Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
Sci Rep ; 11(1): 23285, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857846

ABSTRACT

Machine learning approaches have shown great promise in biology and medicine discovering hidden information to further understand complex biological and pathological processes. In this study, we developed a deep learning-based machine learning algorithm to meaningfully process image data and facilitate studies in vascular biology and pathology. Vascular injury and atherosclerosis are characterized by neointima formation caused by the aberrant accumulation and proliferation of vascular smooth muscle cells (VSMCs) within the vessel wall. Understanding how to control VSMC behaviors would promote the development of therapeutic targets to treat vascular diseases. However, the response to drug treatments among VSMCs with the same diseased vascular condition is often heterogeneous. Here, to identify the heterogeneous responses of drug treatments, we created an in vitro experimental model system using VSMC spheroids and developed a machine learning-based computational method called HETEROID (heterogeneous spheroid). First, we established a VSMC spheroid model that mimics neointima-like formation and the structure of arteries. Then, to identify the morphological subpopulations of drug-treated VSMC spheroids, we used a machine learning framework that combines deep learning-based spheroid segmentation and morphological clustering analysis. Our machine learning approach successfully showed that FAK, Rac, Rho, and Cdc42 inhibitors differentially affect spheroid morphology, suggesting that multiple drug responses of VSMC spheroid formation exist. Overall, our HETEROID pipeline enables detailed quantitative drug characterization of morphological changes in neointima formation, that occurs in vivo, by single-spheroid analysis.


Subject(s)
Machine Learning , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Atherosclerosis/pathology , Cells, Cultured , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/physiology , Humans , Neointima/pathology , Spheroids, Cellular/physiology , Vascular System Injuries/pathology , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/physiology , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/physiology
2.
Mol Cancer Ther ; 20(12): 2420-2432, 2021 12.
Article in English | MEDLINE | ID: mdl-34607932

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer, with a high predisposition for locally invasive and metastatic cancer. With the objective to reduce cancer metastasis, we developed small molecule inhibitors to target the drivers of metastasis, the Rho GTPases Rac and Cdc42. Of these, MBQ-167 inhibits both Rac and Cdc42 with IC50s of 103 and 78 nmol/L, respectively; and consequently, inhibits p21-activated kinase (PAK) signaling, metastatic cancer cell proliferation, migration, and mammosphere growth; induces cell-cycle arrest and apoptosis; and decreases HER2-type mammary fatpad tumor growth and metastasis (Humphries-Bickley and colleagues, 2017). Herein, we used nuclear magnetic resonance to show that MBQ-167 directly interacts with Rac1 to displace specific amino acids, and consequently inhibits Rac.GTP loading and viability in TNBC cell lines. Phosphokinome arrays in the MDA-MB-231 human TNBC cells show that phosphorylation status of kinases independent of the Rac/Cdc42/PAK pathway are not significantly changed following 200 nmol/L MBQ-167 treatment. Western blotting shows that initial increases in phospho-c-Jun and phospho-CREB in response to MBQ-167 are not sustained with prolonged exposure, as also confirmed by a decrease in their transcriptional targets. MBQ-167 inhibits tumor growth, and spontaneous and experimental metastasis in immunocompromised (human TNBC) and immunocompetent (mouse TNBC) models. Moreover, per oral administration of MBQ-167 at 100 mg/kg body weight is not toxic to immunocompetent BALB/c mice and has a half-life of 4.6 hours in plasma. These results highlight the specificity, potency, and bioavailability of MBQ-167, and support its clinical potential as a TNBC therapeutic.


Subject(s)
Triple Negative Breast Neoplasms/genetics , cdc42 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Humans , Mice , Mice, SCID , Triple Negative Breast Neoplasms/pathology
3.
Sci Rep ; 11(1): 17990, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504210

ABSTRACT

Macrothrombocytopenia is a common pathology of missense mutations in genes regulating actin dynamics. Takenouchi-Kosaki syndrome (TKS) harboring the c.191A > G, Tyr64Cys (Y64C) variant in Cdc42 exhibits a variety of clinical manifestations, including immunological and hematological anomalies. In the present study, we investigated the functional abnormalities of the Y64C mutant in HEK293 cells and elucidated the mechanism of macrothrombocytopenia, one of the symptoms of TKS patients, by monitoring the production of platelet-like particles (PLP) using MEG-01 cells. We found that the Y64C mutant was concentrated at the membrane compartment due to impaired binding to Rho-GDI and more active than the wild-type. The Y64C mutant also had lower association with its effectors Pak1/2 and N-WASP. Y64C mutant-expressing MEG-01 cells demonstrated short cytoplasmic protrusions with aberrant F-actin and microtubules, and reduced PLP production. This suggested that the Y64C mutant facilitates its activity and membrane localization, resulting in impaired F-actin dynamics for proplatelet extension, which is necessary for platelet production. Furthermore, such dysfunction was ameliorated by either suppression of Cdc42 activity or prenylation using chemical inhibitors. Our study may lead to pharmacological treatments for TKS patients.


