Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 180
1.
Cell Mol Life Sci ; 81(1): 216, 2024 May 13.
Article En | MEDLINE | ID: mdl-38740643

p50RhoGAP is a key protein that interacts with and downregulates the small GTPase RhoA. p50RhoGAP is a multifunctional protein containing the BNIP-2 and Cdc42GAP Homology (BCH) domain that facilitates protein-protein interactions and lipid binding and the GAP domain that regulates active RhoA population. We recently solved the structure of the BCH domain from yeast p50RhoGAP (YBCH) and showed that it maintains the adjacent GAP domain in an auto-inhibited state through the ß5 strand. Our previous WT YBCH structure shows that a unique kink at position 116 thought to be made by a proline residue between alpha helices α6 and α7 is essential for the formation of intertwined dimer from asymmetric monomers. Here we sought to establish the role and impact of this Pro116. However, the kink persists in the structure of P116A mutant YBCH domain, suggesting that the scaffold is not dictated by the proline residue at this position. We further identified Tyr124 (or Tyr188 in HBCH) as a conserved residue in the crucial ß5 strand. Extending to the human ortholog, when substituted to acidic residues, Tyr188D or Tyr188E, we observed an increase in RhoA binding and self-dimerization, indicative of a loss of inhibition of the GAP domain by the BCH domain. These results point to distinct roles and impact of the non-conserved and conserved amino acid positions in regulating the structural and functional complexity of the BCH domain.


Proline , Proline/metabolism , Proline/chemistry , Proline/genetics , Tyrosine/metabolism , Tyrosine/chemistry , Tyrosine/genetics , Protein Domains , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/chemistry , Models, Molecular , Conserved Sequence , Humans , Protein Binding
2.
Nat Commun ; 14(1): 3733, 2023 06 23.
Article En | MEDLINE | ID: mdl-37353478

Transient receptor potential (TRP) channel TRPV4 is a polymodal cellular sensor that responds to moderate heat, cell swelling, shear stress, and small-molecule ligands. It is involved in thermogenesis, regulation of vascular tone, bone homeostasis, renal and pulmonary functions. TRPV4 is implicated in neuromuscular and skeletal disorders, pulmonary edema, and cancers, and represents an important drug target. The cytoskeletal remodeling GTPase RhoA has been shown to suppress TRPV4 activity. Here, we present a structure of the human TRPV4-RhoA complex that shows RhoA interaction with the membrane-facing surface of the TRPV4 ankyrin repeat domains. The contact interface reveals residues that are mutated in neuropathies, providing an insight into the disease pathogenesis. We also identify the binding sites of the TRPV4 agonist 4α-PDD and the inhibitor HC-067047 at the base of the S1-S4 bundle, and show that agonist binding leads to pore opening, while channel inhibition involves a π-to-α transition in the pore-forming helix S6. Our structures elucidate the interaction interface between hTRPV4 and RhoA, as well as residues at this interface that are involved in TRPV4 disease-causing mutations. They shed light on TRPV4 activation and inhibition and provide a template for the design of future therapeutics for treatment of TRPV4-related diseases.


TRPV Cation Channels , rhoA GTP-Binding Protein , Humans , Ankyrin Repeat , TRPV Cation Channels/chemistry , rhoA GTP-Binding Protein/chemistry
3.
Nat Commun ; 14(1): 3732, 2023 06 23.
Article En | MEDLINE | ID: mdl-37353484

Crosstalk between ion channels and small GTPases is critical during homeostasis and disease, but little is known about the structural underpinnings of these interactions. TRPV4 is a polymodal, calcium-permeable cation channel that has emerged as a potential therapeutic target in multiple conditions. Gain-of-function mutations also cause hereditary neuromuscular disease. Here, we present cryo-EM structures of human TRPV4 in complex with RhoA in the ligand-free, antagonist-bound closed, and agonist-bound open states. These structures reveal the mechanism of ligand-dependent TRPV4 gating. Channel activation is associated with rigid-body rotation of the intracellular ankyrin repeat domain, but state-dependent interaction with membrane-anchored RhoA constrains this movement. Notably, many residues at the TRPV4-RhoA interface are mutated in disease and perturbing this interface by introducing mutations into either TRPV4 or RhoA increases TRPV4 channel activity. Together, these results suggest that RhoA serves as an auxiliary subunit for TRPV4, regulating TRPV4-mediated calcium homeostasis and disruption of TRPV4-RhoA interactions can lead to TRPV4-related neuromuscular disease. These insights will help facilitate TRPV4 therapeutics development.


