scholarly journals SRGAP1 Controls Small Rho GTPases To Regulate Podocyte Foot Process Maintenance

2021 ◽  
Vol 32 (3) ◽  
pp. 563-579
Author(s):  
Manuel Rogg ◽  
Jasmin I. Maier ◽  
Robert Dotzauer ◽  
Nadine Artelt ◽  
Oliver Kretz ◽  
...  

BackgroundPrevious research demonstrated that small Rho GTPases, modulators of the actin cytoskeleton, are drivers of podocyte foot-process effacement in glomerular diseases, such as FSGS. However, a comprehensive understanding of the regulatory networks of small Rho GTPases in podocytes is lacking.MethodsWe conducted an analysis of podocyte transcriptome and proteome datasets for Rho GTPases; mapped in vivo, podocyte-specific Rho GTPase affinity networks; and examined conditional knockout mice and murine disease models targeting Srgap1. To evaluate podocyte foot-process morphology, we used super-resolution microscopy and electron microscopy; in situ proximity ligation assays were used to determine the subcellular localization of the small GTPase-activating protein SRGAP1. We performed functional analysis of CRISPR/Cas9-generated SRGAP1 knockout podocytes in two-dimensional and three-dimensional cultures and quantitative interaction proteomics.ResultsWe demonstrated SRGAP1 localization to podocyte foot processes in vivo and to cellular protrusions in vitro. Srgap1fl/fl*Six2Cre but not Srgap1fl/fl*hNPHS2Cre knockout mice developed an FSGS-like phenotype at adulthood. Podocyte-specific deletion of Srgap1 by hNPHS2Cre resulted in increased susceptibility to doxorubicin-induced nephropathy. Detailed analysis demonstrated significant effacement of podocyte foot processes. Furthermore, SRGAP1-knockout podocytes showed excessive protrusion formation and disinhibition of the small Rho GTPase machinery in vitro. Evaluation of a SRGAP1-dependent interactome revealed the involvement of SRGAP1 with protrusive and contractile actin networks. Analysis of glomerular biopsy specimens translated these findings toward human disease by displaying a pronounced redistribution of SRGAP1 in FSGS.ConclusionsSRGAP1, a podocyte-specific RhoGAP, controls podocyte foot-process architecture by limiting the activity of protrusive, branched actin networks. Therefore, elucidating the complex regulatory small Rho GTPase affinity network points to novel targets for potentially precise intervention in glomerular diseases.

2011 ◽  
Vol 39 (6) ◽  
pp. 1606-1611 ◽  
Author(s):  
Katarzyna Leszczynska ◽  
Sukhbir Kaur ◽  
Eleanor Wilson ◽  
Roy Bicknell ◽  
Victoria L. Heath

RhoJ is an endothelially expressed member of the Cdc42 (cell division cycle 42) subfamily of small Rho GTPases. It is expressed in both the developing mammalian vasculature and the vascular beds of a number of adult tissues, with its expression regulated by the endothelial transcription factor ERG (ETS-related gene). RhoJ has been shown to regulate endothelial motility, tubulogenesis and lumen formation in vitro, and modulates the vascularization of Matrigel plugs in vivo. Both vascular endothelial growth factor and semaphorin 3E have been found to affect its activation. RhoJ has been shown to be a focal-adhesion-localized Rho GTPase which can modulate focal adhesion number, actomyosin contractility and activity of Cdc42 and Rac1. The present review discusses the biology of RhoJ with a focus on recent reports of its role in endothelial cells and angiogenesis.


2021 ◽  
Vol 12 (4) ◽  
Author(s):  
Lifeng Feng ◽  
Miaoqin Chen ◽  
Yiling Li ◽  
Muchun Li ◽  
Shiman Hu ◽  
...  