Subject(s)
Megakaryocytes/drug effects , Megakaryocytes/metabolism , Signal Transduction/drug effects , Thrombocytopenia/metabolism , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/metabolism , Actins/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Benzamides/pharmacology , Blood Platelets/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Membrane/metabolism , HEK293 Cells , Humans , Mutation , Protein Prenylation/drug effects , Pyrazoles/pharmacology , Signal Transduction/genetics , Sulfonamides/pharmacology , Syndrome , Thrombocytopenia/genetics , Thrombopoiesis/drug effects , Thrombopoiesis/genetics , Transfection , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/genetics , p21-Activated Kinases/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism
4.
FASEB J ; 35(8): e21723, 2021 08.
Article in English | MEDLINE | ID: mdl-34224609

ABSTRACT

Sperm acquire the ability to fertilize in a process called capacitation and undergo hyperactivation, a change in the motility pattern, which depends on Ca2+ transport by CatSper channels. CatSper is essential for fertilization and it is subjected to a complex regulation that is not fully understood. Here, we report that similar to CatSper, Cdc42 distribution in the principal piece is confined to four linear domains and this localization is disrupted in CatSper1-null sperm. Cdc42 inhibition impaired CatSper activity and other Ca2+ -dependent downstream events resulting in a severe compromise of the sperm fertilizing potential. We also demonstrate that Cdc42 is essential for CatSper function by modulating cAMP production by soluble adenylate cyclase (sAC), providing a new regulatory mechanism for the stimulation of CatSper by the cAMP-dependent pathway. These results reveal a broad mechanistic insight into the regulation of Ca2+ in mammalian sperm, a matter of critical importance in male infertility as well as in contraception.


Subject(s)
Calcium Channels/metabolism , Spermatozoa/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Calcium/metabolism , Calcium Channels/deficiency , Calcium Channels/genetics , Calcium Signaling , Cyclic AMP/metabolism , Female , Fertilization in Vitro , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Signal Transduction , Sperm Capacitation/physiology , Sperm Motility/physiology , Sperm Tail/metabolism , Spermatozoa/drug effects , Spermatozoa/ultrastructure , cdc42 GTP-Binding Protein/antagonists & inhibitors
5.
Biochem Soc Trans ; 49(3): 1443-1456, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34100887

ABSTRACT

Cdc42 is a member of the Rho family of small GTPases and a key regulator of the actin cytoskeleton, controlling cell motility, polarity and cell cycle progression. It signals downstream of the master regulator Ras and is essential for cell transformation by this potent oncogene. Overexpression of Cdc42 is observed in several cancers, where it is linked to poor prognosis. As a regulator of both cell architecture and motility, deregulation of Cdc42 is also linked to tumour metastasis. Like Ras, Cdc42 and other components of the signalling pathways it controls represent important potential targets for cancer therapeutics. In this review, we consider the progress that has been made targeting Cdc42, its regulators and effectors, including new modalities and new approaches to inhibition. Strategies under consideration include inhibition of lipid modification, modulation of Cdc42-GEF, Cdc42-GDI and Cdc42-effector interactions, and direct inhibition of downstream effectors.