TRPV Cation Channels , rhoA GTP-Binding Protein , Humans , Ankyrin Repeat , Calcium/metabolism , Mutation , TRPV Cation Channels/chemistry , rhoA GTP-Binding Protein/chemistry
4.
PLoS Negl Trop Dis ; 15(11): e0009503, 2021 11.
Article En | MEDLINE | ID: mdl-34843489

BACKGROUND: Schistosoma mansoni histone deacetylase 8 (SmHDAC8) has elicited considerable interest as a target for drug discovery. Invalidation of its transcripts by RNAi leads to impaired survival of the worms in infected mice and its inhibition causes cell apoptosis and death. To determine why it is a promising therapeutic target the study of the currently unknown cellular signaling pathways involving this enzyme is essential. Protein partners of SmHDAC8 were previously identified by yeast two-hybrid (Y2H) cDNA library screening and by mass spectrometry (MS) analysis. Among these partners we characterized SmRho1, the schistosome orthologue of human RhoA GTPase, which is involved in the regulation of the cytoskeleton. In this work, we validated the interaction between SmHDAC8 and SmRho1 and explored the role of the lysine deacetylase in cytoskeletal regulation. METHODOLOGY/PRINCIPAL FINDINGS: We characterized two isoforms of SmRho1, SmRho1.1 and SmRho1.2. Co- immunoprecipitation (Co-IP)/Mass Spectrometry (MS) analysis identified SmRho1 partner proteins and we used two heterologous expression systems (Y2H assay and Xenopus laevis oocytes) to study interactions between SmHDAC8 and SmRho1 isoforms. To confirm SmHDAC8 and SmRho1 interaction in adult worms and schistosomula, we performed Co-IP experiments and additionally demonstrated SmRho1 acetylation using a Nano LC-MS/MS approach. A major impact of SmHDAC8 in cytoskeleton organization was documented by treating adult worms and schistosomula with a selective SmHDAC8 inhibitor or using RNAi followed by confocal microscopy. CONCLUSIONS/SIGNIFICANCE: Our results suggest that SmHDAC8 is involved in cytoskeleton organization via its interaction with the SmRho1.1 isoform. The SmRho1.2 isoform failed to interact with SmHDAC8, but did specifically interact with SmDia suggesting the existence of two distinct signaling pathways regulating S. mansoni cytoskeleton organization via the two SmRho1 isoforms. A specific interaction between SmHDAC8 and the C-terminal moiety of SmRho1.1 was demonstrated, and we showed that SmRho1 is acetylated on K136. SmHDAC8 inhibition or knockdown using RNAi caused extensive disruption of schistosomula actin cytoskeleton.


GTP Phosphohydrolases/chemistry , Histone Deacetylases/chemistry , Schistosoma mansoni/metabolism , rhoA GTP-Binding Protein/chemistry , Acetylation , Animals , Female , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Oocytes , RNA Interference , Schistosoma mansoni/genetics , Tandem Mass Spectrometry , Xenopus laevis , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
5.
Mol Cell ; 81(22): 4622-4634.e8, 2021 11 18.
Article En | MEDLINE | ID: mdl-34551282

AKT is a serine/threonine kinase that plays an important role in metabolism, cell growth, and cytoskeletal dynamics. AKT is activated by two kinases, PDK1 and mTORC2. Although the regulation of PDK1 is well understood, the mechanism that controls mTORC2 is unknown. Here, by investigating insulin receptor signaling in human cells and biochemical reconstitution, we found that insulin induces the activation of mTORC2 toward AKT by assembling a supercomplex with KRAS4B and RHOA GTPases, termed KARATE (KRAS4B-RHOA-mTORC2 Ensemble). Insulin-induced KARATE assembly is controlled via phosphorylation of GTP-bound KRAS4B at S181 and GDP-bound RHOA at S188 by protein kinase A. By developing a KARATE inhibitor, we demonstrate that KRAS4B-RHOA interaction drives KARATE formation. In adipocytes, KARATE controls insulin-dependent translocation of the glucose transporter GLUT4 to the plasma membrane for glucose uptake. Thus, our work reveals a fundamental mechanism that activates mTORC2 toward AKT in insulin-regulated glucose homeostasis.


