Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 173
Filter
1.
Protein Cell ; 13(11): 825-841, 2022 11.
Article in English | MEDLINE | ID: mdl-35217990

ABSTRACT

Metformin is currently a strong candidate anti-tumor agent in multiple cancers. However, its anti-tumor effectiveness varies among different cancers or subpopulations, potentially due to tumor heterogeneity. It thus remains unclear which hepatocellular carcinoma (HCC) patient subpopulation(s) can benefit from metformin treatment. Here, through a genome-wide CRISPR-Cas9-based knockout screen, we find that DOCK1 levels determine the anti-tumor effects of metformin and that DOCK1 is a synthetic lethal target of metformin in HCC. Mechanistically, metformin promotes DOCK1 phosphorylation, which activates RAC1 to facilitate cell survival, leading to metformin resistance. The DOCK1-selective inhibitor, TBOPP, potentiates anti-tumor activity by metformin in vitro in liver cancer cell lines and patient-derived HCC organoids, and in vivo in xenografted liver cancer cells and immunocompetent mouse liver cancer models. Notably, metformin improves overall survival of HCC patients with low DOCK1 levels but not among patients with high DOCK1 expression. This study shows that metformin effectiveness depends on DOCK1 levels and that combining metformin with DOCK1 inhibition may provide a promising personalized therapeutic strategy for metformin-resistant HCC patients.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Metformin , rac GTP-Binding Proteins , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Genome , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mice , Phosphorylation , Synthetic Lethal Mutations , Transcription Factors/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
2.
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
3.
BMC Cancer ; 21(1): 652, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34074257

ABSTRACT

BACKGROUND: Even though targeted therapies are available for cancers expressing oncogenic epidermal growth receptor (EGFR) and (or) human EGFR2 (HER2), acquired or intrinsic resistance often confounds therapy success. Common mechanisms of therapy resistance involve activating receptor point mutations and (or) upregulation of signaling downstream of EGFR/HER2 to Akt and (or) mitogen activated protein kinase (MAPK) pathways. However, additional pathways of resistance may exist thus, confounding successful therapy. METHODS: To determine novel mechanisms of EGFR/HER2 therapy resistance in breast cancer, gefitinib or lapatinib resistant variants were created from SKBR3 breast cancer cells. Syngenic therapy sensitive and resistant SKBR3 variants were characterized for mechanisms of resistance by mammosphere assays, viability assays, and western blotting for total and phospho proteins. RESULTS: Gefitinib and lapatinib treatments reduced mammosphere formation in the sensitive cells, but not in the therapy resistant variants, indicating enhanced mesenchymal and cancer stem cell-like characteristics in therapy resistant cells. The therapy resistant variants did not show significant changes in known therapy resistant pathways of AKT and MAPK activities downstream of EGFR/HER2. However, these cells exhibited elevated expression and activation of the small GTPase Rac, which is a pivotal intermediate of GFR signaling in EMT and metastasis. Therefore, the potential of the Rac inhibitors EHop-016 and MBQ-167 to overcome therapy resistance was tested, and found to inhibit viability and induce apoptosis of therapy resistant cells. CONCLUSIONS: Rac inhibition may represent a viable strategy for treatment of EGFR/HER2 targeted therapy resistant breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , rac GTP-Binding Proteins/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carbazoles/pharmacology , Carbazoles/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Gain of Function Mutation , Gefitinib/pharmacology , Gefitinib/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Point Mutation , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Spheroids, Cellular , Up-Regulation
4.
Sci Rep ; 11(1): 5009, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33658535