Subject(s)
Actin Cytoskeleton/metabolism , Neoplasms/metabolism , Signal Transduction/physiology , cdc42 GTP-Binding Protein/metabolism , Aminoquinolines/therapeutic use , Animals , Benzamides/therapeutic use , Benzazepines/therapeutic use , Humans , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/genetics , Oximes/therapeutic use , Protein Binding/drug effects , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfonamides/therapeutic use , Thiourea/analogs & derivatives , Thiourea/therapeutic use , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics
6.
Sci Rep ; 11(1): 13170, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162972

ABSTRACT

Gene targeting of Cdc42 GTPase has been shown to inhibit platelet activation. In this study, we investigated a hypothesis that inhibition of Cdc42 activity by CASIN, a small molecule Cdc42 Activity-Specific INhibitor, may down regulate platelet activation and thrombus formation. We investigated the effects of CASIN on platelet activation in vitro and thrombosis in vivo. In human platelets, CASIN, but not its inactive analog Pirl7, blocked collagen induced activation of Cdc42 and inhibited phosphorylation of its downstream effector, PAK1/2. Moreover, addition of CASIN to washed human platelets inhibited platelet spreading on immobilized fibrinogen. Treatment of human platelets with CASIN inhibited collagen or thrombin induced: (a) ATP secretion and platelet aggregation; and (b) phosphorylation of Akt, ERK and p38-MAPK. Pre-incubation of platelets with Pirl7, an inactive analog of CASIN, failed to inhibit collagen induced aggregation. Washing of human platelets after incubation with CASIN eliminated its inhibitory effect on collagen induced aggregation. Intraperitoneal administration of CASIN to wild type mice inhibited ex vivo aggregation induced by collagen but did not affect the murine tail bleeding times. CASIN administration, prior to laser-induced injury in murine cremaster muscle arterioles, resulted in formation of smaller and unstable thrombi compared to control mice without CASIN treatment. These data suggest that pharmacologic targeting of Cdc42 by specific and reversible inhibitors may lead to the discovery of novel antithrombotic agents.


Subject(s)
Carbazoles/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thrombosis/prevention & control , cdc42 GTP-Binding Protein/antagonists & inhibitors , Abdominal Muscles/blood supply , Adenosine Triphosphate/metabolism , Animals , Arterioles , Carbazoles/administration & dosage , Drug Evaluation, Preclinical , Female , Humans , Lasers , Male , Mice , Mice, Inbred C57BL , P-Selectin/metabolism , Platelet Aggregation/drug effects , rac1 GTP-Binding Protein/antagonists & inhibitors
7.
Front Immunol ; 12: 619069, 2021.
Article in English | MEDLINE | ID: mdl-34108958

ABSTRACT

Natural killer (NK) cells are innate effector lymphocytes with strong antitumor effects against hematologic malignancies such as chronic lymphocytic leukemia (CLL). However, NK cells fail to control CLL progression on the long term. For effective lysis of their targets, NK cells use a specific cell-cell interface, known as the immunological synapse (IS), whose assembly and effector function critically rely on dynamic cytoskeletal changes in NK cells. Here we explored the role of CLL cell actin cytoskeleton during NK cell attack. We found that CLL cells can undergo fast actin cytoskeleton remodeling which is characterized by a NK cell contact-induced accumulation of actin filaments at the IS. Such polarization of the actin cytoskeleton was strongly associated with resistance against NK cell-mediated cytotoxicity and reduced amounts of the cell-death inducing molecule granzyme B in target CLL cells. Selective pharmacological targeting of the key actin regulator Cdc42 abrogated the capacity of CLL cells to reorganize their actin cytoskeleton during NK cell attack, increased levels of transferred granzyme B and restored CLL cell susceptibility to NK cell cytotoxicity. This resistance mechanism was confirmed in primary CLL cells from patients. In addition, pharmacological inhibition of actin dynamics in combination with blocking antibodies increased conjugation frequency and improved CLL cell elimination by NK cells. Together our results highlight the critical role of CLL cell actin cytoskeleton in driving resistance against NK cell cytotoxicity and provide new potential therapeutic point of intervention to target CLL immune escape.


Subject(s)
Actin Cytoskeleton/metabolism , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , cdc42 GTP-Binding Protein/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Fluorescent Antibody Technique , HLA-G Antigens/immunology , Humans , Immunological Synapses/immunology , Immunological Synapses/metabolism , Immunophenotyping , Killer Cells, Natural/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
8.
Front Immunol ; 12: 564720, 2021.
Article in English | MEDLINE | ID: mdl-33679729

ABSTRACT

Neutrophil granulocytes represent the first line of defense against invading pathogens. In addition to the production of Reactive Oxygen Species, degranulation, and phagocytosis, these specialized cells are able to extrude Neutrophil Extracellular Traps. Extensive work was done to elucidate the mechanism of this special form of cell death. However, the exact mechanisms are still not fully uncovered. Here we demonstrate that the small GTPase Cdc42 is a negative regulator of NET formation in primary human and murine neutrophils. We present a functional role for Cdc42 activity in NET formation that differs from the already described NETosis pathways. We show that Cdc42 deficiency induces NETs independent of the NADPH-oxidase but dependent on protein kinase C. Furthermore, we demonstrate that Cdc42 deficiency induces NETosis through activation of SK-channels and that mitochondria play a crucial role in this process. Our data therefore suggests a mechanistic role for Cdc42 activity in primary human neutrophils, and identify Cdc42 activity as a target to modulate the formation of Neutrophil Extracellular Traps.