Cyclic AMP-Dependent Protein Kinases/chemistry , Glucose/metabolism , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 2/chemistry , rhoA GTP-Binding Protein/chemistry , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Dictyostelium , Glucose Transporter Type 4/metabolism , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , HEK293 Cells , Humans , Mice , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism
6.
Int J Mol Sci ; 22(18)2021 Sep 20.
Article En | MEDLINE | ID: mdl-34576292

The cytotoxic necrotizing factor 1 (CNF1) toxin from uropathogenic Escherichia coli constitutively activates Rho GTPases by catalyzing the deamidation of a critical glutamine residue located in the switch II (SWII). In crystallographic structures of the CNF1 catalytic domain (CNF1CD), surface-exposed P768 and P968 peptidyl-prolyl imide bonds (X-Pro) adopt an unusual cis conformation. Here, we show that mutation of each proline residue into glycine abrogates CNF1CD in vitro deamidase activity, while mutant forms of CNF1 remain functional on RhoA in cells. Using molecular dynamics simulations coupled to protein-peptide docking, we highlight the long-distance impact of peptidyl-prolyl cis-trans isomerization on the network of interactions between the loops bordering the entrance of the catalytic cleft. The energetically favorable isomerization of P768 compared with P968, induces an enlargement of loop L1 that fosters the invasion of CNF1CD catalytic cleft by a peptide encompassing SWII of RhoA. The connection of the P968 cis isomer to the catalytic cysteine C866 via a ladder of stacking interactions is alleviated along the cis-trans isomerization. Finally, the cis-trans conversion of P768 favors a switch of the thiol side chain of C866 from a resting to an active orientation. The long-distance impact of peptidyl-prolyl cis-trans isomerizations is expected to have implications for target modification.


Bacterial Toxins/chemistry , Catalytic Domain , Escherichia coli Proteins/chemistry , Molecular Dynamics Simulation , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Isomerism , Molecular Docking Simulation , Protein Binding , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism
7.
STAR Protoc ; 2(2): 100541, 2021 06 18.
Article En | MEDLINE | ID: mdl-34036285

Ras GTPases in complex with Guanosine triphosphate (GTP) or GTP analog exhibit dynamic equilibrium between two interconvertible conformations-an inactive state 1 and an active state 2. Unlike Ras, it remains unclear if the GTP-bound form of Rho GTPases also exhibits multiple conformational states. Here, we describe a protocol for structural and biochemical analyses of RhoA GTPase. This protocol can be adapted for the characterization of other Rho GTPases. For details on the use and execution of this protocol, please refer to Lin et al. (2021).


rho GTP-Binding Proteins , Escherichia coli , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/isolation & purification , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/isolation & purification , rhoA GTP-Binding Protein/metabolism
8.
Structure ; 29(6): 553-563.e5, 2021 06 03.
Article En | MEDLINE | ID: mdl-33497604

By using 31P NMR, we present evidence that the Rho family GTPase RhoA, similar to Ras GTPases, exists in an equilibrium of conformations when bound to GTP. High-resolution crystal structures of RhoA bound to the GTP analog GMPPNP and to GDP show that they display a similar overall inactive conformation. In contrast to the previously reported crystal structures of GTP analog-bound forms of two RhoA dominantly active mutants (G14V and Q63L), GMPPNP-bound RhoA assumes an open conformation in the Switch I loop with a previously unseen interaction between the γ-phosphate and Pro36, instead of the canonical Thr37. Molecular dynamics simulations found that the oncogenic RhoAG14V mutant displays a reduced flexibility in the Switch regions, consistent with a crystal structure of GDP-bound RhoAG14V. Thus, GDP- and GTP-bound RhoA can present similar inactive conformations, and the molecular dynamics in the Switch regions are likely to have a role in RhoA activation.


Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism , Crystallography, X-Ray , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Humans , Models, Molecular , Molecular Dynamics Simulation , Mutation , Protein Conformation , rhoA GTP-Binding Protein/genetics
9.
Curr Stem Cell Res Ther ; 15(7): 607-613, 2020.
Article En | MEDLINE | ID: mdl-32223738

Gastric cancer (GC) is the fourth-most common cancer in the world, with an estimated 1.034 million new cases in 2015, and the third-highest cause of cancer deaths, estimated at 785,558, in 2014. Early diagnosis and treatment greatly affect the survival rate in patients with GC: the 5-year survival rate of early GC reaches 90%-95%, while the mortality rate significantly increases if GC develops to the late stage. Recently, studies for the role of RhoA in the diseases have become a hot topic, especially in the development of tumors. A study found that RhoA can regulate actin polymerization, cell adhesion, motor-myosin, cell transformation, and the ability to participate in the activities of cell movement, proliferation, migration, which are closely related to the invasion and metastasis of tumor cells. However, the specific role of RhoA in tumor cells remains to be studied. Therefore, our current study aimed to briefly review the role of RhoA in GC, especially for its associated signaling pathways involved in the GC progression.