Abstractp62/SQSTM1 is frequently up-regulated in many cancers including hepatocellular carcinoma. Highly expressed p62 promotes hepato-carcinogenesis by activating many signaling pathways including Nrf2, mTORC1, and NFκB signaling. However, the underlying mechanism for p62 up-regulation in hepatocellular carcinoma remains largely unclear. Herein, we confirmed that p62 was up-regulated in hepatocellular carcinoma and its higher expression was associated with shorter overall survival in patients. The knockdown of p62 in hepatocellular carcinoma cells decreased cell growth in vitro and in vivo. Intriguingly, p62 protein stability could be reduced by its acetylation at lysine 295, which was regulated by deacetylase Sirt1 and acetyltransferase GCN5. Acetylated p62 increased its association with the E3 ligase Keap1, which facilitated its poly-ubiquitination-dependent proteasomal degradation. Moreover, Sirt1 was up-regulated to deacetylate and stabilize p62 in hepatocellular carcinoma. Additionally, Hepatocyte Sirt1 conditional knockout mice developed much fewer liver tumors after Diethynitrosamine treatment, which could be reversed by the re-introduction of exogenous p62. Taken together, Sirt1 deacetylates p62 at lysine 295 to disturb Keap1-mediated p62 poly-ubiquitination, thus up-regulating p62 expression to promote hepato-carcinogenesis. Therefore, targeting Sirt1 or p62 is a reasonable strategy for the treatment of hepatocellular carcinoma.


2019 ◽  
Vol 119 (05) ◽  
pp. 744-757 ◽  
Author(s):  
Vanessa Scanlon ◽  
Alexandra Teixeira ◽  
Tarun Tyagi ◽  
Siying Zou ◽  
Ping-Xia Zhang ◽  
...  

AbstractCadherins play a major role in mediating cell–cell adhesion, which shares many parallels with platelet–platelet interactions during aggregate formation and clot stabilization. Platelets express epithelial (E)-cadherin, but its contribution to platelet function and/or platelet production is currently unknown. To assess the role of E-cadherin in platelet production and function in vitro and in vivo, we utilized a megakaryocyte-specific E-cadherin knockout mouse model. Loss of E-cadherin in megakaryocytes does not affect megakaryocyte maturation, platelet number or size. However, platelet dysfunction in the absence of E-cadherin is revealed when conditional knockout mice are challenged with acute antibody-mediated platelet depletion. Unlike wild-type mice that recover fully, knockout mice die within 72 hours post-antibody administration, likely from haemorrhage. Furthermore, conditional knockout mice have prolonged tail bleeding times, unstable clot formation, reduced clot retraction and reduced fibrin deposition in in vivo injury models. Murine platelet aggregation in vitro in response to thrombin and thrombin receptor activating peptide is compromised in E-cadherin null platelets, while aggregation in response to adenosine diphosphate (ADP) is not significantly different. Consistent with this, in vitro aggregation of primary human platelets in response to thrombin is decreased by an inhibitory E-cadherin antibody. Integrin activation and granule secretion in response to ADP and thrombin are not affected in E-cadherin null platelets, but Akt and glycogen synthase kinase 3β (GSK3β) activation are attenuated, suggesting a that E-cadherin contributes to aggregation, clot stabilization and retraction that is mediated by phosphoinositide 3-kinase/Akt/GSK3β signalling. In summary, E-cadherin plays a salient role in platelet aggregation and clot stability.


2001 ◽  
Vol 194 (1) ◽  
pp. 57-70 ◽  
Author(s):  
David A. Ingram ◽  
Kelly Hiatt ◽  
Alastair J. King ◽  
Lucy Fisher ◽  
Rama Shivakumar ◽  
...  

Mutations in the NF1 tumor suppressor gene cause neurofibromatosis type I (NF1), a disease characterized by the formation of cutaneous neurofibromas infiltrated with a high density of degranulating mast cells. A hallmark of cell lines generated from NF1 patients or Nf1-deficient mice is their propensity to hyperproliferate. Neurofibromin, the protein encoded by NF1, negatively regulates p21ras activity by accelerating the conversion of Ras-GTP to Ras-GDP. However, identification of alterations in specific p21ras effector pathways that control proliferation in NF1-deficient cells is incomplete and critical for understanding disease pathogenesis. Recent studies have suggested that the proliferative effects of p21ras may depend on signaling outputs from the small Rho GTPases, Rac and Rho, but the physiologic importance of these interactions in an animal disease model has not been established. Using a genetic intercross between Nf1+/− and Rac2−/− mice, we now provide genetic evidence to support a biochemical model where hyperactivation of the extracellular signal–regulated kinase (ERK) via the hematopoietic-specific Rho GTPase, Rac2, directly contributes to the hyperproliferation of Nf1-deficient mast cells in vitro and in vivo. Further, we demonstrate that Rac2 functions as mediator of cross-talk between phosphoinositide 3-kinase (PI-3K) and the classical p21ras-Raf-Mek-ERK pathway to confer a distinct proliferative advantage to Nf1+/− mast cells. Thus, these studies identify Rac2 as a novel mediator of cross-talk between PI-3K and the p21ras-ERK pathway which functions to alter the cellular phenotype of a cell lineage involved in the pathologic complications of a common genetic disease.