Subject(s)
Extracellular Traps/metabolism , Mitochondria/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neutrophils/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Extracellular Traps/genetics , Humans , Mice, Knockout , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/genetics , Neutrophils/cytology , Potassium Channels, Calcium-Activated/metabolism , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics
9.
J Recept Signal Transduct Res ; 41(5): 504-510, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32998602

ABSTRACT

BACKGROUND: Cell division control (CDC) 42 has been involved in the regulation of diverse cancers. Macrophage recruitment plays an important role in the pathogenesis and development of tumor. However, it remains unclear whether CDC42 contributes to macrophage recruitment and lung tumorigenesis in vivo. METHODS: Small interference RNA (siRNA) was used to knock down CDC42 in the Lewis lung carcinoma (LLC)1. The invasion capability of CDC42 knockdown LLC1 cells was evaluated. LLC1 cells with CDC42 targeted small hairpin RNA (shRNA) were inoculated into C57BL/6 mice to establish the tumor-bearing animal model Tumor size and metastasis related proteins were measured. In addition, the invasion of macrophages in the tumor site as well as macrophage chemokine were also determined in the model. RESULTS: The capacity of invasion and metastasis of LLC1 cells significantly decreased when CDC42 was knocked down. When inoculated with CDC42 knockdown LLC1 cells in vivo, the tumor size and metastasis related proteins levels both decreased. The invasion capacity of macrophages and the associated macrophage chemokine were also significantly down-regulated. CONCLUSION: Our data suggest that the inhibition of CDC42 expression in lung cancer cells can significantly prevent the pathogenesis and development of tumor in an allograft tumor model in vivo, which might provide a novel therapeutic target and potential strategy for lung cancer treatment in the future.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Lewis Lung/prevention & control , Disease Models, Animal , Macrophages/immunology , RNA, Small Interfering/genetics , cdc42 GTP-Binding Protein/antagonists & inhibitors , Animals , Apoptosis , Carcinogenesis/immunology , Carcinogenesis/metabolism , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , cdc42 GTP-Binding Protein/genetics
10.
Biosci Rep ; 40(7)2020 07 31.
Article in English | MEDLINE | ID: mdl-32578854

ABSTRACT

Recently, Rho GTPases substrates include Rac (Rac1 and Rac2) and Cdc42 that have been reported to exert multiple cellular functions in osteoclasts, the most prominent of which includes regulating the dynamic actin cytoskeleton rearrangements. In addition, natural products and their molecular frameworks have a long tradition as valuable starting points for medicinal chemistry and drug discovery. Although currently, there are reports about the natural product, which could play a therapeutic role in bone loss diseases (osteoporosis and osteolysis) through the regulation of Rac1/2 and Cdc42 during osteoclasts cytoskeletal structuring. There have been several excellent studies for exploring the therapeutic potentials of various natural products for their role in inhibiting cancer cells migration and function via regulating the Rac1/2 and Cdc42. Herein in this review, we try to focus on recent advancement studies for extensively understanding the role of Rho GTPases substrates Rac1, Rac2 and Cdc42 in osteoclastogenesis, as well as therapeutic potentials of natural medicinal products for their properties on the regulation of Rac1, and/or Rac2 and Cdc42, which is in order to inspire drug discovery in regulating osteoclastogenesis.