Signal Transduction , Stomach Neoplasms/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Humans , Integrins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , rhoA GTP-Binding Protein/chemistry
10.
Pharmacogenomics J ; 20(4): 601-612, 2020 08.
Article En | MEDLINE | ID: mdl-32015453

Previously, we identified Ras homologous A (RHOA) as a major signaling hub in gastric cancer (GC), the third most common cause of cancer death in the world, prompting us to rationally design an efficacious inhibitor of this oncogenic GTPase. Here, based on that previous work, we extend those computational analyses to further pharmacologically optimize anti-RHOA hydrazide derivatives for greater anti-GC potency. Two of these, JK-136 and JK-139, potently inhibited cell viability and migration/invasion of GC cell lines, and mouse xenografts, diversely expressing RHOA. Moreover, JK-136's binding affinity for RHOA was >140-fold greater than Rhosin, a nonclinical RHOA inhibitor. Network analysis of JK-136/-139 vs. Rhosin treatments indicated downregulation of the sphingosine-1-phosphate, as an emerging cancer metabolic pathway in cell migration and motility. We assert that identifying and targeting oncogenic signaling hubs, such as RHOA, represents an emerging strategy for the design, characterization, and translation of new antineoplastics, against gastric and other cancers.


Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Drug Design , Stomach Neoplasms/drug therapy , rhoA GTP-Binding Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Mice , Mice, SCID , Molecular Docking Simulation/methods , Protein Structure, Secondary , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays/methods , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism
11.
Proc Natl Acad Sci U S A ; 117(2): 1027-1035, 2020 01 14.
Article En | MEDLINE | ID: mdl-31888991

Epithelial cell transforming 2 (Ect2) protein activates Rho GTPases and controls cytokinesis and many other cellular processes. Dysregulation of Ect2 is associated with various cancers. Here, we report the crystal structure of human Ect2 and complementary mechanistic analyses. The data show the C-terminal PH domain of Ect2 folds back and blocks the canonical RhoA-binding site at the catalytic center of the DH domain, providing a mechanism of Ect2 autoinhibition. Ect2 is activated by binding of GTP-bound RhoA to the PH domain, which suggests an allosteric mechanism of Ect2 activation and a positive-feedback loop reinforcing RhoA signaling. This bimodal RhoA binding of Ect2 is unusual and was confirmed with Förster resonance energy transfer (FRET) and hydrogen-deuterium exchange mass spectrometry (HDX-MS) analyses. Several recurrent cancer-associated mutations map to the catalytic and regulatory interfaces, and dysregulate Ect2 in vitro and in vivo. Together, our findings provide mechanistic insights into Ect2 regulation in normal cells and under disease conditions.


Epithelial Cells/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Binding Sites , Cytokinesis/physiology , Fluorescence Resonance Energy Transfer , Gene Knockdown Techniques , Humans , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Protein Conformation , Protein Domains , Proto-Oncogene Proteins/genetics , Signal Transduction , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism
12.
Hum Mutat ; 41(3): 591-599, 2020 03.
Article En | MEDLINE | ID: mdl-31821646

RHOA is a member of the Rho family of GTPases that are involved in fundamental cellular processes including cell adhesion, migration, and proliferation. RHOA can stimulate the formation of stress fibers and focal adhesions and is a key regulator of actomyosin dynamics in various tissues. In a Genematcher-facilitated collaboration, we were able to identify four unrelated individuals with a specific phenotype characterized by hypopigmented areas of the skin, dental anomalies, body asymmetry, and limb length discrepancy due to hemihypotrophy of one half of the body, as well as brain magnetic resonance imaging (MRI) anomalies. Using whole-exome and ultra-deep amplicon sequencing and comparing genomic data of affected and unaffected areas of the skin, we discovered that all four individuals carried the identical RHOA missense variant, c.139G>A; p.Glu47Lys, in a postzygotic state. Molecular modeling and in silico analysis of the affected p.Glu47Lys residue in RHOA indicated that this exchange is predicted to specifically alter the interaction of RHOA with its downstream effectors containing a PKN-type binding domain and thereby disrupts its ability to activate signaling. Our findings indicate that the recurrent postzygotic RHOA missense variant p.Glu47Lys causes a specific mosaic disorder in humans.