ABSTRACT

This study aimed to explore the role of LINC00665, miR-4458 and DOCK1 and their interactions in the development of acute myeloid leukemia (AML). The relative expression of LINC00665, miR-4458 and DOCK1 in AML samples was measured using qRT-PCR, and the protein level of DOCK1 in AML cell lines was examined using western blot. CCK8, BrdU, transwell, cell adhesion, and caspase-3 activity assays were carried out to evaluate the viability, proliferation, migration, adhesion, and apoptosis of AML cells, respectively. Luciferase reporter, RIP, and RNA pull-down assays were also performed to confirm the target relationship among LINC00665, miR-4458 and DOCK1. Findings revealed that LINC00665 and DOCK1 were aberrantly overexpressed in AML tissues and that the expression of miR-4458 was low in AML tissues. Silencing LINC00665 or DOCK1 presented significant restriction to the proliferation, migration and adhesion of AML cells. Apart from that, it was found that inhibiting miR-4458 could enhance the proliferation, migration and adhesion of AML cells but suppress the apoptosis of AML cells. Experimental results also indicated that LINC00665 exerted its positive function on AML cells by sponging miR-4458 and that miR-4458 influenced the progression of AML cells by targeting DOCK1 directly. Overall, this finding not only provided a novel molecular pathway for the diagnosis and treatment of AML but also showed that LINC00665 could enhance the progression of AML by regulating the miR-4458/DOCK1 pathway.


Subject(s)
Gene Regulatory Networks , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , rac GTP-Binding Proteins/genetics , Adult , Aged , Apoptosis/genetics , Base Pairing , Case-Control Studies , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HL-60 Cells , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Middle Aged , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
5.
Biochem Biophys Res Commun ; 529(4): 1173-1179, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32819582

ABSTRACT

PURPOSE: Among the members of the DOCK family, DOCK1-5 function as guanine-nucleotide exchange factors for small GTPase Rac1, which regulates the actin cytoskeleton. It has been reported that in model organisms the Dock-Rac axis is required for myoblast fusion. We examined the role of DOCK1-5 in trophoblast fusion herein. METHODS: We used a quantitative polymerase chain reaction (qPCR) to examine the mRNA expressions of DOCK1-5 and differentiation-related genes, i.e., fusogenic genes, in human trophoblastic cell lines, BeWo and JEG-3. We treated BeWo cells with TBOPP and C21 to inhibit DOCK1 and DOCK5. Cell dynamics and cell fusion were assessed by live imaging and immunostaining. The signaling pathways induced by DOCK1/5 inhibition were examined by western blotting. RESULTS: DOCK1 and DOCK5 were expressed in BeWo cells. The inhibition of DOCK1 or DOCK5 did not prevent the cell fusion induced by forskolin (a common reagent for cell fusion); it induced cell fusion. DOCK1 inhibition induced cell death, as did forskolin. DOCK1 and DOCK5 inhibition for 24 and 48 h increased the expression of the genes ASCT2 and SYNCYTIN2, which code responsive proteins of trophoblast cell fusion, respectively. CONCLUSION: DOCK1 and DOCK5 inhibition participates in BeWo cell fusion, probably via pathways independent from forskolin-mediated pathways.


Subject(s)
Trophoblasts/cytology , Trophoblasts/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , Cell Aggregation/drug effects , Cell Death/drug effects , Cell Fusion , Cell Line, Tumor , Colforsin/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trophoblasts/drug effects , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
6.
Prostate ; 80(13): 1045-1057, 2020 09.
Article in English | MEDLINE | ID: mdl-32687658