Subject(s)
Biological Products/pharmacology , Osteogenesis/drug effects , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Biological Products/therapeutic use , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Movement/physiology , Drug Discovery , Humans , Models, Animal , Molecular Targeted Therapy/methods , Osteoblasts/drug effects , Osteoblasts/physiology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteolysis/drug therapy , Osteolysis/pathology , Osteoporosis/drug therapy , Osteoporosis/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , cdc42 GTP-Binding Protein/antagonists & inhibitors , rac GTP-Binding Proteins/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
11.
J Biol Chem ; 295(14): 4498-4512, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32071086

ABSTRACT

The small GTPase cell division cycle 42 (CDC42) plays essential roles in neurogenesis and brain development. Previously, using murine embryonic P19 cells as a model system, we showed that CDC42 stimulates mTOR complex 1 (mTORC1) activity and thereby up-regulates transcription factors required for the formation of neural progenitor cells. However, paradoxically, although endogenous CDC42 is required for both the initial transition of undifferentiated P19 cells to neural progenitors and their ultimate terminal differentiation into neurons, ectopic CDC42 overexpression promotes only the first stage of neurogenesis (i.e. the formation of neuroprogenitors) and not the second phase (differentiation into neurons). Here, using both P19 cells and mouse embryonic stem cells, we resolve this paradox, demonstrating that two splice variants of CDC42, differing only in nine amino acid residues in their very C-terminal regions, play distinct roles in neurogenesis. We found that a CDC42 splice variant that has a ubiquitous tissue distribution, termed here as CDC42u, specifically drives the formation of neuroprogenitor cells, whereas a brain-specific CDC42 variant, CDC42b, is essential for promoting the transition of neuroprogenitor cells to neurons. We further show that the specific roles of CDC42u and CDC42b in neurogenesis are due to their opposing effects on mTORC1 activity. Specifically, CDC42u stimulated mTORC1 activity and thereby induced neuroprogenitor formation, whereas CDC42b worked together with activated CDC42-associated kinase (ACK) in down-regulating mTOR expression and promoting neuronal differentiation. These findings highlight the remarkable functional specificities of two highly similar CDC42 splice variants in regulating distinct stages of neurogenesis.


Subject(s)
Neurogenesis/physiology , cdc42 GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Cell Differentiation , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nestin/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sequence Alignment , Signal Transduction , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics
12.
Eur Cell Mater ; 39: 48-64, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31917459

ABSTRACT

The roles of cell division control protein 42 homologue (CDC42) and actin polymerisation in regulating the phenotype of superficial-zone chondrocytes (SZCs) have been demonstrated in vitro; however, the signalling pathway(s) downstream have yet to be fully elucidated. The study hypothesis was that Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) act downstream to regulate proteoglycan 4 (PRG4) and tenascin C (TNC). Bovine SZCs grown in monolayer were treated with ML141 (CDC42 inhibitor) or the actin depolymerising agents, latrunculin B and cytochalasin D, to determine the effect on YAP/TAZ. Verteporfin (YAP/TAZ inhibitor) and YAP/TAZ siRNA-mediated knockdown were used to determine their role in regulating PRG4 and TNC. ML141 treatment reduced total YAP/TAZ protein, nuclear TAZ levels and the YAP/TAZ target gene, connective tissue growth factor (CTGF) mRNA levels. Latrunculin B decreased nuclear TAZ, while cytochalasin D treatment trended towards increased nuclear TAZ (p = 0.06), correlating with decreased and increased CTGF mRNA levels, respectively. Verteporfin treatment decreased PRG4 and TNC expression, with no effect on actin polymerisation. siRNA-mediated knockdown of YAP/TAZ revealed that PRG4 was regulated by YAP/TAZ while TNC was regulated by TAZ only. As cytochalasin D can activate myocardin-related transcription factor-A (MRTF-A), siRNA-mediated knockdown was performed to determine the role of MRTF-A in regulating YAP/TAZ. Although nuclear TAZ decreased, no significant changes in total protein levels were observed. Findings suggested that CDC42 and actin polymerisation regulated SZCs through multiple actin-regulated pathways. Understanding the regulation of these chondroprotective molecules may have important implications for prevention/treatment of osteoarthritis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens/metabolism , Chondrocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Proteoglycans/metabolism , Tenascin/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Cattle , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chondrocytes/drug effects , Phenotype , Protein Transport/drug effects , Trans-Activators/metabolism , Verteporfin/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/metabolism
13.
J Biol Chem ; 295(9): 2866-2884, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31959628

ABSTRACT

Aberrant Ras signaling drives 30% of cancers, and inhibition of the Rho family small GTPase signaling has been shown to combat Ras-driven cancers. Here, we present the discovery of a 16-mer cyclic peptide that binds to Cdc42 with nanomolar affinity. Affinity maturation of this sequence has produced a panel of derived candidates with increased affinity and modulated specificity for other closely-related small GTPases. The structure of the tightest binding peptide was solved by NMR, and its binding site on Cdc42 was determined. Addition of a cell-penetrating sequence allowed the peptides to access the cell interior and engage with their target(s), modulating signaling pathways. In Ras-driven cancer cell models, the peptides have an inhibitory effect on proliferation and show suppression of both invasion and motility. As such, they represent promising candidates for Rho-family small GTPase inhibitors and therapeutics targeting Ras-driven cancers. Our data add to the growing literature demonstrating that peptides are establishing their place in the biologics arm of drug discovery.