Alleles , Codon , Genetic Association Studies , Genetic Variation , Neural Plate/metabolism , Phenotype , rhoA GTP-Binding Protein/genetics , Adolescent , Adult , Brain/abnormalities , Brain/diagnostic imaging , Child , Child, Preschool , Female , Humans , Magnetic Resonance Imaging , Models, Molecular , Neural Plate/abnormalities , Neural Plate/embryology , Protein Conformation , Structure-Activity Relationship , Young Adult , rhoA GTP-Binding Protein/chemistry
13.
J Biol Chem ; 295(2): 645-656, 2020 01 10.
Article En | MEDLINE | ID: mdl-31806702

Deleted-in-liver cancer 1 (DLC1) exerts its tumor suppressive function mainly through the Rho-GTPase-activating protein (RhoGAP) domain. When activated, the domain promotes the hydrolysis of RhoA-GTP, leading to reduced cell migration. DLC1 is kept in an inactive state by an intramolecular interaction between its RhoGAP domain and the DLC1 sterile α motif (SAM) domain. We have shown previously that this autoinhibited state of DLC1 may be alleviated by tensin-3 (TNS3) or PTEN. We show here that the TNS3/PTEN-DLC1 interactions are mediated by the C2 domains of the former and the SAM domain of the latter. Intriguingly, the DLC1 SAM domain was capable of binding to specific peptide motifs within the C2 domains. Indeed, peptides containing the binding motifs were highly effective in blocking the C2-SAM domain-domain interaction. Importantly, when fused to the tat protein-transduction sequence and subsequently introduced into cells, the C2 peptides potently promoted the RhoGAP function in DLC1, leading to decreased RhoA activation and reduced tumor cell growth in soft agar and migration in response to growth factor stimulation. To facilitate the development of the C2 peptides as potential therapeutic agents, we created a cyclic version of the TNS3 C2 domain-derived peptide and showed that this peptide readily entered the MDA-MB-231 breast cancer cells and effectively inhibited their migration. Our work shows, for the first time, that the SAM domain is a peptide-binding module and establishes the framework on which to explore DLC1 SAM domain-binding peptides as potential therapeutic agents for cancer treatment.


Breast Neoplasms/metabolism , Cell Proliferation , GTPase-Activating Proteins/metabolism , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Female , GTPase-Activating Proteins/chemistry , HEK293 Cells , Humans , Models, Molecular , PTEN Phosphohydrolase/chemistry , PTEN Phosphohydrolase/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Maps , Sterile Alpha Motif , Tensins/chemistry , Tensins/metabolism , Tumor Suppressor Proteins/chemistry , rhoA GTP-Binding Protein/chemistry
14.
Biosci Rep ; 40(1)2020 01 31.
Article En | MEDLINE | ID: mdl-31844879

C3larvinA is a putative virulence factor produced by Paenibacillus larvae enterobacterial-repetitive-intergenic-consensus (ERIC) III/IV (strain 11-8051). Biochemical, functional and structural analyses of C3larvinA revealed that it belongs to the C3-like mono-ADP-ribosylating toxin subgroup. Mammalian RhoA was the target substrate for its transferase activity suggesting that it may be the biological target of C3larvinA. The kinetic parameters of the NAD+ substrate for the transferase (KM = 75 ± 10 µM) and glycohydrolase (GH) (KM = 107 ± 20 µM) reactions were typical for a C3-like bacterial toxin, including the Plx2A virulence factor from Paenibacillus larvae ERIC I. Upon cytoplasmic expression in yeast, C3larvinA caused a growth-defective phenotype indicating that it is an active C3-like toxin and is cytotoxic to eukaryotic cells. The catalytic variant of the Q187-X-E189 motif in C3larvinA showed no cytotoxicity toward yeast confirming that the cytotoxicity of this factor depends on its enzymatic activity. A homology consensus model of C3larvinA with NAD+ substrate was built on the structure of Plx2A, provided additional confirmation that C3larvinA is a member of the C3-like mono-ADP-ribosylating toxin subgroup. A homology model of C3larvinA with NADH and RhoA was built on the structure of the C3cer-NADH-RhoA complex which provided further evidence that C3larvinA is a C3-like toxin that shares an identical catalytic mechanism with C3cer from Bacillus cereus. C3larvinA induced actin cytoskeleton reorganization in murine macrophages, whereas in insect cells, vacuolization and bi-nucleated cells were observed. These cellular effects are consistent with C3larvinA disrupting RhoA function by covalent modification that is shared among C3-like bacterial toxins.


ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , Bees/microbiology , Paenibacillus larvae/enzymology , Virulence Factors/metabolism , rhoA GTP-Binding Protein/metabolism , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Actin Cytoskeleton/enzymology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Host-Pathogen Interactions , Kinetics , Macrophages/enzymology , Mutation , Paenibacillus larvae/genetics , Paenibacillus larvae/pathogenicity , Protein Conformation , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sf9 Cells , Spodoptera , Structure-Activity Relationship , Substrate Specificity , Virulence , Virulence Factors/chemistry , Virulence Factors/genetics , rhoA GTP-Binding Protein/chemistry
15.
Int J Mol Sci ; 20(24)2019 Dec 12.
Article En | MEDLINE | ID: mdl-31842335

In recent years, hypersensitivity reactions to the Shuanghuanglian injection have attracted broad attention. However, the componential chief culprits inducing the reactions and the underlying mechanisms involved have not been completely defined. In this study, we used a combination of approaches based on the mouse model, human umbilical vein endothelial cell monolayer, real-time cellular monitoring, immunoblot analysis, pharmacological inhibition, and molecular docking. We demonstrated that forsythoside A and forsythoside B contributed to Shuanghuanglian injection-induced pseudoallergic reactions through activation of the RhoA/ROCK signaling pathway. Forsythoside A and forsythoside B could trigger dose-dependent vascular leakage in mice. Moreover, forsythoside A and forsythoside B slightly elicited mast cell degranulation. Correspondingly, treatment with forsythoside A and forsythoside B disrupted the endothelial barrier and augmented the expression of GTP-RhoA, p-MYPT1, and p-MLC2 in a concentration-dependent manner. Additionally, the ROCK inhibitor effectively alleviated forsythoside A/forsythoside B-induced hyperpermeability in both the endothelial cells and mice. Similar responses were not observed in the forsythoside E-treated animals and cells. These differences may be related to the potential of the tested compounds to react with RhoA-GTPγS and form stable interactions. This study innovatively revealed that some forsythosides may cause vascular leakage, and therefore, limiting their contents in injections should be considered.


Caffeic Acids/pharmacology , Drugs, Chinese Herbal/chemistry , Glucosides/pharmacology , Glycosides/pharmacology , Signal Transduction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Caffeic Acids/chemistry , Capillary Permeability/drug effects , Capillary Permeability/immunology , Cell Degranulation , Drugs, Chinese Herbal/administration & dosage , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glucosides/chemistry , Glycosides/chemistry , Humans , Mast Cells/immunology , Mast Cells/metabolism , Mice , Models, Molecular , Molecular Conformation , Molecular Structure , Structure-Activity Relationship , rho-Associated Kinases/chemistry , rhoA GTP-Binding Protein/chemistry
16.
Cell Death Dis ; 10(10): 770, 2019 10 10.
Article En | MEDLINE | ID: mdl-31601793

Ovarian cancer remains the most lethal gynecologic malignancy with late detection and acquired chemoresistance. Advanced understanding of the pathophysiology and novel treatment strategies are urgently required. A growing body of proteomic investigations suggest that phosphorylation has a pivotal role in the regulation of ovarian cancer associated signaling pathways. Matrine has been extensively studied for its potent anti-tumor activities. However, its effect on ovarian cancer cells and underlying molecular mechanisms remain unclear. Herein we showed that matrine treatment inhibited the development and progression of ovarian cancer cells by regulating proliferation, apoptosis, autophagy, invasion and angiogenesis. Matrine treatment retarded the cancer associated signaling transduction by decreasing the phosphorylation levels of ERK1/2, MEK1/2, PI3K, Akt, mTOR, FAK, RhoA, VEGFR2, and Tie2 in vitro and in vivo. Moreover, matrine showed excellent antitumor effect on chemoresistant ovarian cancer cells. No obvious toxic side effects were observed in matrine-administrated mice. As the natural agent, matrine has the potential to be the targeting drug against ovarian cancer cells with the advantages of overcoming the chemotherapy resistance and decreasing the toxic side effects.