ABSTRACT

BACKGROUND: There is a need to develop novel therapies which could be beneficial to patients with prostate cancer (CaP) including those who are predisposed to poor outcome, such as African-Americans. This study investigates the role of ROBO1-pathway in predicting outcome and race-based disparity in patients with CaP. METHODS AND RESULTS: Aided by RNA sequencing-based DECIPHER-testing and immunohistochemical (IHC) analysis of tumors we show that ROBO1 is lost during the progressive stages of CaP, a prevalent feature in African-Americans. We show that the loss of ROBO1 predicts high-risk of recurrence, metastasis and poor outcome of androgen-deprivation therapy in radical prostatectomy-treated patients. These data identified an aggressive ROBO1deficient /DOCK1+ve sub-class of CaP. Combined genetic and IHC data showed that ROBO1 loss is accompanied by DOCK1/Rac1 elevation in grade-III/IV primary-tumors and Mets. We observed that the hypermethylation of ROBO1-promoter contributes to loss of expression that is highly prevalent in African-Americans. Because of limitations in restoring ROBO1 function, we asked if targeting the DOCK1 could be an ideal strategy to inhibit progression or treat ROBO1deficient metastatic-CaP. We tested the pharmacological efficacy of CPYPP, a selective inhibitor of DOCK1 under in vitro and in vivo conditions. Using ROBO1-ve and ROBO1+ve CaP models, we determined the median effective concentration of CPYPP for growth. DOCK1-inhibitor treatment significantly decreased the (a) Rac1-GTP/ß-catenin activity, (b) transmigration of ROBO1deficient cells across endothelial lining, and (c) metastatic spread of ROBO1deficient cells through the vasculature of transgenicfl Zebrafish model. CONCLUSION: We suggest that ROBO1 status forms as predictive biomarker of outcome in high-risk populations such as African-Americans and DOCK1-targeting therapy has a clinical potential for treating metastatic-CaP.


Subject(s)
Black or African American/genetics , Nerve Tissue Proteins/genetics , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/genetics , Receptors, Immunologic/genetics , rac GTP-Binding Proteins/genetics , Animals , Cell Line, Tumor , DNA Methylation , Health Status Disparities , Humans , Immunohistochemistry , Male , Neoplasm Metastasis , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Promoter Regions, Genetic , Prostatectomy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/deficiency , White People/genetics , Zebrafish , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , Roundabout Proteins
7.
Mol Med Rep ; 22(2): 1187-1194, 2020 08.
Article in English | MEDLINE | ID: mdl-32626999

ABSTRACT

The treatment of renal cell carcinoma (RCC) with chemotherapy remains a challenge; therefore, improving the knowledge of the molecular mechanisms underlying RCC chemoresistance and developing novel therapeutic strategies is important. Dedicator of cytokinesis 1 (DOCK1), the first member of the DOCK family to be discovered, displays various roles during tumorigenesis; however, its role during RCC progression is not completely understood. Therefore, the present study aimed to clarify the function of DOCK1 and 1­[2­(3'­(trifluoromethyl)­(1,1'­biphenyl)­4­yl)­2­oxoethyl]­5­pyrrolidinylsulfonyl­2 (1H)­pyridone (TBOPP), a DOCK1­sensitive inhibitor, during RCC development and chemoresistance. The results of CCK­8 and EdU assay indicated that TBOPP decreased RCC cell viability and proliferation compared with the control group, and sensitized RCC cells to cisplatin. Moreover, RCC cells with high DOCK1 expression levels displayed increased resistance to cisplatin, whereas DOCK1 knockdown enhanced the lethal effects of cisplatin on RCC cells. Furthermore, the results determined by western blotting, CCK­8 and cell apoptosis assay indicated that TBOPP effectively reduced DOCK1 expression levels compared with the control group, and the TBOPP­mediated cisplatin sensitizing effect was mediated by DOCK1 inhibition. The present study suggests that DOCK1 plays a vital role in RCC cell chemoresistance to cisplatin; therefore, TBOPP may serve as a novel therapeutic agent for RCC chemoresistance.


Subject(s)
Carcinogenesis , Carcinoma, Renal Cell , Cisplatin , Drug Resistance, Neoplasm/drug effects , Kidney Neoplasms , Pyridones/pharmacology , rac GTP-Binding Proteins , Apoptosis/drug effects , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/physiology
8.
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
9.
Cancer Res ; 80(14): 3009-3022, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32366477