Subject(s)
Drug Discovery , Peptides, Cyclic/pharmacology , Signal Transduction/drug effects , cdc42 GTP-Binding Protein/antagonists & inhibitors , ras Proteins/metabolism , Binding Sites , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell-Penetrating Peptides , GTP Phosphohydrolases/antagonists & inhibitors , Humans , Molecular Structure , Neoplasm Invasiveness/prevention & control , Neoplasms/drug therapy , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , cdc42 GTP-Binding Protein/metabolism
14.
Small GTPases ; 11(6): 430-440, 2020 11.
Article in English | MEDLINE | ID: mdl-29969362

ABSTRACT

The Rho GTPase Cdc42 is highly conserved in structure and function. Mechanical or chemical cues in the microenvironment stimulate the localized activation of Cdc42 to rearrange the actin cytoskeleton and establish cell polarity. A role for Cdc42 in cell polarization was first discovered in the budding yeast Saccharomyces cerevisiae, and subsequently shown to also regulate directional motility in animal cells. Accordingly, in cancer Cdc42 promotes migration, invasion, and spread of tumor cells. Therefore, we targeted Cdc42 as a therapeutic strategy to treat metastatic breast cancer and designed the small molecule MBQ-167 as a potent inhibitor against Cdc42 and the homolog Rac. MBQ-167 inhibited cancer cell proliferation and migration in-vitro, and tumor growth and spread in-vivo in a mouse xenograft model of metastatic breast cancer. Since haploid budding yeast express a single Cdc42 gene, and do not express Rac, we used this well characterized model of polarization to define the contribution of Cdc42 inhibition to the effects of MBQ-167 in eukaryotic cells. Growth, budding pattern, and Cdc42 activity was determined in wildtype yeast or cells expressing a conditional knockdown of Cdc42 in response to vehicle or MBQ-167 treatment. As expected, growth and budding polarity were reduced by knocking-down Cdc42, with a parallel effect observed with MBQ-167. Cdc42 activity assays confirmed that MBQ-167 inhibits Cdc42 activation in yeast, and thus, bud polarity. Hence, we have validated MBQ-167 as a Cdc42 inhibitor in another biological context and present a method to screen Cdc42 inhibitors with potential as anti-metastatic cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Polarity/drug effects , Saccharomycetales/drug effects , cdc42 GTP-Binding Protein/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Molecular Structure , Saccharomycetales/cytology , Saccharomycetales/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
15.
J Cell Sci ; 132(24)2019 12 13.
Article in English | MEDLINE | ID: mdl-31727638

ABSTRACT

Constitutive secretion from the trans-Golgi-network (TGN) is facilitated by a concerted regulation of vesicle biogenesis and fission processes. The protein kinase D family (PKD) has been previously described to enhance vesicle fission by modifying the lipid environment. PKD also phosphorylates the actin regulatory protein cortactin at S298 to impair synergistic actin polymerization. We here report additional functions for PKD2 (also known as PRKD2) and cortactin in the regulation of actin polymerization during the fission of transport carriers from the TGN. Phosphorylation of cortactin at S298 impairs the interaction between WIP (also known as WIPF1) and cortactin. WIP stabilizes the autoinhibited conformation of N-WASP (also known as WASL). This leads to an inhibition of synergistic Arp2/3-complex-dependent actin polymerization at the TGN. PKD2 activity at the TGN is controlled by active CDC42-GTP which directly activates N-WASP, inhibits PKD2 and shifts the balance to non-S298-phosphorylated cortactin, which can in turn sequester WIP from N-WASP. Consequently, synergistic actin polymerization at the TGN and constitutive secretion are enhanced.