Alkaloids/therapeutic use , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Quinolizines/therapeutic use , Alkaloids/adverse effects , Alkaloids/pharmacology , Animals , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/chemistry , Focal Adhesion Kinase 1/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Quinolizines/adverse effects , Quinolizines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , Transplantation, Heterologous , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism , Matrines
17.
Cell Death Dis ; 10(6): 450, 2019 06 06.
Article En | MEDLINE | ID: mdl-31171774

Non-obstructive azoospermia (NOA) severely affects male infertility, however, the deep mechanisms of this disease are rarely interpreted. In this study, we find that undifferentiated spermatogonial stem cells (SSCs) still exist in the basal compartment of the seminiferous tubules and the blood-testis barrier (BTB) formed by the interaction of neighbor Sertoli cells (SCs) is incomplete in NOA patients with spermatogenic maturation arrest. The adhesions between SCs and germ cells (GCs) are also broken in NOA patients. Meanwhile, the expression level of geranylgeranyl diphosphate synthase (Ggpps), a key enzyme in mevalonate metabolic pathway, is lower in NOA patients than that in obstructive azoospermia (OA) patients. After Ggpps deletion specifically in SCs, the mice are infertile and the phenotype of the SC-Ggpps-/- mice is similar to the NOA patients, where the BTB and the SC-GC adhesions are severely destroyed. Although SSCs are still found in the basal compartment of the seminiferous tubules, fewer mature spermatocyte and spermatid are found in SC-Ggpps-/- mice. Further examination suggests that the defect is mediated by the aberrant protein isoprenylation of RhoA and Ras family after Ggpps deletion. The exciting finding is that when the knockout mice are injected with berberine, the abnormal cell adhesions are ameliorated and spermatogenesis is partially restored. Our data suggest that the reconstruction of disrupted BTB is an effective treatment strategy for NOA patients with spermatogenic maturation arrest and hypospermatogenesis.


Azoospermia/metabolism , Blood-Testis Barrier/metabolism , Farnesyltranstransferase/metabolism , Multienzyme Complexes/metabolism , Protein Prenylation , Spermatogenesis/genetics , ras Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Azoospermia/enzymology , Berberine/pharmacology , Blood-Testis Barrier/drug effects , Cells, Cultured , Farnesyltranstransferase/genetics , Germ Cells/metabolism , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Multienzyme Complexes/genetics , Sertoli Cells/enzymology , Sertoli Cells/metabolism , Spermatocytes/metabolism , Spermatogenesis/drug effects , Testis/metabolism , Tight Junctions/genetics , ras Proteins/chemistry , ras Proteins/genetics , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
18.
Proc Natl Acad Sci U S A ; 116(9): 3594-3603, 2019 02 26.
Article En | MEDLINE | ID: mdl-30808751

The contractile actin cortex is a thin layer of filamentous actin, myosin motors, and regulatory proteins beneath the plasma membrane crucial to cytokinesis, morphogenesis, and cell migration. However, the factors regulating actin assembly in this compartment are not well understood. Using the Dictyostelium model system, we show that the three Diaphanous-related formins (DRFs) ForA, ForE, and ForH are regulated by the RhoA-like GTPase RacE and synergize in the assembly of filaments in the actin cortex. Single or double formin-null mutants displayed only moderate defects in cortex function whereas the concurrent elimination of all three formins or of RacE caused massive defects in cortical rigidity and architecture as assessed by aspiration assays and electron microscopy. Consistently, the triple formin and RacE mutants encompassed large peripheral patches devoid of cortical F-actin and exhibited severe defects in cytokinesis and multicellular development. Unexpectedly, many forA- /E-/H- and racE- mutants protruded efficiently, formed multiple exaggerated fronts, and migrated with morphologies reminiscent of rapidly moving fish keratocytes. In 2D-confinement, however, these mutants failed to properly polarize and recruit myosin II to the cell rear essential for migration. Cells arrested in these conditions displayed dramatically amplified flow of cortical actin filaments, as revealed by total internal reflection fluorescence (TIRF) imaging and iterative particle image velocimetry (PIV). Consistently, individual and combined, CRISPR/Cas9-mediated disruption of genes encoding mDia1 and -3 formins in B16-F1 mouse melanoma cells revealed enhanced frequency of cells displaying multiple fronts, again accompanied by defects in cell polarization and migration. These results suggest evolutionarily conserved functions for formin-mediated actin assembly in actin cortex mechanics.