ABSTRACT

HACE1 is an E3 ubiquitin ligase with important roles in tumor biology and tissue homeostasis. Loss or mutation of HACE1 has been associated with the occurrence of a variety of neoplasms, but the underlying mechanisms have not been defined yet. Here, we report that HACE1 is frequently mutated in human lung cancer. In mice, loss of Hace1 led to enhanced progression of KRasG12D -driven lung tumors. Additional ablation of the oncogenic GTPase Rac1 partially reduced progression of Hace1-/- lung tumors. RAC2, a novel ubiquitylation target of HACE1, could compensate for the absence of its homolog RAC1 in Hace1-deficient, but not in HACE1-sufficient tumors. Accordingly, ablation of both Rac1 and Rac2 fully averted the increased progression of KRasG12D -driven lung tumors in Hace1-/- mice. In patients with lung cancer, increased expression of HACE1 correlated with reduced levels of RAC1 and RAC2 and prolonged survival, whereas elevated expression of RAC1 and RAC2 was associated with poor prognosis. This work defines HACE1 as a crucial regulator of the oncogenic activity of RAC-family GTPases in lung cancer development. SIGNIFICANCE: These findings reveal that mutation of the tumor suppressor HACE1 disrupts its role as a regulator of the oncogenic activity of RAC-family GTPases in human and murine lung cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/14/3009/F1.large.jpg.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/prevention & control , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/pathology , Cell Proliferation , Humans , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , RAC2 GTP-Binding Protein
10.
Development ; 147(12)2020 06 24.
Article in English | MEDLINE | ID: mdl-32467241

ABSTRACT

Tendons and ligaments are crucial components of the musculoskeletal system, yet the pathways specifying these fates remain poorly defined. Through a screen of known bioactive chemicals in zebrafish, we identified a new pathway regulating tendon cell induction. We established that statin, through inhibition of the mevalonate pathway, causes an expansion of the tendon progenitor population. Co-expression and live imaging studies indicate that the expansion does not involve an increase in cell proliferation, but rather results from re-specification of cells from the neural crest-derived sox9a+/sox10+ skeletal lineage. The effect on tendon cell expansion is specific to the geranylgeranylation branch of the mevalonate pathway and is mediated by inhibition of Rac activity. This work establishes a novel role for the mevalonate pathway and Rac activity in regulating specification of the tendon lineage.


Subject(s)
Mevalonic Acid/metabolism , Tendons/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Animals, Genetically Modified/metabolism , Atorvastatin/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Morpholinos/metabolism , Neural Crest/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tendons/cytology , Tendons/pathology , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
11.
Cell Physiol Biochem ; 53(6): 933-947, 2019.
Article in English | MEDLINE | ID: mdl-31805226

ABSTRACT

BACKGROUND/AIMS: We showed that patho-physiological concentrations of either 7-keto-cholesterol (7-KC), or cholestane-3beta, 5alpha, 6beta-triol (TRIOL) caused the eryptotic death of human red blood cells (RBC), strictly dependent on the early production of reactive oxygen species (ROS). The goal of the current study was to assess the contribution of the erythrocyte ROS-generating enzymes, NADPH oxidase (RBC-NOX), nitric oxide synthase (RBC-NOS) and xanthine oxido-reductase (XOR) to the oxysterol-dependent eryptosis and pertinent activation pathways. METHODS: Phosphatidylserine exposure at the cell surface was estimated from annexin-V-binding, reactive oxygen/nitrogen species (RONS) and nitric oxide formation from 2',7'-dichloro-dihydrofluorescein (DCF-DA) and 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM DA) -dependent fluorescence, respectively; Akt1, phospho-NOS3 Ser1177, and PKCζ from Western blot analysis. The activity of individual 7-KC (7 µM) and TRIOL (2, µM) on ROS-generating enzymes and relevant activation pathways was assayed in the presence of Diphenylene iodonium chloride (DPI), N-nitro-L-arginine methyl ester (L-NAME), allopurinol, NSC23766 and LY294002, inhibitors in this order of RBC-NOX, RBC-NOS, XOR and upstream regulatory proteins Rac GTPase and phosphoinositide3 Kinase (PI3K); hemoglobin oxidation from spectrophotometric analysis. RESULTS: RBC-NOX was the target of 7-KC, through a signaling including Rac GTPase and PKCζ, whereas TRIOL caused activation of RBC-NOS according to the pathway PI3K/Akt, with the concurrent activity of a Rac-GTPase. In concomitance with the TRIOL-induced .NO production, formation of methemoglobin with global loss of heme were observed, ascribable to nitrosative stress. XOR, activated after modification of the redox environment by either RBC-NOX or RBC-NOS activity, concurred to the overall oxidative/nitrosative stress by either oxysterols. When 7-KC and TRIOL were combined, they acted independently and their effect on ROS/RONS production and PS exposure appeared the result of the effects of the oxysterols on RBC-NOX and RBC-NOS. CONCLUSION: Eryptosis of human RBCs may be caused by either 7-KC or TRIOL by oxidative/nitrosative stress through distinct signaling cascades activating RBC-NOX and RBC-NOS, respectively, with the complementary activity of XOR; when combined, the oxysterols act independently and both concur to the final eryptotic effect.