2020 ◽  
Author(s):  
Robert Beal ◽  
Ana Alonso-Carriazo Fernandez ◽  
Dimitris K. Grammatopoulos ◽  
Karl Matter ◽  
Maria S. Balda

SUMMARYCoordination of cell-cell adhesion, actomyosin dynamics and gene expression is crucial for morphogenetic processes underlying tissue and organ development. Rho GTPases are main regulators of the cytoskeleton and adhesion. They are activated by guanine nucleotide exchange factors in a spatially and temporally controlled manner. However, the roles of these Rho GTPase activators during complex developmental processes are still poorly understood. ARHGEF18/p114RhoGEF is a tight junction-associated RhoA activator that forms complexes with myosin II, and regulates actomyosin contractility. Here we show that p114RhoGEF/ ARHGEF18 is required for mouse syncytiotrophoblast differentiation and placenta development. In vitro and in vivo experiments identify that p114RhoGEF controls expression of AKAP12, a protein regulating PKA signalling, and is required for PKA-induced actomyosin remodelling, CREB-driven gene expression of proteins required for trophoblast differentiation, and, hence, trophoblast cell-cell fusion. Our data thus indicate that p114RhoGEF links actomyosin dynamics and cell-cell junctions to PKA/CREB signalling, gene expression and cell-cell fusion.


2021 ◽  
Author(s):  
Kosei Nagata ◽  
Hironori Hojo ◽  
Song Ho Chang ◽  
Hiroyuki Okada ◽  
Fumiko Yano ◽  
...  

Abstract The Runt-related transcription factor (Runx) family plays various roles in the homeostasis of cartilage. Here, we examined the role of Runx2 and Runx3 for osteoarthritis (OA) development in vivo and in vitro. Runx3 knockout mice accelerated OA by surgical induction, accompanied with decreased expression of lubricin and aggrecan. Meanwhile, Runx2 conditional knockout mice showed biphasic phenotypes; OA was inhibited by hetero-knockout accompanied with decreased matrix metallopeptidase 13 (Mmp13) expression, but accelerated in homo-knockout of Runx2 accompanied with reduction of type II collagen (Col2a1) expression. Comprehensive transcriptional analyses revealed lubricin and aggrecan as transcriptional target genes of Runx3, and indicated that Runx2 sustained Col2a1 expression through an intron 6 enhancer when Sox9 was decreased. Intra-articular administration of Runx3 adenovirus ameliorated development of surgically induced OA. Runx3 protects adult articular cartilage through extracellular matrix protein production under the normal condition, while Runx2 exerts both catabolic and anabolic effects under the inflammatory condition.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 80-80
Author(s):  
Abel Sanchez-Aguilera ◽  
Yun-Jung Lee ◽  
Cristina Lo Celso ◽  
Kristina Brumme ◽  
Charles P Lin ◽  
...  

Abstract Abstract 80 Background: Rho GTPases are molecular switches that regulate actin cytoskeleton dynamics, cell proliferation and survival. In hematopoietic stem cells and progenitors (HSC/P), several Rho GTPases (including Rac1, Rac2 and Cdc42) function as critical regulators of engraftment through the integration of diverse extracellular signals, such as those transmitted by growth factor, chemokine and adhesion receptors. In addition, Rac-deficient mice show significantly increased numbers of mobilized HSC/P. GTPase activation downstream of these and other receptors is mediated by a large family of guanine nucleotide exchange factors (GEF). Functional interactions between receptors, GEF and Rho GTPases are potentially complex and the crucial biochemical pathways regulating HSC activity have not been defined. Among the Rho/Rac GEFs, Vav1 shows hematopoietic-specific expression and has been previously implicated in immune cell processes, such as immunoreceptor signaling in lymphocytes and neutrophil migration. To further explore the mechanism of Rho GTPase regulation of HSC engraftment, we investigated the role of Vav1 GEF in Rho GTPase activation after ligation of multiple HSC receptors and the effect of genetic deletion of Vav1 on HSC homing, retention and engraftment in the hematopoietic microenvironment. Methods: GTPase activation (Rac, Cdc42, RhoA) was analyzed by in vitro pulldown assays. The HSC/P compartment of Vav1−/− mice was studied by flow cytometry, colony forming cell (CFC) assays, progenitor (CFC) homing, competitive and non-competitive repopulation assays. HSC localization in the endosteal niche was determined by intravital microscopy 1 h and 48 h after transplant. Results: At the biochemical level, Vav1−/− hematopoietic progenitors showed a dysfunctional Rho GTPase activation pattern, with increased baseline levels of GTP-bound Rac, Cdc42 and RhoA; however, in the absence of Vav1, these GTPases were unresponsive to stimulation by stem cell factor and SDF1α, critical proteins in HSC engraftment. In spite of this biochemical abnormality, Vav1−/− mice at baseline had nearly normal numbers of immunophenotypically defined HSC, myeloid and lymphoid progenitors in the bone marrow (BM), and normal hematopoietic progenitor content as defined by CFC, although reduced rather than increased circulating HSC/P. Vav1−/− HSC/P transplanted into irradiated recipients exhibited normal BM CFC homing efficiency (∼5%) and normal early endosteal localization of HSC in vivo (1 h after injection) as determined by intravital microscopy. Surprisingly-but in concordance with the normal BM homing of HSC/P in vivo- the loss of Vav1 did not affect hematopoietic progenitor chemotaxis or short-term adhesion to fibronectin in vitro. However, there was a significant decrease in the retention of HSC in the endosteal space at 48 h after transplant (Vav1−/− HSC numbers were reduced to 46%, relative to WT HSC) and this defect was associated with a profound loss of short- and long-term engraftment. In competitive repopulation assays, Vav1−/− cells virtually did not contribute to the graft (Table 1), whereas in a non-competitive setting, they either failed to rescue the recipient (60% survival vs 100% at 1 month, Vav1−/− vs WT) or showed significantly delayed hematopoietic reconstitution (Table 2). Conclusions: The hematopoietic-specific GEF Vav1 is essential for the appropriate microenvironment-induced Rho GTPase activation in HSC/P after transplant and is required for the retention of HSC/P in the BM endosteal niche and subsequent engraftment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 631-631
Author(s):  
Marie-Dominique Filippi ◽  
Pierre-Yves Berclaz ◽  
Kathleen Szczur ◽  
Chad Harris ◽  
David A. Williams

Abstract Neutrophils are a critical cell in inflammatory processes by moving rapidly to tissue sites of inflammation to perform phagocytosis, cytokine and reactive oxygen species release. Members of the small Rho GTPase family, Rac1, Rac2, CDC42 and RhoA, are central regulators of cell movement via cytoskeleton rearrangement. We have previously demonstrated that the Rho family GTPase Rac2 is a critical regulator of neutrophil functions in vitro and in vivo (Roberts et al, Immunity 1999). We have also demonstrated that in response to formyl-methionyl-leucyl-phenylalanine (fMLP), the related GTPase Rac1 plays a distinct, but as yet ill-defined role in tail retraction during cell movement and cell spreading in vitro (Gu and Filippi et al, Science 2003). Here, we further demonstrate that Rac1 appears to be critical for β2-integrin mediated adhesion and migration likely via cross talk with another Rho GTPase, RhoA. Although, Rac1−/− PMNs show normal in vitro migration in response to fMLP using the Boyden chamber assay, Rac1−/− PMNs demonstrate a dramatic defect compared with WT cells in haptotaxis using transwell precoated with fibrinogen (1.3±0.3x103 vs 9.8±0.5x103). In addition, Rac1−/− PMNs displayed increased frequency in pseudopodia formation associated with lack of cell body contraction upon integrin ligation compared with WT (80% vs 40%). We noted that this phenotype closely mimics deregulation of the related Rho GTPase, RhoA. Remarkably, Rac1-deficiency leads to mislocalization of RhoA in neutrophils after integrin ligation and reintroduction of Rac1 into Rac1−/− cells completely restores the correct localization of RhoA. These data are consistent with the hypothesis that Rho GTPases interact in a time- and space-dependent manner. Because fMLP-induced PMN migration into the lung has previously been shown to be beta2-integrin dependent (Mackarel, Am. J. Respir. Cell. Mol. Biol 2000), we used a model of neutrophil associated lung inflammation induced by intratracheal (IT) injection of fMLP to address the physiological role of Rac1 in neutrophil-derived inflammatory processes in vivo,. To study the role of Rac1 specifically in bone marrow-derived cells, we reconstituted C57BL/6 mice with either wild type or Rac1Flox/Flox bone marrow cells. After Cre-mediated deletion of Rac1, reconstituted mice were treated with one dose of fMLP (20mg) IT. One day after fMLP exposure, bronchoalveolar lavage (BAL) from reconstituted animals showed complete loss of Rac1 expression and demonstrated significantly reduced numbers of migrated neutrophils in BAL compared with mice reconstituted with WT cells (3.1±0.65 vs 9.56±2, p<0.05). Importantly, 5 weeks after fMLP exposure IT, Rac1−/− recipients displayed a significant reduction in emphysematous lesions as compared with WT as assessed by morphometric measurement of alveolar spaces (57.6±7.8 vs 73.3±3.04, p<0.05), demonstrating the physiological relevance of Rac1 in neutrophil-related inflammatory responses in vivo. Taken together, these results suggest that Rac1 activity regulates b2 integrin-induced cell shape change and RhoA subcellular localization in PMNs and demonstrate the existence of physiological cross talk between Rac1 and RhoA where RhoA activity depends at least in part on Rac1. Thus, Rac1 and RhoA appear to coordinately regulate PMN migration into the lung during inflammation.