Actin Cytoskeleton/genetics , Carrier Proteins/genetics , Contractile Proteins/genetics , Melanoma, Experimental/genetics , Actin Cytoskeleton/chemistry , Actins/genetics , Animals , CRISPR-Cas Systems , Cell Movement/genetics , Cell Polarity/genetics , Contractile Proteins/chemistry , Dictyostelium/genetics , Disease Models, Animal , Formins , Humans , Melanoma, Experimental/pathology , Mice , Microscopy, Electron , Muscle Contraction/genetics , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
19.
Sci Signal ; 12(569)2019 02 19.
Article En | MEDLINE | ID: mdl-30783010

The C-terminal guanine nucleotide exchange factor (GEF) module of Trio (TrioC) transfers signals from the Gαq/11 subfamily of heterotrimeric G proteins to the small guanosine triphosphatase (GTPase) RhoA, enabling Gαq/11-coupled G protein-coupled receptors (GPCRs) to control downstream events, such as cell motility and gene transcription. This conserved signal transduction axis is crucial for tumor growth in uveal melanoma. Previous studies indicate that the GEF activity of the TrioC module is autoinhibited, with release of autoinhibition upon Gαq/11 binding. Here, we determined the crystal structure of TrioC in its basal state and found that the pleckstrin homology (PH) domain interacts with the Dbl homology (DH) domain in a manner that occludes the Rho GTPase binding site, thereby suggesting the molecular basis of TrioC autoinhibition. Biochemical and biophysical assays revealed that disruption of the autoinhibited conformation destabilized and activated the TrioC module in vitro. Last, mutations in the DH-PH interface found in patients with cancer activated TrioC and, in the context of full-length Trio, led to increased abundance of guanosine triphosphate-bound RhoA (RhoA·GTP) in human cells. These mutations increase mitogenic signaling through the RhoA axis and, therefore, may represent cancer drivers operating in a Gαq/11-independent manner.


GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Protein Domains , Rho Guanine Nucleotide Exchange Factors/chemistry , rhoA GTP-Binding Protein/chemistry , Binding Sites/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Melanoma/genetics , Melanoma/pathology , Models, Molecular , Mutation , Protein Binding , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
20.
Hum Mol Genet ; 28(10): 1620-1628, 2019 05 15.
Article En | MEDLINE | ID: mdl-30608580

Missense mutations in the gene, MAP3K1, are a common cause of 46,XY gonadal dysgenesis, accounting for 15-20% of cases [Ostrer, 2014, Disorders of sex development (DSDs): an update. J. Clin. Endocrinol. Metab., 99, 1503-1509]. Functional studies demonstrated that all of these mutations cause a protein gain-of-function that alters co-factor binding and increases phosphorylation of the downstream MAP kinase pathway targets, MAPK11, MAP3K and MAPK1. This dysregulation of the MAP kinase pathway results in increased CTNNB1, increased expression of WNT4 and FOXL2 and decreased expression of SRY and SOX9. Unique and recurrent pathogenic mutations cluster in three semi-contiguous domains outside the kinase region of the protein, a newly identified N-terminal domain that shares homology with the Guanine Exchange Factor (residues Met164 to Glu231), a Plant HomeoDomain (residues Met442 to Trp495) and an ARMadillo repeat domain (residues Met566 to Glu862). Despite the presence of the mutation clusters and clinical data, there exists a dearth of mechanistic insights behind the development imbalance. In this paper, we use structural modeling and functional data of these mutations to understand alterations of the MAP3K1 protein and the effects on protein folding, binding and downstream target phosphorylation. We show that these mutations have differential effects on protein binding depending on the domains in which they occur. These mutations increase the binding of the RHOA, MAP3K4 and FRAT1 proteins and generally decrease the binding of RAC1. Thus, pathologies in MAP3K1 disrupt the balance between the pro-kinase activities of the RHOA and MAP3K4 binding partners and the inhibitory activity of RAC1.


Disorders of Sex Development/genetics , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 4/genetics , rac1 GTP-Binding Protein/genetics , Adaptor Proteins, Signal Transducing/genetics , Armadillo Domain Proteins/genetics , Disorder of Sex Development, 46,XY , Disorders of Sex Development/pathology , Female , Forkhead Box Protein L2/genetics , Gene Expression Regulation/genetics , Gonadal Dysgenesis, 46,XY/genetics , Gonadal Dysgenesis, 46,XY/pathology , Humans , MAP Kinase Kinase Kinase 1/chemistry , MAP Kinase Kinase Kinase 4/chemistry , MAP Kinase Signaling System/genetics , Male , Mutation, Missense/genetics , Protein Binding/genetics , Proto-Oncogene Proteins/genetics , Sex-Determining Region Y Protein/genetics , rac1 GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
...