Subject(s)
Cholestanols/pharmacology , Eryptosis/drug effects , Ketocholesterols/pharmacology , NADPH Oxidases/metabolism , Nitric Oxide Synthase/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Hemoglobins/chemistry , Humans , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
12.
Curr Biol ; 28(22): 3674-3684.e6, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30393033

ABSTRACT

Cell migration often involves the formation of sheet-like lamellipodia generated by branched actin filaments. The branches are initiated when Arp2/3 complex [1] is activated by WAVE regulatory complex (WRC) downstream of small GTPases of the Rac family [2]. Recent structural studies defined two independent Rac binding sites on WRC within the Sra-1/PIR121 subunit of the pentameric WRC [3, 4], but the functions of these sites in vivo have remained unknown. Here we dissect the mechanism of WRC activation and the in vivo relevance of distinct Rac binding sites on Sra-1, using CRISPR/Cas9-mediated gene disruption of Sra-1 and its paralog PIR121 in murine B16-F1 cells combined with Sra-1 mutant rescue. We show that the A site, positioned adjacent to the binding region of WAVE-WCA mediating actin and Arp2/3 complex binding, is the main site for allosteric activation of WRC. In contrast, the D site toward the C terminus is dispensable for WRC activation but required for optimal lamellipodium morphology and function. These results were confirmed in evolutionarily distant Dictyostelium cells. Moreover, the phenotype seen in D site mutants was recapitulated in Rac1 E31 and F37 mutants; we conclude these residues are important for Rac-D site interaction. Finally, constitutively activated WRC was able to induce lamellipodia even after both Rac interaction sites were lost, showing that Rac interaction is not essential for membrane recruitment. Our data establish that physical interaction with Rac is required for WRC activation, in particular through the A site, but is not mandatory for WRC accumulation in the lamellipodium.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dictyostelium/metabolism , Multiprotein Complexes/metabolism , Pseudopodia/physiology , rac1 GTP-Binding Protein/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Movement , Dictyostelium/cytology , Dictyostelium/genetics , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Neuropeptides/antagonists & inhibitors , Neuropeptides/metabolism , Protein Conformation , Tumor Cells, Cultured , Wiskott-Aldrich Syndrome Protein Family/chemistry , Wiskott-Aldrich Syndrome Protein Family/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors , RAC2 GTP-Binding Protein
13.
Cell Physiol Biochem ; 49(4): 1523-1538, 2018.
Article in English | MEDLINE | ID: mdl-30212812

ABSTRACT

BACKGROUND/AIMS: Thrombin induces the activation of human platelets through protease-activated receptor (PAR) 1 and PAR4, and Rac, a member of the Rho family of small GTPases, is implicated in PAR activation. We previously reported that phosphorylated-heat shock protein 27 (HSP27) is released from the thrombin receptor-activating peptide (TRAP)-stimulated platelets of diabetic patients. In the present study, we investigated the role of Rac in the TRAP-elicited release of phosphorylated-HSP27 from human platelets. METHODS: Platelet aggregation was measured using an aggregometer with laser scattering. Protein phosphorylation was analyzed by Western blotting. The levels of phosphorylated-HSP27 and platelet-derived growth factor-AB (PDGF-AB) were measured by enzyme-linked immunosorbent assays. RESULTS: NSC23766, an inhibitor of Rac-guanine nucleotide exchange factor interaction, suppressed the TRAP-elicited release of phosphorylated-HSP27 as well as platelet aggregation. The TRAP-induced phosphorylation of HSP27, p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) was attenuated by NSC23766. SB203580, a p38 MAPK inhibitor, but not SP600125, a JNK inhibitor, suppressed the release of phosphorylated-HSP27 in addition to HSP27 phosphorylation. On the other hand, both SB203580 and SP600125 reduced the TRAP-stimulated secretion of PDGF-AB. CONCLUSION: Our results strongly suggest that Rac acts as a positive regulator of the PAR-elicited release of phosphorylated-HSP27 from human platelets via p38 MAPK but not JNK.