2006 ◽  
Vol 17 (11) ◽  
pp. 4760-4768 ◽  
Author(s):  
Céline DerMardirossian ◽  
Gabriel Rocklin ◽  
Ji-Yeon Seo ◽  
Gary M. Bokoch

Rho GTPases (Rac, Rho, and Cdc42) play important roles in regulating cell function through their ability to coordinate the actin cytoskeleton, modulate the formation of signaling reactive oxidant species, and control gene transcription. Activation of Rho GTPase signaling pathways requires the regulated release of Rho GTPases from RhoGDI complexes, followed by their reuptake after membrane cycling. We show here that Src kinase binds and phosphorylates RhoGDI both in vitro and in vivo at Tyr156. Analysis of Rho GTPase–RhoGDI complexes using in vitro assays of complexation and in vivo by coimmunoprecipitation analysis indicates that Src-mediated phosphorylation of Tyr156 causes a dramatic decrease in the ability of RhoGDI to form a complex with RhoA, Rac1, or Cdc42. Phosphomimetic mutation of Tyr156→Glu results in the constitutive association of RhoGDIY156E with the plasma membrane and/or associated cortical actin. Substantial cortical localization of tyrosine-phosphorylated RhoGDI is also observed in fibroblasts expressing active Src, where it is most evident in podosomes and regions of membrane ruffling. Expression of membrane-localized RhoGDIY156E mutant is associated with enhanced cell spreading and membrane ruffling. These results suggest that Src-mediated RhoGDI phosphorylation is a novel physiological mechanism for regulating Rho GTPase cytosol membrane–cycling and activity.


Author(s):  
Robert Beal ◽  
Ana Alonso-Carriazo Fernandez ◽  
Dimitris K. Grammatopoulos ◽  
Karl Matter ◽  
Maria S. Balda

Coordination of cell-cell adhesion, actomyosin dynamics and gene expression is crucial for morphogenetic processes underlying tissue and organ development. Rho GTPases are main regulators of the cytoskeleton and adhesion. They are activated by guanine nucleotide exchange factors in a spatially and temporally controlled manner. However, the roles of these Rho GTPase activators during complex developmental processes are still poorly understood. ARHGEF18/p114RhoGEF is a tight junction-associated RhoA activator that forms complexes with myosin II, and regulates actomyosin contractility. Here we show that p114RhoGEF/ARHGEF18 is required for mouse syncytiotrophoblast differentiation and placenta development. In vitro and in vivo experiments identify that p114RhoGEF controls expression of AKAP12, a protein regulating protein kinase A (PKA) signaling, and is required for PKA-induced actomyosin remodeling, cAMP-responsive element binding protein (CREB)-driven gene expression of proteins required for trophoblast differentiation, and, hence, trophoblast cell-cell fusion. Our data thus indicate that p114RhoGEF links actomyosin dynamics and cell-cell junctions to PKA/CREB signaling, gene expression and cell-cell fusion.


Sign in / Sign up

Export Citation Format

Share Document