Subject(s)
Cortactin/metabolism , TRPP Cation Channels/metabolism , Actins , Animals , Blotting, Western , Fluorescence Resonance Energy Transfer , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , MCF-7 Cells , Mice , NIH 3T3 Cells , Polymerization , Pyrazoles/pharmacology , Sulfonamides/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/metabolism , trans-Golgi Network/genetics
16.
Biochem Biophys Res Commun ; 519(1): 134-140, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31477271

ABSTRACT

Tumor microenvironment is composed of biological, chemical and physical factors. Mechanical factors are more and more focused these years. Therefore, mimicking mechanical factors' contribution to cancer cell malignancy will greatly improve the advance in this field. Although the induced malignant behaviors are present under many stimuli such as growth or inflammatory factors, the cell key physical migration mechanisms are still missing. In this study, we identify that low shear stress significantly promotes the formation of needle-shaped membrane protrusions, which is called filopodia and important for the sense and interact of a cell with extracellular matrix in the tumor microenvironment. Under low shear stress, the migration is promoted while it is inhibited in the presence of ROCK inhibitor Y27632, which could abolish the F-actin network. Using cell imaging, we further unravel that key to these protrusions is Cell division cycle 42 (Cdc42) dependent. After Cdc42 activation, the filopodia is more and longer, acting as massagers to pass the information from a cell to the microenvironment for its malignant phenotype. In the Cdc42 inhibition, the filopodia is greatly reduced. Moreover, small GTPases Cdc42 rather than Rac1 and Rho directly controls the filopodia formation. Our work highlights that low shear stress and Cdc42 activation are sufficient to promote filopodia formation, it not only points out the novel structure for cancer progression but also provides the experimental physical basis for the efficient drug anti-cancer strategies.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Cytoskeleton/metabolism , Stress, Mechanical , cdc42 GTP-Binding Protein/metabolism , Amides/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Movement/drug effects , Cytoskeleton/drug effects , Female , Humans , Pyridines/pharmacology , Tumor Cells, Cultured , cdc42 GTP-Binding Protein/antagonists & inhibitors
17.
Biomolecules ; 9(9)2019 09 07.
Article in English | MEDLINE | ID: mdl-31500289

ABSTRACT

During development, growth cones are essential for axon pathfinding by sensing numerous guidance cues in their environment. Retinoic acid, the metabolite of vitamin A, is important for neurite outgrowth during vertebrate development, but may also play a role in axon guidance, though little is known of the cellular mechanisms involved. Our previous studies showed that retinoid-induced growth cone turning of invertebrate motorneurons requires local protein synthesis and calcium influx. However, the signalling pathways that link calcium influx to cytoskeletal dynamics involved in retinoid-mediated growth cone turning are not currently known. The Rho GTPases, Cdc42 and Rac, are known regulators of the growth cone cytoskeleton. Here, we demonstrated that inhibition of Cdc42 or Rac not only prevented growth cone turning toward retinoic acid but could also induce a switch in growth cone responsiveness to chemorepulsion or growth cone collapse. However, the effects of Cdc42 or Rac inhibition on growth cone responsiveness differed, depending on whether the turning was induced by the all-trans or 9-cis retinoid isomer. The effects also differed depending on whether the growth cones maintained communication with the cell body. These data strongly suggest that Cdc42 and Rac are downstream effectors of retinoic acid during growth cone guidance.


Subject(s)
Enzyme Inhibitors/pharmacology , Growth Cones/drug effects , Lymnaea , Tretinoin/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , Animals , Growth Cones/metabolism , Tretinoin/chemistry , cdc42 GTP-Binding Protein/antagonists & inhibitors
18.
Curr Pharm Biotechnol ; 20(9): 733-744, 2019.
Article in English | MEDLINE | ID: mdl-31258074