Subject(s)
Blood Platelets/drug effects , HSP27 Heat-Shock Proteins/metabolism , Peptide Fragments/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , rac GTP-Binding Proteins/metabolism , Aminoquinolines/pharmacology , Blood Platelets/cytology , Blood Platelets/metabolism , Cells, Cultured , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Platelet-Derived Growth Factor/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , rac GTP-Binding Proteins/antagonists & inhibitors
14.
J Biol Chem ; 293(40): 15397-15418, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30108175

ABSTRACT

The MET proto-oncogene-encoded receptor tyrosine kinase (MET) and AXL receptor tyrosine kinase (AXL) are independently operating receptor tyrosine kinases (RTKs) that are functionally associated with aggressive and invasive cancer cell growth. However, how MET and AXL regulate the migratory properties of cancer cells remains largely unclear. We report here that the addition of hepatocyte growth factor (HGF), the natural ligand of MET, to serum-starved human glioblastoma cells induces the rapid activation of both MET and AXL and formation of highly polarized MET-AXL clusters on the plasma membrane. HGF also promoted the formation of the MET and AXL protein complexes and phosphorylation of AXL, independent of AXL's ligand, growth arrest-specific 6 (GAS6). The HGF-induced MET-AXL complex stimulated rapid and dynamic cytoskeleton reorganization by activating the small GTPase RAC1, a process requiring both MET and AXL kinase activities. We further found that HGF also promotes the recruitment of ELMO2 and DOCK180, a bipartite guanine nucleotide exchange factor for RAC1, to the MET-AXL complex and thereby stimulates the RAC1-dependent cytoskeleton reorganization. We also demonstrated that the MET-AXL-ELMO2-DOCK180 complex is critical for HGF-induced cell migration and invasion in glioblastoma or other cancer cells. Our findings uncover a critical HGF-dependent signaling pathway that involves the assembly of a large protein complex consisting of MET, AXL, ELMO2, and DOCK180 on the plasma membrane, leading to RAC1-dependent cell migration and invasion in various cancer cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/pharmacology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Neoplasm Invasiveness , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Protein Transport/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/agonists , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism , Axl Receptor Tyrosine Kinase
15.
Cancer Res ; 78(12): 3101-3111, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29858187

ABSTRACT

Rac and Cdc42 are small GTPases that have been linked to multiple human cancers and are implicated in epithelial to mesenchymal transition, cell-cycle progression, migration/invasion, tumor growth, angiogenesis, and oncogenic transformation. With the exception of the P29S driver mutation in melanoma, Rac and Cdc42 are not generally mutated in cancer, but are overexpressed (gene amplification and mRNA upregulation) or hyperactivated. Rac and Cdc42 are hyperactivated via signaling through oncogenic cell surface receptors, such as growth factor receptors, which converge on the guanine nucleotide exchange factors that regulate their GDP/GTP exchange. Hence, targeting Rac and Cdc42 represents a promising strategy for precise cancer therapy, as well as for inhibition of bypass signaling that promotes resistance to cell surface receptor-targeted therapies. Therefore, an understanding of the regulatory mechanisms of these pivotal signaling intermediates is key for the development of effective inhibitors. In this review, we focus on the role of Rac and Cdc42 in cancer and summarize the regulatory mechanisms, inhibitory efficacy, and the anticancer potential of Rac- and Cdc42-targeting agents. Cancer Res; 78(12); 3101-11. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , cdc42 GTP-Binding Protein/antagonists & inhibitors , rac GTP-Binding Proteins/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/pathology , Protein Binding/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism
16.
Cytoskeleton (Hoboken) ; 75(7): 307-317, 2018 07.
Article in English | MEDLINE | ID: mdl-29752871

