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/deficiency , rhoA GTP-Binding Protein
10.
Exp Cell Res ; 371(1): 72-82, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30056063

ABSTRACT

Animal cells divide by a process called cytokinesis which relies on the constriction of a contractile actomyosin ring leading to the production of two daughter cells. Cytokinesis is an intrinsic property of cells which occurs even for artificially isolated cells. During division, isolated cells undergo dramatic changes in shape such as rounding and membrane deformation as the division furrow ingresses. However, cells are often embedded in tissues and thus are surrounded by neighbouring cells. How these neighbours might influence, or might themselves be influenced by, the shape changes of cytokinesis is poorly understood in vertebrates. Here, we show that during cytokinesis of epithelial cells in the Xenopus embryo, lateral cell-cell contacts remain almost perpendicular to the epithelial plane. Depletion of the tight junction-associated protein GEF-H1 leads to a transient and stereotyped deformation of cell-cell contacts. Although, this deformation occurs only during cytokinesis, we show that it originates from immediate neighbours of the dividing cell. Moreover, we show that exocyst and recycling endosome regulation by GEF-H1 are involved in adaptation of cell-cell contacts to deformation. Our results highlight the crucial role of tight junctions and GEF-H1 in cell-cell contact adaptation when cells are exposed to a mechanical stress such as cytokinesis.


Subject(s)
Cytokinesis/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Mechanotransduction, Cellular , Rho Guanine Nucleotide Exchange Factors/genetics , Tight Junctions/metabolism , Xenopus Proteins/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Adherens Junctions/metabolism , Adherens Junctions/ultrastructure , Amides/pharmacology , Animals , Cell Communication , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Shape , Embryo, Nonmammalian , Epithelial Cells/ultrastructure , Morpholinos/genetics , Morpholinos/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/deficiency , Tight Junctions/ultrastructure , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/deficiency , Xenopus laevis , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
11.
Sci Rep ; 8(1): 8811, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891904

ABSTRACT

The characteristic six layers of the mammalian neocortex develop sequentially as neurons are generated by neural progenitors and subsequently migrate past older neurons to their final position in the cortical plate. One of the earliest steps of neuronal differentiation is the formation of an axon. Small GTPases play essential roles during this process by regulating cytoskeletal dynamics and intracellular trafficking. While the function of GTPases has been studied extensively in cultured neurons and in vivo much less is known about their upstream regulators. Here we show that Arhgef7 (also called ßPix or Cool1) is essential for axon formation during cortical development. The loss of Arhgef7 results in an extensive loss of axons in cultured neurons and in the developing cortex. Arhgef7 is a guanine-nucleotide exchange factor (GEF) for Cdc42, a GTPase that has a central role in directing the formation of axons during brain development. However, active Cdc42 was not able to rescue the knockdown of Arhgef7. We show that Arhgef7 interacts with the GTPase TC10 that is closely related to Cdc42. Expression of active TC10 can restore the ability to extend axons in Arhgef7-deficient neurons. Our results identify an essential role of Arhgef7 during neuronal development that promotes axon formation upstream of TC10.


Subject(s)
Axons/physiology , Cell Differentiation , Cerebral Cortex/embryology , Rho Guanine Nucleotide Exchange Factors/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Mice, Inbred C57BL , Mice, Knockout , Rats , Rho Guanine Nucleotide Exchange Factors/deficiency , cdc42 GTP-Binding Protein/metabolism
12.
Neural Plast ; 2018: 6015753, 2018.
Article in English | MEDLINE | ID: mdl-29670649