ABSTRACT

BACKGROUND: Recurrent pharyngotonsillitis due to Streptococcus pyogenes develops regardless of whether infecting strains are resistant or susceptible to first-line antimicrobials. Causation for recurrent infection is associated with the use of first-line antimicrobials that fail to penetrate deep tissue and host cell membranes, enabling intracellular S. pyogenes to survive throughout repeated rounds of antimicrobial therapy. OBJECTIVE: To determine whether simvastatin, a therapeutic approved for use in the treatment of hypercholesterolemia, and ML141, a first-in-class small molecule inhibitor with specificity for human CDC42, limit host cell invasion by S. pyogenes. METHODS: Assays to assess host cell invasion, bactericidal activity, host cell viability, actin depolymerization, and fibronectin binding were performed using the RAW 267.4 macrophage cell line and Human Umbilical Vein Endothelial Cells (HUVEC) infected with S. pyogenes (90-226) and treated with simvastatin, ML141, structural analogs of ML141, or vehicle control. RESULTS: Simvastatin and ML141 decreased intracellular infection by S. pyogenes in a dose-dependent manner. Inhibition by simvastatin persisted following 1 h washout whereas inhibition by ML141 was reversed. During S. pyogenes infection, actin stress fibers depolymerized in vehicle control treated cells, yet remained intact in simvastatin and in ML141 treated cells. Consistent with the previous characterization of ML141, simvastatin decreased host cell binding to fibronectin. Structural analogs of ML141, designated as the RSM series, decreased intracellular infection through non-cytotoxic, nonbactericidal mechanisms. CONCLUSION: Our findings demonstrate the potential of repurposing simvastatin and of developing CDC42-targeted therapeutics for eradicating intracellular S. pyogenes infection to break the cycle of recurrent infection through a host-directed approach.


Subject(s)
Anti-Bacterial Agents/pharmacology , Pyrazoles/pharmacology , Simvastatin/pharmacology , Streptococcal Infections/drug therapy , Streptococcus pyogenes/drug effects , Sulfonamides/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , Animals , Anti-Bacterial Agents/chemistry , Cells, Cultured , Fibronectins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/microbiology , Humans , Macrophages/metabolism , Macrophages/microbiology , Mice , Molecular Structure , Pyrazoles/chemistry , RAW 264.7 Cells , Simvastatin/chemistry , Sulfonamides/chemistry
19.
Cells ; 8(6)2019 06 10.
Article in English | MEDLINE | ID: mdl-31185627

ABSTRACT

Neuronal migration is essential for the orchestration of brain development and involves several contiguous steps: interkinetic nuclear movement (INM), multipolar-bipolar transition, locomotion, and translocation. Growing evidence suggests that Rho GTPases, including RhoA, Rac, Cdc42, and the atypical Rnd members, play critical roles in neuronal migration by regulating both actin and microtubule cytoskeletal components. This review focuses on the spatiotemporal-specific regulation of Rho GTPases as well as their regulators and effectors in distinct steps during the neuronal migration process. Their roles in bridging extracellular signals and cytoskeletal dynamics to provide optimal structural support to the migrating neurons will also be discussed.


Subject(s)
Neurons/metabolism , rho GTP-Binding Proteins/metabolism , Adherens Junctions/metabolism , Animals , Cell Movement , Ependymoglial Cells/chemistry , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Humans , Neurogenesis , Neurons/cytology , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors
20.
Sci Rep ; 9(1): 4418, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872706

ABSTRACT

The combined phenotype of thrombocytopenia accompanied by intellectual disability in patients with a de novo heterozygous mutation, i.e., p.Tyr64Cys in CDC42, signifies a clinically recognizable novel syndrome that has been eponymized as "Takenouchi-Kosaki syndrome" (OMIM #616737). In the present study, a detailed phenotypic analysis performed for a total of five patients with Takenouchi-Kosaki syndrome revealed that intellectual disability, macrothrombocytopenia, camptodactyly, structural brain abnormalities with sensorineural deafness, hypothyroidism, and frequent infections comprise the cardinal features of this condition. A morphologic analysis of platelets derived from three affected individuals was performed using electron microscopy. The platelets of the three patients were large and spherical in shape. Furthermore, platelet α-granules were decreased, while vacuoles were increased. We further performed a functional analysis of p.Tyr64Cys in CDC42 through CRISPR/Cas9-mediated gene editing in a Caenorhabditis elegans model. This functional analysis suggested that the mutant allele has hypomorphic effects. Takenouchi-Kosaki syndrome is clinically recognizable by the combined phenotype of intellectual disability, macrothrombocytopenia, camptodactyly, structural brain abnormalities with sensorineural deafness, hypothyroidism, and frequent infections as well as the identification of a heterozygous de novo mutation in CDC42, i.e., p.Tyr64Cys.


Subject(s)
Blood Platelets/pathology , Caenorhabditis elegans/physiology , Microscopy, Electron/methods , Mutation , Takayasu Arteritis/pathology , cdc42 GTP-Binding Protein/metabolism , Adolescent , Adult , Animals , Apoptosis , Caenorhabditis elegans/ultrastructure , Child , Child, Preschool , Female , Gene Editing , Humans , Male , Phenotype , Young Adult , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...