ABSTRACT

The migration and invasion of cells through tissues in the body is facilitated by a dynamic actin cytoskeleton. The actin-associating protein, tropomyosin Tpm3.1 has emerged to play important roles in cell migration and invasion. To date, investigations have focused on single cell migration and invasion where Tpm3.1 expression is inversely associated with Rac GTPase-mediated cell invasion. While single cell and collective cell invasion have many features in common, collective invasion is additionally impacted by cell-cell adhesion, and the role of Tpm3.1 in collective invasion has not been established. In the present study we have modelled multicellular invasion using neuroblastoma spheroids embedded in 3D collagen and analysed the function of Tpm3.1 using recently established compounds that target the Tpm3.1 C-terminus. The major findings from our study reveal that combined Rac inhibition and Tpm3.1 targeting result in greater inhibition of multicellular invasion than either treatment alone. Together, the data suggest that Tpm3.1 disruption sensitises neuroblastoma cells to inhibition of Rac-mediated multicellular invasion.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Enzyme Inhibitors/pharmacology , Neuroblastoma/drug therapy , Tropomyosin/antagonists & inhibitors , rac GTP-Binding Proteins/antagonists & inhibitors , Actins/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/administration & dosage , Humans , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Invasiveness , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Tropomyosin/metabolism , rac GTP-Binding Proteins/metabolism
17.
Biochem Biophys Res Commun ; 497(1): 298-304, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29432733

ABSTRACT

Rac1 is a member of the Rho family of small GTPases that regulates cytoskeletal reorganization, membrane polarization, cell migration and proliferation. Recently, a self-activating mutation of Rac1, Rac1P29S, has been identified as a recurrent somatic mutation frequently found in sun-exposed melanomas, which possesses increased inherent GDP/GTP exchange activity and cell transforming ability. However, the role of cellular Rac1-interacting proteins in the transforming potential of Rac1P29S remains unclear. We found that the catalytic domain of DOCK1, a Rac-specific guanine nucleotide exchange factor (GEF) implicated in malignancy of a variety of cancers, can greatly accelerate the GDP/GTP exchange of Rac1P29S. Enforced expression of Rac1P29S induced matrix invasion and macropinocytosis in wild-type (WT) mouse embryonic fibroblasts (MEFs), but not in DOCK1-deficient MEFs. Consistently, a selective inhibitor of DOCK1 that blocks its GEF function suppressed the invasion and macropinocytosis in WT MEFs expressing Rac1P29S. Human melanoma IGR-1 and breast cancer MDA-MB-157 cells harbor Rac1P29S mutation and express DOCK1 endogenously. Genetic inactivation and pharmacological inhibition of DOCK1 suppressed their invasion and macropinocytosis. Taken together, these results indicate that DOCK1 is a critical regulator of the malignant phenotypes induced by Rac1P29S, and suggest that targeting DOCK1 might be an effective approach to treat cancers associated with Rac1P29S mutation.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Pinocytosis/genetics , rac GTP-Binding Proteins/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , Cell Line, Tumor , Humans , Mutation/genetics , Neoplasm Invasiveness
18.
Nat Immunol ; 18(12): 1353-1360, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058702

ABSTRACT

The polarization of leukocytes toward chemoattractants is essential for the directed migration (chemotaxis) of leukocytes. How leukocytes acquire polarity after encountering chemical gradients is not well understood. We found here that leukocyte polarity was generated by TIPE2 (TNFAIP8L2), a transfer protein for phosphoinositide second messengers. TIPE2 functioned as a local enhancer of phosphoinositide-dependent signaling and cytoskeleton remodeling, which promoted leading-edge formation. Conversely, TIPE2 acted as an inhibitor of the GTPase Rac, which promoted trailing-edge polarization. Consequently, TIPE2-deficient leukocytes were defective in polarization and chemotaxis, and TIPE2-deficient mice were resistant to leukocyte-mediated neural inflammation. Thus, the leukocyte polarizer is a dual-role phosphoinositide-transfer protein and represents a potential therapeutic target for the treatment of inflammatory diseases.