ABSTRACT

The hippocampal dentate gyrus plays a role in spatial learning and memory and is thought to encode differences between similar environments. The integrity of excitatory and inhibitory transmission and a fine balance between them is essential for efficient processing of information. Therefore, identification and functional characterization of crucial molecular players at excitatory and inhibitory inputs is critical for understanding the dentate gyrus function. In this minireview, we discuss recent studies unraveling molecular mechanisms of excitatory/inhibitory synaptic transmission, long-term synaptic plasticity, and dentate granule cell excitability in the hippocampus of live animals. We focus on the role of three major postsynaptic proteins localized at excitatory (neuroligin-1) and inhibitory synapses (neuroligin-2 and collybistin). In vivo recordings of field potentials have the advantage of characterizing the effects of the loss of these proteins on the input-output function of granule cells embedded in a network with intact connectivity. The lack of neuroligin-1 leads to deficient synaptic plasticity and reduced excitation but normal granule cell output, suggesting unaltered excitation-inhibition ratio. In contrast, the lack of neuroligin-2 and collybistin reduces inhibition resulting in a shift towards excitation of the dentate circuitry.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Dentate Gyrus/physiology , Excitatory Postsynaptic Potentials/physiology , Nerve Tissue Proteins/deficiency , Neuronal Plasticity/physiology , Rho Guanine Nucleotide Exchange Factors/deficiency , Animals , Cell Adhesion Molecules, Neuronal/genetics , Gene Knockout Techniques , Humans , Nerve Net/physiology , Nerve Tissue Proteins/genetics , Neural Inhibition/physiology , Rho Guanine Nucleotide Exchange Factors/genetics , Synapses/genetics , Synapses/metabolism
13.
Mol Autism ; 9: 11, 2018.
Article in English | MEDLINE | ID: mdl-29456827

ABSTRACT

Background: Impaired social interaction is one of the essential features of autism spectrum disorder (ASD). Our previous copy number variation (CNV) study discovered a novel deleted region associated with ASD. One of the genes included in the deleted region is ARHGEF10. A missense mutation of ARHGEF10 has been reported to be one of the contributing factors in several diseases of the central nervous system. However, the relationship between the loss of ARHGEF10 and the clinical symptoms of ASD is unclear. Methods: We generated Arhgef10 knockout mice as a model of ASD and characterized the social behavior and the biochemical changes in the brains of the knockout mice. Results: Compared with their wild-type littermates, the Arhgef10-depleted mice showed social interaction impairment, hyperactivity, and decreased depression-like and anxiety-like behavior. Behavioral measures of learning in the Morris water maze were not affected by Arhgef10 deficiency. Moreover, neurotransmitters including serotonin, norepinephrine, and dopamine were significantly increased in different brain regions of the Arhgef10 knockout mice. In addition, monoamine oxidase A (MAO-A) decreased in several brain regions. Conclusions: These results suggest that ARHGEF10 is a candidate risk gene for ASD and that the Arhgef10 knockout model could be a tool for studying the mechanisms of neurotransmission in ASD. Trial registration: Animal studies were approved by the Institutional Animal Care and Use Committee of National Taiwan University (IACUC 20150023). Registered 1 August 2015.


Subject(s)
Autism Spectrum Disorder/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Social Behavior , Animals , Brain/metabolism , Brain/physiology , Learning , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Norepinephrine/metabolism , Rho Guanine Nucleotide Exchange Factors/deficiency , Serotonin/metabolism
14.
J Thromb Haemost ; 15(10): 2053-2064, 2017 10.
Article in English | MEDLINE | ID: mdl-28799234

ABSTRACT

Essentials ARHGEF10 single-nucleotide polymorphism provides risk of ischemic and atherothrombotic stroke. The role of ARHGEF10 in platelet function was examined using ARHGEF10 knockout mice. ARHGEF10 deficiency inhibits platelet function and arterial thrombus formation. ARHGEF10 knockout protects mice from stroke-induced infarction. SUMMARY: Background ARHGEF10, a member of the Rho guanine nucleotide exchange factor (GEF) family, stimulates Rho GTPases. Rho GTPases have been reported to regulate a variety of cellular behaviors, such as cell polarity, cytoskeletal organization, and gene transcription. ARHGEF10 single-nucleotide polymorphisms are linked to the risk of ischemic stroke. However, the role of ARHGEF10 in platelet function remains unknown. Objective To examine the role of ARHGEF10 in platelet function. Methods ARHGEF10-/- were generated. We examined the in vitro and in vivo effects of ARHGEF10 knockout on platelet function and arterial thrombosis formation. Results ARHGEF10-/- mice had normal platelet counts, but showed altered aggregation in response to thrombin, collagen, ADP, protease-activated receptor-4 peptide, and U46619 stimulation. ARHGEF10 knockout influenced platelet spreading on fibrinogen-coated surfaces, and caused the platelets to show less lamellipodia-like extension than wild-type platelets. ARHGEF10 knockout also inhibited platelet clot retraction induced by thrombin stimulation. ARHGEF10 knockout resulted in prolonged tail bleeding time and inhibited the stable thrombus formation induced by FeCl3 in the carotid artery. Conclusions ARHGEF10 serves as an important regulator in platelet shape change, spreading, and aggregation. Moreover, ARHGEF10 also plays an important role in arterial thrombosis formation.


Subject(s)
Arterial Occlusive Diseases/prevention & control , Blood Platelets/metabolism , Carotid Artery Diseases/prevention & control , Hemostasis , Platelet Aggregation , Rho Guanine Nucleotide Exchange Factors/deficiency , Thrombosis/prevention & control , Animals , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/genetics , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Cell Shape , Chlorides , Disease Models, Animal , Ferric Compounds , Gene Knockout Techniques , Genotype , Male , Mice, 129 Strain , Mice, Knockout , Myosin Light Chains/metabolism , Phenotype , Phosphorylation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Rho Guanine Nucleotide Exchange Factors/blood , Rho Guanine Nucleotide Exchange Factors/genetics , Selenoprotein P/blood , Thrombosis/blood , Thrombosis/genetics , Time Factors
15.
J Cell Sci ; 130(14): 2329-2343, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28596238

ABSTRACT

During healing of the skin, the cytoskeleton of keratinocytes and their matrix adhesions, including focal adhesions (FAs), undergo reorganization. These changes are coordinated by small GTPases and their regulators, including the guanine nucleotide exchange factor ß-PIX (also known as ARHGEF7). In fibroblasts, ß-PIX activates small GTPases, thereby enhancing migration. In keratinocytes in vitro, ß-PIX localizes to FAs. To study ß-PIX functions, we generated ß-PIX knockdown keratinocytes. During wound closure of ß-PIX knockdown cell monolayers, disassembly of FAs is impaired, and their number and size are increased. In addition, in the ß-PIX knockdown cells, phosphorylated myosin light chain (MLC; also known as MYL2) is present not only in the leading edge of cells at the wound front, but also in the cells following the front, while p21-activated kinase 2 (PAK2), a regulator of MLC kinase (MYLK), is mislocalized. Inhibition or depletion of MYLK restores FA distribution in ß-PIX knockdown cells. Traction forces generated by ß-PIX knockdown cells are increased relative to those in control cells, a result consistent with an unexpected enhancement in the migration of single ß-PIX knockdown cells and monolayers of such cells. We propose that targeting ß-PIX might be a means of promoting epithelialization of wounds in vivo.


Subject(s)
Focal Adhesions/metabolism , Keratinocytes/metabolism , Myosin Light Chains/metabolism , Rho Guanine Nucleotide Exchange Factors/deficiency , Calcium-Binding Proteins/metabolism , Cell Line , Cell Movement/physiology , Gene Knockdown Techniques , Humans , Keratinocytes/cytology , Myosin-Light-Chain Kinase/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , p21-Activated Kinases/metabolism
16.
J Cell Biol ; 215(1): 107-119, 2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27738004

ABSTRACT

Leukocyte crawling and transendothelial migration (TEM) are potentiated by shear stress caused by blood flow. The mechanism that couples shear stress to migration has not been fully elucidated. We found that mice lacking GEF-H1 (GEF-H1-/-), a RhoA-specific guanine nucleotide exchange factor (GEF), displayed limited migration and recruitment of neutrophils into inflamed tissues. GEF-H1-/- leukocytes were deficient in in vivo crawling and TEM in the postcapillary venules. We demonstrated that although GEF-H1 deficiency had little impact on the migratory properties of neutrophils under static conditions, shear stress triggered GEF-H1-dependent spreading and crawling of neutrophils and relocalization of GEF-H1 to flotillin-2-rich uropods. Our results identify GEF-H1 as a component of the shear stress response machinery in neutrophils required for a fully competent immune response to bacterial infection.


Subject(s)
Cell Movement , Inflammation/pathology , Neutrophils/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Stress, Mechanical , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , HL-60 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Microtubules/drug effects , Microtubules/metabolism , Models, Biological , Muscles/drug effects , Myosin Light Chains/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Phosphorylation/drug effects , Polymerization/drug effects , Rho Guanine Nucleotide Exchange Factors/deficiency , Sepsis/pathology
17.
Thromb Haemost ; 116(3): 506-16, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27345948

ABSTRACT

Leukemia-Associated RhoGEF (LARG) is highly expressed in platelets, which are essential for maintaining normal haemostasis. We studied the function of LARG in murine and human megakaryocytes and platelets with Larg knockout (KO), shRNA-mediated knockdown and small molecule-mediated inhibition. We found that LARG is important for human, but not murine, megakaryocyte maturation. Larg KO mice exhibit macrothrombocytopenia, internal bleeding in the ovaries and prolonged bleeding times. KO platelets have impaired aggregation, α-granule release and integrin α2bß3 activation in response to thrombin and thromboxane, but not to ADP. The same agonist-specific reductions in platelet aggregation occur in human platelets treated with a LARG inhibitor. Larg KO platelets have reduced RhoA activation and myosin light chain phosphorylation, suggesting that Larg plays an agonist-specific role in platelet signal transduction. Using two different in vivo assays, Larg KO mice are protected from in vivo thrombus formation. Together, these results establish that LARG regulates human megakaryocyte maturation, and is critical for platelet function in both humans and mice.


Subject(s)
Blood Platelets/metabolism , Rho Guanine Nucleotide Exchange Factors/blood , rho GTP-Binding Proteins/blood , rhoA GTP-Binding Protein/blood , Animals , Bleeding Time , Blood Platelets/drug effects , Gene Knockdown Techniques , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Light Chains/blood , Platelet Function Tests , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Thrombin/metabolism , Thrombin/pharmacology , Thrombopoiesis/genetics , Thrombopoiesis/physiology , Thromboxanes/blood , Thromboxanes/pharmacology , rho GTP-Binding Proteins/agonists , rhoA GTP-Binding Protein/agonists
18.
Eur J Hum Genet ; 24(1): 59-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25898924

ABSTRACT

Protein synthesis regulation via mammalian target of rapamycin complex 1 (mTORC1) signaling pathway has key roles in neural development and function, and its dysregulation is involved in neurodevelopmental disorders associated with autism and intellectual disability. mTOR regulates assembly of the translation initiation machinery by interacting with the eukaryotic initiation factor eIF3 complex and by controlling phosphorylation of key translational regulators. Collybistin (CB), a neuron-specific Rho-GEF responsible for X-linked intellectual disability with epilepsy, also interacts with eIF3, and its binding partner gephyrin associates with mTOR. Therefore, we hypothesized that CB also binds mTOR and affects mTORC1 signaling activity in neuronal cells. Here, by using induced pluripotent stem cell-derived neural progenitor cells from a male patient with a deletion of entire CB gene and from control individuals, as well as a heterologous expression system, we describe that CB physically interacts with mTOR and inhibits mTORC1 signaling pathway and protein synthesis. These findings suggest that disinhibited mTORC1 signaling may also contribute to the pathological process in patients with loss-of-function variants in CB.


Subject(s)
Autistic Disorder/genetics , Eukaryotic Initiation Factor-3/genetics , Gene Deletion , Intellectual Disability/genetics , Multiprotein Complexes/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , TOR Serine-Threonine Kinases/genetics , Adolescent , Autistic Disorder/metabolism , Autistic Disorder/physiopathology , Case-Control Studies , Eukaryotic Initiation Factor-3/metabolism , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Intellectual Disability/metabolism , Intellectual Disability/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Peptide Chain Initiation, Translational , Primary Cell Culture , Protein Binding , Rho Guanine Nucleotide Exchange Factors/deficiency , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transfection
19.
J Thromb Haemost ; 13(11): 2102-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26334261

ABSTRACT

BACKGROUND: RhoA is an important regulator of platelet responses downstream of Gα13 , yet we still know little about its regulation in platelets. Leukemia-associated Rho guanine-nucleotide exchange factor (GEF [LARG]), a RhoA GEF, is highly expressed in platelets and may constitute a major upstream activator of RhoA. To this end, it is important to determine the role of LARG in platelet function and thrombosis. METHODS AND RESULTS: Using a platelet-specific gene knockout, we show that the absence of LARG results in a marked reduction in aggregation and dense-granule secretion in response to the thromboxane mimetic U46619 and proteinase-activated receptor 4-activating peptide, AYPGKF, but not to adenosine diphosphate. In a ferric chloride thrombosis model in vivo, this translated into a defect, under mild injury conditions. Importantly, agonist-induced RhoA activation was not affected by the absence of LARG, although basal activity was reduced, suggesting that LARG may play a housekeeper role in regulating constitutive RhoA activity. CONCLUSIONS: LARG plays an important role in platelet function and thrombosis in vivo. However, although LARG may have a role in regulating the resting activation state of RhoA, its role in regulating platelet function may principally be through RhoA-independent pathways, possibly through other Rho family members.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , Thrombosis/blood , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Cell Degranulation/drug effects , Chlorides/toxicity , Crosses, Genetic , Ferric Compounds/toxicity , Gene Knockout Techniques , Mice , Mice, Knockout , Oligopeptides/pharmacology , Organ Specificity , Platelet Aggregation , Rho Guanine Nucleotide Exchange Factors/blood , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Thrombosis/chemically induced
20.
Nature ; 516(7530): 254-8, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25274307

ABSTRACT

Germinal centre B-cell-like diffuse large B-cell lymphoma (GCB-DLBCL) is a common malignancy, yet the signalling pathways that are deregulated and the factors leading to its systemic dissemination are poorly defined. Work in mice showed that sphingosine-1-phosphate receptor-2 (S1PR2), a Gα12 and Gα13 coupled receptor, promotes growth regulation and local confinement of germinal centre B cells. Recent deep sequencing studies of GCB-DLBCL have revealed mutations in many genes in this cancer, including in GNA13 (encoding Gα13) and S1PR2 (refs 5,6, 7). Here we show, using in vitro and in vivo assays, that GCB-DLBCL-associated mutations occurring in S1PR2 frequently disrupt the receptor's Akt and migration inhibitory functions. Gα13-deficient mouse germinal centre B cells and human GCB-DLBCL cells were unable to suppress pAkt and migration in response to S1P, and Gα13-deficient mice developed germinal centre B-cell-derived lymphoma. Germinal centre B cells, unlike most lymphocytes, are tightly confined in lymphoid organs and do not recirculate. Remarkably, deficiency in Gα13, but not S1PR2, led to germinal centre B-cell dissemination into lymph and blood. GCB-DLBCL cell lines frequently carried mutations in the Gα13 effector ARHGEF1, and Arhgef1 deficiency also led to germinal centre B-cell dissemination. The incomplete phenocopy of Gα13- and S1PR2 deficiency led us to discover that P2RY8, an orphan receptor that is mutated in GCB-DLBCL and another germinal centre B-cell-derived malignancy, Burkitt's lymphoma, also represses germinal centre B-cell growth and promotes confinement via Gα13. These findings identify a Gα13-dependent pathway that exerts dual actions in suppressing growth and blocking dissemination of germinal centre B cells that is frequently disrupted in germinal centre B-cell-derived lymphoma.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/pathology , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Germinal Center/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Signal Transduction , Animals , Blood/immunology , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Line, Tumor , Cell Movement/genetics , Humans , Lymph/cytology , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Mice, Inbred C57BL , Mutation/genetics , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Receptors, Purinergic P2Y/genetics , Receptors, Purinergic P2Y/metabolism , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Sphingosine-1-Phosphate Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...