Subject(s)
Chemotaxis, Leukocyte/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Intracellular Signaling Peptides and Proteins/genetics , T-Lymphocytes/immunology , Animals , Cell Polarity/genetics , Chemotaxis, Leukocyte/physiology , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositols/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , rac GTP-Binding Proteins/antagonists & inhibitors
19.
PLoS One ; 12(9): e0184595, 2017.
Article in English | MEDLINE | ID: mdl-28886159

ABSTRACT

Homing of peripheral stem cells is regulated by one of the most representative homing factors, stromal cell-derived factor 1 alpha (SDF-1α), which specifically binds to the plasma membrane receptor CXCR4 of mesenchymal stem cells (MSCs) in order to initiate the signaling pathways that lead to directional migration and homing of stem cells. This complex homing process and directional migration of stem cells have been mimicked on a microfluidic device that is capable of generating a chemokine gradient within the collagen matrix and embedding endothelial cell (EC) monolayers to mimic blood vessels. On the microfluidic device, stem cells showed directional migration toward the higher concentration of SDF-1α, whereas treatment with the CXCR4 antagonist AMD3100 caused loss of directionality of stem cells. Furthermore, inhibition of stem cell's main migratory signaling pathways, Rho-ROCK and Rac pathways, caused blockage of actomyosin and lamellipodia formation, decreasing the migration distance but maintaining directionality. Stem cell homing regulated by SDF-1α caused directional migration of stem cells, while the migratory ability was affected by the activation of migration-related signaling pathways.


Subject(s)
Chemokine CXCL12/chemistry , Lab-On-A-Chip Devices , Mesenchymal Stem Cells/cytology , Amides/pharmacology , Aminoquinolines/pharmacology , Benzylamines , Cell Movement/drug effects , Cyclams , Heterocyclic Compounds/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Mesenchymal Stem Cells/drug effects , Microscopy, Confocal , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism
20.
Mol Biol Cell ; 27(17): 2653-61, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27413008

ABSTRACT

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of skeletal muscle origin in children and adolescents. Among RMS subtypes, alveolar rhabdomyosarcoma (ARMS), which is characterized by the presence of the PAX3-FOXO1A or PAX7-FOXO1A chimeric oncogenic transcription factor, is associated with poor prognosis and a strong risk of metastasis compared with the embryonal subtype (ERMS). To identify molecular pathways involved in ARMS aggressiveness, we first characterized the migratory behavior of cell lines derived from ARMS and ERMS biopsies using a three-dimensional spheroid cell invasion assay. ARMS cells were more invasive than ERMS cells and adopted an ellipsoidal morphology to efficiently invade the extracellular matrix. Moreover, the invasive potential of ARMS cells depended on ROCK activity, which is regulated by the GTPase RhoE. Specifically, RhoE expression was low in ARMS biopsies, and its overexpression in ARMS cells reduced their invasion potential. Conversely, ARHGAP25, a GTPase-activating protein for Rac, was up-regulated in ARMS biopsies. Moreover, we found that ARHGAP25 inhibits Rac activity downstream of ROCKII and is required for ARMS cell invasion. Our results indicate that the RhoE/ROCK/ARHGAP25 signaling pathway promotes ARMS invasive potential and identify these proteins as potential therapeutic targets for ARMS treatment.


Subject(s)
GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , Cell Differentiation , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Muscle, Skeletal/metabolism , Oncogene Proteins, Fusion/metabolism , Rhabdomyosarcoma , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/metabolism , Signal Transduction/genetics , rac GTP-Binding Proteins/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL