Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 16(11): 1142-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26414765

ABSTRACT

Mitochondria need to be juxtaposed to phagosomes for the synergistic production of ample reactive oxygen species (ROS) in phagocytes to kill pathogens. However, how phagosomes transmit signals to recruit mitochondria has remained unclear. Here we found that the kinases Mst1 and Mst2 functioned to control ROS production by regulating mitochondrial trafficking and mitochondrion-phagosome juxtaposition. Mst1 and Mst2 activated the GTPase Rac to promote Toll-like receptor (TLR)-triggered assembly of the TRAF6-ECSIT complex that is required for the recruitment of mitochondria to phagosomes. Inactive forms of Rac, including the human Rac2(D57N) mutant, disrupted the TRAF6-ECSIT complex by sequestering TRAF6 and substantially diminished ROS production and enhanced susceptibility to bacterial infection. Our findings demonstrate that the TLR-Mst1-Mst2-Rac signaling axis is critical for effective phagosome-mitochondrion function and bactericidal activity.


Subject(s)
Phagocytes/immunology , Phagocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Infections/etiology , Bacterial Infections/immunology , Bacterial Infections/metabolism , Blood Bactericidal Activity/immunology , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/microbiology , Phagocytes/microbiology , Phagosomes/immunology , Phagosomes/metabolism , Phagosomes/microbiology , Protein Kinase C-alpha/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Sepsis/etiology , Sepsis/immunology , Sepsis/metabolism , Serine-Threonine Kinase 3 , Signal Transduction , TNF Receptor-Associated Factor 6 , Toll-Like Receptors/metabolism , Ubiquitination , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
2.
Mol Carcinog ; 63(5): 977-990, 2024 May.
Article in English | MEDLINE | ID: mdl-38376344

ABSTRACT

Nickel pollution is a recognized factor contributing to lung cancer. Understanding the molecular mechanisms of its carcinogenic effects is crucial for lung cancer prevention and treatment. Our previous research identified the downregulation of a long noncoding RNA, maternally expressed gene 3 (MEG3), as a key factor in transforming human bronchial epithelial cells (HBECs) into malignant cells following nickel exposure. In our study, we found that deletion of MEG3 also reduced the expression of RhoGDIß. Notably, artificially increasing RhoGDIß levels counteracted the malignant transformation caused by MEG3 deletion in HBECs. This indicates that the reduction in RhoGDIß contributes to the transformation of HBECs due to MEG3 deletion. Further exploration revealed that MEG3 downregulation led to enhanced c-Jun activity, which in turn promoted miR-200c transcription. High levels of miR-200c subsequently increased the translation of AUF1 protein, stabilizing SOX2 messenger RNA (mRNA). This stabilization affected the regulation of miR-137, SP-1 protein translation, and the suppression of RhoGDIß mRNA transcription and protein expression, leading to cell transformation. Our study underscores the co-regulation of RhoGDIß expression by long noncoding RNA MEG3, multiple microRNAs (miR-200c and miR-137), and RNA-regulated transcription factors (c-Jun, SOX2, and SP1). This intricate network of molecular events sheds light on the nature of lung tumorigenesis. These novel findings pave the way for developing targeted strategies for the prevention and treatment of human lung cancer based on the MEG3/RhoGDIß pathway.


Subject(s)
Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Down-Regulation , Epithelial Cells/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Nickel , rho Guanine Nucleotide Dissociation Inhibitor beta/antagonists & inhibitors , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger , SOXB1 Transcription Factors/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism
3.
BMC Biol ; 21(1): 181, 2023 08 28.
Article in English | MEDLINE | ID: mdl-37635218

ABSTRACT

BACKGROUND: Functional role of Rho GDP-dissociation inhibitor beta (RhoGDIß) in tumor biology appears to be contradictory across various studies. Thus, the exploration of the molecular mechanisms underlying the differential functions of this protein in urinary bladder carcinogenesis is highly significant in the field. Here, RhoGDIß expression patterns, biological functions, and mechanisms leading to transformation and progression of human urothelial cells (UROtsa cells) were evaluated following varying lengths of exposure to the bladder carcinogen N-butyl-N-(4-hydmoxybutyl) nitrosamine (BBN). RESULTS: It was seen that compared to expression in vehicle-treated control cells, RhoGDIß protein expression was downregulated after 2-month of BBN exposure, but upregulated after 6-month of exposure. Assessments of cell function showed that RhoGDIß inhibited UROtsa cell growth in cells with BBN for 2-month exposure, whereas it promoted the invasion of cells treated with BBN for 6 months. Mechanistic studies revealed that 2-month of BBN exposure markedly attenuated DNMT3a abundance, and this led to reduced miR-219a promoter methylation, increased miR-219a binding to the RhoGDIß mRNA 3'UTR, and reduced RhoGDIß protein translation. While after 6-mo of BBN treatment, the cells showed increased PP2A/JNK/C-Jun axis phosphorylation and this in turn mediated overall RhoGDIß mRNA transcription and protein expression as well as invasion. CONCLUSIONS: These findings indicate that RhoGDIß is likely to inhibit the transformation of human urothelial cells during the early phase of BBN exposure, whereas it promotes invasion of the transformed/progressed urothelial cells in the late stage of BBN exposure. The studies also suggest that RhoGDIß may be a useful biomarker for evaluating the progression of human bladder cancers.


Subject(s)
MicroRNAs , Nitrosamines , Humans , rho Guanine Nucleotide Dissociation Inhibitor beta , Nitrosamines/toxicity , Epithelial Cells , Carcinogenesis
4.
J Transl Med ; 21(1): 620, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37700323

ABSTRACT

BACKGROUND: A significant proportion of septic patients with acute lung injury (ALI) are recognized late due to the absence of an efficient diagnostic test, leading to the postponed treatments and consequently higher mortality. Identifying diagnostic biomarkers may improve screening to identify septic patients at high risk of ALI earlier and provide the potential effective therapeutic drugs. Machine learning represents a powerful approach for making sense of complex gene expression data to find robust ALI diagnostic biomarkers. METHODS: The datasets were obtained from GEO and ArrayExpress databases. Following quality control and normalization, the datasets (GSE66890, GSE10474 and GSE32707) were merged as the training set, and four machine learning feature selection methods (Elastic net, SVM, random forest and XGBoost) were applied to construct the diagnostic model. The other datasets were considered as the validation sets. To further evaluate the performance and predictive value of diagnostic model, nomogram, Decision Curve Analysis (DCA) and Clinical Impact Curve (CIC) were constructed. Finally, the potential small molecular compounds interacting with selected features were explored from the CTD database. RESULTS: The results of GSEA showed that immune response and metabolism might play an important role in the pathogenesis of sepsis-induced ALI. Then, 52 genes were identified as putative biomarkers by consensus feature selection from all four methods. Among them, 5 genes (ARHGDIB, ALDH1A1, TACR3, TREM1 and PI3) were selected by all methods and used to predict ALI diagnosis with high accuracy. The external datasets (E-MTAB-5273 and E-MTAB-5274) demonstrated that the diagnostic model had great accuracy with AUC value of 0.725 and 0.833, respectively. In addition, the nomogram, DCA and CIC showed that the diagnostic model had great performance and predictive value. Finally, the small molecular compounds (Curcumin, Tretinoin, Acetaminophen, Estradiol and Dexamethasone) were screened as the potential therapeutic agents for sepsis-induced ALI. CONCLUSION: This consensus of multiple machine learning algorithms identified 5 genes that were able to distinguish ALI from septic patients. The diagnostic model could identify septic patients at high risk of ALI, and provide potential therapeutic targets for sepsis-induced ALI.


Subject(s)
Acute Lung Injury , Sepsis , Humans , Consensus , Sepsis/complications , Acetaminophen , Acute Lung Injury/diagnosis , Acute Lung Injury/etiology , Machine Learning , rho Guanine Nucleotide Dissociation Inhibitor beta
5.
FASEB J ; 36(8): e22465, 2022 08.
Article in English | MEDLINE | ID: mdl-35867072

ABSTRACT

Anlotinib has been demonstrated to be effective in advanced non-small cell lung cancer (NSCLC) patients. The response stratification of anlotinib remains unclear. In this study, plasma samples from 28 anlotinib-treated NSCLC patients (discovery cohort: 14 responders and 14 non-responders) were subjected to proteomic analysis, and plasma samples from 35 anlotinib-treated NSCLC patients (validation cohort) were subjected to validation analysis. Liquid chromatography-tandem mass spectrometry analysis was performed on samples with different time points, namely baseline (BL), best response (BR), and progression disease (PD). Bioinformatics analysis was performed to screen for the underlying differential proteins. Enzyme-linked immunosorbent assay was performed to detect plasma ARHGDIB, FN1, CDH1, and KNG1 levels respectively. The Kaplan-Meier survival analysis was used for biomarker-based responsive stratification. Our results indicated that differential proteins between responders and non-responders showed that proteomic technology potentially contributes to biomarker screening in plasma samples at BL. Furthermore, our results suggested that the detection of plasma ARHGDIB, FN1, CDH1, and KNG1 levels have potential predictive value for anlotinib response both in the discovery cohort and validation cohort. Collectively, this study offers novel insights into the value of plasma biomarker screening via proteomic examination and suggests that plasma ARHGDIB, FN1, CDH1, and KNG1 levels could be used as biomarkers for anlotinib stratification in NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Quinolines , Biomarkers , Carcinoma, Non-Small-Cell Lung/drug therapy , Early Detection of Cancer , Humans , Indoles , Lung Neoplasms/drug therapy , Proteomics , Quinolines/therapeutic use , rho Guanine Nucleotide Dissociation Inhibitor beta
6.
Environ Toxicol ; 38(5): 1063-1077, 2023 May.
Article in English | MEDLINE | ID: mdl-36793247

ABSTRACT

Leukemia is a type of disease in which hematopoietic stem cells proliferate clonally at the genetic level. We discovered previously by high-resolution mass spectrometry that diallyl disulfide (DADS), which is one of the effective ingredients of garlic, reduces the performance of RhoGDI2 from APL HL-60 cells. Although RhoGDI2 is oversubscribed in several cancer categories, the effect of RhoGDI2 in HL-60 cells has remained unexplained. We aimed to investigate the influence of RhoGDI2 on DADS-induced differentiation of HL-60 cells to elucidate the association among the effect of inhibition or over-expression of RhoGDI2 with HL-60 cell polarization, migration and invasion, which is important for establishing a novel generation of inducers to elicit leukemia cell polarization. Co-transfection with RhoGDI2-targeted miRNAs apparently decreases the malignant biological behavior of cells and upregulates cytopenias in DADS-treated HL-60 cell lines, which increases CD11b and decreases CD33 and mRNA levels of Rac1, PAK1 and LIMK1. Meanwhile, we generated HL-60 cell lines with high-expressing RhoGDI2. The proliferation, migration and invasion capacity of such cells were significantly increased by the treated with DADS, while the reduction capacity of the cells was decreased. There was a reduction in CD11b and an increase in CD33 production, as well as an increase in the mRNA levels of Rac1, PAK1 and LIMK1. It also confirmed that inhibition of RhoGDI2 attenuates the EMT cascade via the Rac1/Pak1/LIMK1 pathway, thereby inhibiting the malignant biological behavior of HL-60 cells. Thus, we considered that inhibition of RhoGDI2 expression might be a new therapeutic direction for the treatment of human promyelocytic leukemia. The anti-cancer property of DADS against HL-60 leukemia cells might be regulated by RhoGDI2 through the Rac1-Pak1-LIMK1 pathway, which provides new evidence for DADS as a clinical anti-cancer medicine.


Subject(s)
Leukemia , rho Guanine Nucleotide Dissociation Inhibitor beta , Humans , Allyl Compounds/pharmacology , Cell Differentiation/drug effects , Disulfides/pharmacology , HL-60 Cells/drug effects , HL-60 Cells/metabolism , Leukemia/metabolism , Leukemia/pathology , Lim Kinases/genetics , Lim Kinases/metabolism , p21-Activated Kinases/metabolism , p21-Activated Kinases/pharmacology , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/pharmacology , rho Guanine Nucleotide Dissociation Inhibitor beta/drug effects , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , RNA, Messenger , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
7.
Int J Mol Sci ; 24(4)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36835422

ABSTRACT

RhoGDI2 is a guanine nucleotide dissociation inhibitor (GDI) specific for the Rho family of small GTPases. It is highly expressed in hematopoietic cells but is also present in a large array of other cell types. RhoGDI2 has been implicated in multiple human cancers and immunity regulation, where it can display a dual role. Despite its involvement in various biological processes, we still do not have a clear understanding of its mechanistic functions. This review sheds a light on the dual opposite role of RhoGDI2 in cancer, highlights its underappreciated role in immunity and proposes ways to explain its intricate regulatory functions.


Subject(s)
Immunity , Neoplasms , rho Guanine Nucleotide Dissociation Inhibitor beta , Humans , Neoplasms/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
8.
J Proteome Res ; 21(9): 2146-2159, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35939567

ABSTRACT

High-grade serous ovarian cancer (HGSOC) represents the major histological type of ovarian cancer, and the lack of effective screening tools and early detection methods significantly contributes to the poor prognosis of HGSOC. Currently, there are no reliable diagnostic biomarkers for HGSOC. In this study, we performed liquid chromatography data-independent acquisition tandem mass spectrometry (MS) on depleted serum samples from 26 HGSOC cases and 24 healthy controls (HCs) to discover potential HGSOC diagnostic biomarkers. A total of 1,847 proteins were identified across all samples, among which 116 proteins showed differential expressions between HGSOC patients and HCs. Network modeling showed activations of coagulation and complement cascades, platelet activation and aggregation, neutrophil extracellular trap formation, toll-like receptor 4, insulin-like growth factor, and transforming growth factor ß signaling, as well as suppression of lipoprotein assembly and Fc gamma receptor activation in HGSOC. Based on the network model, we prioritized 28 biomarker candidates and validated 18 of them using targeted MS assays in an independent cohort. Predictive modeling showed a sensitivity of 1 and a specificity of 0.91 in the validation cohort. Finally, in vitro functional assays on four potential biomarkers (FGA, VWF, ARHGDIB, and SERPINF2) suggested that they may play an important role in cancer cell proliferation and migration in HGSOC. All raw data were deposited in PRIDE (PXD033169).


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Biomarkers, Tumor , Cohort Studies , Cystadenocarcinoma, Serous/diagnosis , Cystadenocarcinoma, Serous/pathology , Female , Humans , Mass Spectrometry , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , rho Guanine Nucleotide Dissociation Inhibitor beta
9.
Biochemistry ; 60(19): 1533-1551, 2021 05 18.
Article in English | MEDLINE | ID: mdl-33913706

ABSTRACT

There are three RhoGDIs in mammalian cells, which were initially defined as negative regulators of Rho family small GTPases. However, it is now accepted that RhoGDIs not only maintain small GTPases in their inactive GDP-bound form but also act as chaperones for small GTPases, targeting them to specific intracellular membranes and protecting them from degradation. Studies to date with RhoGDIs have usually focused on the interactions between the "typical" or "classical" small GTPases, such as the Rho, Rac, and Cdc42 subfamily members, and either the widely expressed RhoGDI-1 or the hematopoietic-specific RhoGDI-2. Less is known about the third member of the family, RhoGDI-3 and its interacting partners. RhoGDI-3 has a unique N-terminal extension and is found to localize in both the cytoplasm and the Golgi. RhoGDI-3 has been shown to target RhoB and RhoG to endomembranes. In order to facilitate a more thorough understanding of RhoGDI function, we undertook a systematic study to determine all possible Rho family small GTPases that interact with the RhoGDIs. RhoGDI-1 and RhoGDI-2 were found to have relatively restricted activity, mainly binding members of the Rho and Rac subfamilies. RhoGDI-3 displayed wider specificity, interacting with the members of Rho, Rac, and Cdc42 subfamilies but also forming complexes with "atypical" small Rho GTPases such as Wrch2/RhoV, Rnd2, Miro2, and RhoH. Levels of RhoA, RhoB, RhoC, Rac1, RhoH, and Wrch2/RhoV bound to GTP were found to decrease following coexpression with RhoGDI-3, confirming its role as a negative regulator of these small Rho GTPases.


Subject(s)
rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , rho Guanine Nucleotide Dissociation Inhibitor gamma/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , HEK293 Cells , Humans , Monomeric GTP-Binding Proteins/metabolism , Protein Binding , rho GTP-Binding Proteins/chemistry , rho Guanine Nucleotide Dissociation Inhibitor alpha/physiology , rho Guanine Nucleotide Dissociation Inhibitor beta/physiology , rho Guanine Nucleotide Dissociation Inhibitor gamma/physiology , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/physiology
10.
J Cell Biochem ; 122(7): 739-751, 2021 07.
Article in English | MEDLINE | ID: mdl-33586155

ABSTRACT

The appropriate regulation of spindle orientation maintains proper tissue homeostasis and avoids aberrant tissue repair or regeneration. Spindle misorientation due to imbalance or improper functioning leads to a loss of tissue integrity and aberrant growth, such as tissue loss or overgrowth. Pharmacological manipulation to prevent spindle misorientation will enable a better understanding of how spindle orientation is involved in physiological and pathological conditions and will provide therapeutic possibilities to treat patients associated with abnormal tissue function caused by spindle misorientation. N-terminal-deleted Rho guanine nucleotide dissociation inhibitor ß (RhoGDIß/RhoGDI2/LyGDI) produced by caspase-3 activation perturbs spindle orientation in surviving cells following exposure to either ionizing radiation or UVC. Thus, presumably, RhoGDIß cleaved by caspase-3 activation acts as a determinant of radiation-induced spindle misorientation that promote aberrant tissue repair due to deregulation of directional organization of cell population and therefore becomes a potential target of drugs to prevent such response. The objective of this study was to screen and identify chemicals that suppress RhoGDIß expression. We focused our attention on ascorbic acid (AA) derivatives because of their impact on the maintenance of skin tissue homeostasis. Here, we screened for AA derivatives that suppress RhoGDIß expression in HeLa cells and identified a lipophilic derivative, 2-O-octadecylascorbic acid (2-OctadecylAA), as a novel RhoGDIß inhibitor that ameliorated ionizing radiation-induced abnormal spindle orientations. Among all examined AA derivatives, which were also antioxidative, the inhibition activity was specific to 2-OctadecylAA. Therefore, this activity was not due to simple antioxidant properties. 2-OctadecylAA was previously shown to prevent hepatocellular carcinoma development. Our findings suggest that the anticarcinogenic effects of 2-OctadecylAA are partly due to RhoGDIß inhibition mechanisms by which spindle orientation perturbations are attenuated. Thus, the molecular targeting features of RhoGDIß warrant its further development for the treatment or control of spindle orientation abnormalities that affect epithelial homeostasis.


Subject(s)
Ascorbic Acid/analogs & derivatives , DNA Damage , Gene Expression Regulation/drug effects , Spindle Apparatus/pathology , rho Guanine Nucleotide Dissociation Inhibitor beta/antagonists & inhibitors , Ascorbic Acid/pharmacology , HeLa Cells , Humans , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
11.
Mol Cancer ; 20(1): 77, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006303

ABSTRACT

BACKGROUND: KDM6A, a histone demethylase, is frequently mutated in bladder cancer (BCa). However, the role and detailed molecular mechanism of KDM6A involved in bladder cancer progression remains unknown. METHODS: Tissue specimens were used to determine the expression levels and prognostic values of KDM6A and ARHGDIB. The MTT, colony formation, wound healing and Transwell migration and invasion assays were employed to detect the BCa cell proliferation, migration and invasion, respectively. Chemotaxis of macrophages was used to evaluate the ability of KDM6A to recruit macrophages. A subcutaneous tumour model and tail vein tumour injection in nude mice were used to assess the role of KDM6A in vivo. RNA sequencing, qPCR, Western blot, ChIP and phalloidin staining assay were performed to investigate the molecular functions of KDM6A. Dual-luciferase reporter assay was used to determine the effects of KDM6A and FOXA1 on the promoters of the ARHGDIB and KDM6A. RESULTS: We showed that the KDM6A inhibited the motility and invasiveness of the BCa cells. Mechanistically, KDM6A promotes the transcription of ARHGDIB by demethylating histone H3 lysine di/trimethylation (H3K27me2/3) and consequently leads to inhibition of Rac1. EZH2, which catalyses the methylation of H3K27, functions to silence ARHGDIB expression, and an EZH2 inhibitor can neutralize the metastatic effect caused by KDM6A deficiency. Furthermore, we demonstrated that FOXA1 directly binds to the KDM6A promoter and thus transactivates KDM6A, leading to diminished metastatic potential. CONCLUSION: Our findings establish the critical role of the FOXA1-KDM6A-ARHGDIB axis in restraining the malignancy of BCa and identify KDM6A and EZH2 as potential therapeutic targets in the management of BCa.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Histone Demethylases/metabolism , Urinary Bladder Neoplasms/pathology , rac1 GTP-Binding Protein/biosynthesis , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Animals , Cell Movement/physiology , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/pathology
12.
Acta Biochim Biophys Sin (Shanghai) ; 53(10): 1321-1332, 2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34508625

ABSTRACT

The fusion gene of ABL1 is closely related to tumor proliferation, invasion, and migration. It has been reported recently that ABL1 itself is required for T-cell acute lymphoblastic leukemia (T-ALL) cell migration induced by CXCL12. Further experiments revealed that ABL1 inhibitor Nilotinib inhibited leukemia cell migration induced by CXCL12, indicating the possible application of Nilotinib in T-ALL leukemia treatment. However, the interacting proteins of ABL1 and the specific mechanisms of their involvement in this process need further investigation. In the present study, ABL1 interacting proteins were characterized and their roles in the process of leukemia cell migration induced by CXCL12 were investigated. Co-immunoprecipitation in combination with mass spectrometry analysis identified 333 proteins that interact with ABL1, including Cofilin1. Gene ontology analysis revealed that many of them were enriched in the intracellular organelle or cytoplasm, including nucleic acid binding components, transfectors, or co-transfectors. Kyoto Encyclopedia of Genes and Genomes analysis showed that the top three enriched pathways were translation, glycan biosynthesis, and metabolism, together with human diseases. ABL1 and Cofilin1 were in the same complex. Cofilin1 binds the SH3 domain of ABL1 directly; however, ABL1 is not required for the phosphorylation of Cofilin1. Molecular docking analysis shows that ABL1 interacts with Cofilin1 mainly through hydrogen bonds and ionic interaction between amino acid residues. The mobility of leukemic cells was significantly decreased by Cofilin1 siRNA. These results demonstrate that Cofilin1 is a novel ABL1 binding partner. Furthermore, Cofilin1 participates in the migration of leukemia cells induced by CXCL12. These data indicate that ABL1 and Cofilin1 are possible targets for T-ALL treatment.


Subject(s)
Cell Movement/immunology , Cofilin 1/immunology , Cofilin 1/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-abl/immunology , Proto-Oncogene Proteins c-abl/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Cofilin 1/genetics , Computational Biology , Cytoskeleton/metabolism , Humans , Male , Mice , Mice, Inbred DBA , Molecular Docking Simulation , Protein Domains , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/pharmacology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
13.
J Cell Sci ; 131(4)2018 02 22.
Article in English | MEDLINE | ID: mdl-29361540

ABSTRACT

Unconventional myosin VIIA (Myo7a) is an actin-based motor molecule that normally functions in the cochlear hair cells of the inner ear. Mutations of MYO7A/Myo7a have been implicated in inherited deafness in both humans and mice. However, there is limited information about the functions of Myo7a outside of the specialized cells of the ears. Herein, we report a previously unidentified function of Myo7a by demonstrating that it plays an important role in melanoma progression. We found that silencing Myo7a by means of RNAi inhibited melanoma cell growth through upregulation of cell cycle regulator p21 (also known as CDKN1A) and suppressed melanoma cell migration and invasion through downregulation of RhoGDI2 (also known as ARHGDIB) and MMP9. Furthermore, Myo7a depletion suppressed melanoma cell metastases to the lung, kidney and bone in mice. In contrast, overexpression of Myo7a promoted melanoma xenograft growth and lung metastasis. Importantly, Myo7a levels are remarkably elevated in human melanoma patients. Collectively, we demonstrated for the first time that Myo7a is able to function in non-specialized cells, a finding that reveals the complicated disease-related roles of Myo7a, especially in melanomas.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Melanoma/genetics , Myosins/genetics , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Ear, Inner/metabolism , Ear, Inner/pathology , Gene Expression Regulation, Neoplastic , Gene Silencing , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Matrix Metalloproteinase 9/genetics , Melanoma/pathology , Mice , Mutation , Myosin VIIa , Myosins/antagonists & inhibitors , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Xenograft Model Antitumor Assays
14.
Am J Transplant ; 19(12): 3335-3344, 2019 12.
Article in English | MEDLINE | ID: mdl-31194283

ABSTRACT

The clinical significance of non-HLA antibodies on renal allograft survival is a matter of debate, due to differences in reported results and lack of large-scale studies incorporating analysis of multiple non-HLA antibodies simultaneously. We developed a multiplex non-HLA antibody assay against 14 proteins highly expressed in the kidney. In this study, the presence of pretransplant non-HLA antibodies was correlated to renal allograft survival in a nationwide cohort of 4770 recipients transplanted between 1995 and 2006. Autoantibodies against Rho GDP-dissociation inhibitor 2 (ARHGDIB) were significantly associated with graft loss in recipients transplanted with a deceased-donor kidney (N = 3276) but not in recipients of a living-donor kidney (N = 1496). At 10 years after deceased-donor transplantation, recipients with anti-ARHGDIB antibodies (94/3276 = 2.9%) had a 13% lower death-censored covariate-adjusted graft survival compared to the anti-ARHGDIB-negative (3182/3276 = 97.1%) population (hazard ratio 1.82; 95% confidence interval, 1.32-2.53; P = .0003). These antibodies occur independently from donor-specific anti-HLA antibodies (DSA) or other non-HLA antibodies investigated. No significant relations with graft loss were found for the other 13 non-HLA antibodies. We suggest that pretransplant risk assessment can be improved by measuring anti-ARHGDIB antibodies in all patients awaiting deceased-donor transplantation.


Subject(s)
Autoantibodies/immunology , Graft Rejection/mortality , Graft Survival/immunology , HLA Antigens/immunology , Kidney Transplantation/adverse effects , Postoperative Complications/mortality , rho Guanine Nucleotide Dissociation Inhibitor beta/immunology , Adult , Female , Follow-Up Studies , Graft Rejection/diagnosis , Graft Rejection/etiology , Humans , Isoantibodies/immunology , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/mortality , Kidney Failure, Chronic/surgery , Living Donors/statistics & numerical data , Male , Middle Aged , Postoperative Complications/diagnosis , Postoperative Complications/etiology , Prognosis , Retrospective Studies , Risk Factors
15.
Mol Carcinog ; 58(5): 777-793, 2019 05.
Article in English | MEDLINE | ID: mdl-30604907

ABSTRACT

Although overexpression of the non-canonical NFκB subunit p52 has been observed in several tumors, the function and mechanism of p52 in bladder cancer (BC) are less well understood. Here, we aimed at understanding the role and mechanism underlying p52 regulation of BC invasion. Human p52 was stably knockdown with shRNA targeting p52 in two bladder cancer cell lines (T24 and UMUC3). Two constitutively expressing constructs, p52 and p100, were stably transfected in to T24 or UMUC3, respectively. The stable transfectants were used to determine function and mechanisms responsible for p52 regulation of BC invasion. We demonstrate that p52 mediates human BC invasion. Knockdown of p52 impaired bladder cancer invasion by reduction of rhogdiß mRNA stability and expression. Positively regulation of rhogdiß mRNA stability was mediated by p52 promoting AUF1 protein degradation, consequently resulting in reduction of AUF1 binding to rhogdiß mRNA. Further studies indicated that AUF1 protein degradation was mediated by upregulating USP8 transcription, which was modulated by its negative regulatory transcription factor Sp1. Moreover, we found that p52 upregulated miR-145, which directly bound to the 3'-UTR of sp1 mRNA, leading to downregulation of Sp1 protein translation. Our results reveal a comprehensive pathway that p52 acts as a positive regulator of BC invasion by initiating a novel miR-145/Sp1/USP8/AUF1/RhoGDIß axis. These findings provide insight into the understanding of p52 in the pathology of human BC invasion and progression, which may be useful information in the development of preventive and therapeutic approaches for using p52 as a potential target.


Subject(s)
Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , MicroRNAs/metabolism , NF-kappa B p52 Subunit/metabolism , RNA Stability , Sp1 Transcription Factor/metabolism , Ubiquitin Thiolesterase/metabolism , Urinary Bladder Neoplasms/pathology , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Humans , MicroRNAs/genetics , NF-kappa B p52 Subunit/genetics , Protein Biosynthesis , Proteolysis , Sp1 Transcription Factor/genetics , Tumor Cells, Cultured , Ubiquitin Thiolesterase/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/chemistry , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics
16.
Int J Cancer ; 142(10): 2040-2055, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29250796

ABSTRACT

Our recent studies demonstrate that X-linked inhibitor of apoptosis protein (XIAP) is essential for regulating colorectal cancer invasion. Here, we discovered that RhoGDIß was a key XIAP downstream effector mediating bladder cancer (BC) invasion in vitro and in vivo. We found that both XIAP and RhoGDIß expressions were consistently elevated in BCs of N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-treated mice in comparison to bladder tissues from vehicle-treated mice and human BCs in comparison to the paired adjacent normal bladder tissues. Knockdown of XIAP attenuated RhoGDIß expression and reduced cancer cell invasion, whereas RhoGDIß expression was attenuated in BBN-treated urothelium of RING-deletion knockin mice. Mechanistically, XIAP stabilized RhoGDIß mRNA by its positively regulating nucleolin mRNA stability via Erks-dependent manner. Moreover, ectopic expression of GFP-RhoGDIß in T24T(shXIAP) cells restored its lung metastasis in nude mice. Our results demonstrate that XIAP-regulated Erks/nucleolin/RhoGDIß axis promoted BC invasion and lung metastasis.


Subject(s)
Inhibitor of Apoptosis Proteins/biosynthesis , Lung Neoplasms/secondary , Phosphoproteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Urinary Bladder Neoplasms/pathology , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics , Animals , Cell Line, Tumor , Female , HCT116 Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Invasiveness , RNA, Messenger/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Nucleolin
17.
Am J Physiol Cell Physiol ; 312(4): C527-C536, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28148498

ABSTRACT

On activation at sites of vascular injury, platelets undergo morphological alterations essential to hemostasis via cytoskeletal reorganizations driven by the Rho GTPases Rac1, Cdc42, and RhoA. Here we investigate roles for Rho-specific guanine nucleotide dissociation inhibitor proteins (RhoGDIs) in platelet function. We find that platelets express two RhoGDI family members, RhoGDI and Ly-GDI. Whereas RhoGDI localizes throughout platelets in a granule-like manner, Ly-GDI shows an asymmetric, polarized localization that largely overlaps with Rac1 and Cdc42 as well as microtubules and protein kinase C (PKC) in platelets adherent to fibrinogen. Antibody interference and platelet spreading experiments suggest a specific role for Ly-GDI in platelet function. Intracellular signaling studies based on interactome and pathways analyses also support a regulatory role for Ly-GDI, which is phosphorylated at PKC substrate motifs in a PKC-dependent manner in response to the platelet collagen receptor glycoprotein (GP) VI-specific agonist collagen-related peptide. Additionally, PKC inhibition diffuses the polarized organization of Ly-GDI in spread platelets relative to its colocalization with Rac1 and Cdc42. Together, our results suggest a role for Ly-GDI in the localized regulation of Rho GTPases in platelets and hypothesize a link between the PKC and Rho GTPase signaling systems in platelet function.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/physiology , Platelet Activation/physiology , Platelet Adhesiveness/physiology , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , Cells, Cultured , Hemostasis/physiology , Humans , Signal Transduction/physiology , Subcellular Fractions/metabolism
18.
Biochim Biophys Acta ; 1861(12 Pt A): 2011-2019, 2016 12.
Article in English | MEDLINE | ID: mdl-27720952

ABSTRACT

Retinoic acid (RA) has a variety of biological effects in mammalian cells and tissues. It is well known that RA induces differentiation of human acute promyelocytic leukemia (APL) HL60 cells, fresh human APL cells, and clinical remission in patients with APL. Retinoylation (acylation of proteins by RA) is a possible pathway for RA action. However, an understanding of the role that retinoylation plays in the actions of RA is lacking. In the current study, several retinoylated proteins were detected in RA-treated HL60 fractions following Mono Q anion exchange chromatography and analysis using two-dimensional polyacrylamide gel electrophoresis. One of the retinoylated proteins was identified as Rho-GDIß (28kDa) by TOF-MS and co-migration with Rho-GDIß (28kDa). Truncated Rho-GDIß (23kDa, N∆19), a product of cleavage by caspase-3, was not retinoylated. RA covalently bound to the Thr2 residue in Rho-GDIß (5kDa), which is the second product resulting from the cleavage of Rho-GDIß (28kDa) by caspase-3. RA treatment increased the level of Rho-GDIß (28kDa) and decreased the level of Rho-GDIß (23kDa). RA did not induce caspase-3 activity or Rho-GDIß mRNA expression. It is likely that retinoylation of Rho-GDIß increases its metabolic stability.


Subject(s)
Acylation/physiology , Leukemia, Myeloid/metabolism , Tretinoin/pharmacology , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Acylation/drug effects , Amino Acid Sequence , Caspase 3/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , HL-60 Cells , Humans , RNA/metabolism , RNA, Messenger/metabolism
19.
Breast Cancer Res ; 19(1): 74, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28666462

ABSTRACT

BACKGROUND: The importance of the mTOR complex 2 (mTORC2) signaling complex in tumor progression is becoming increasingly recognized. HER2-amplified breast cancers use Rictor/mTORC2 signaling to drive tumor formation, tumor cell survival and resistance to human epidermal growth factor receptor 2 (HER2)-targeted therapy. Cell motility, a key step in the metastatic process, can be activated by mTORC2 in luminal and triple negative breast cancer cell lines, but its role in promoting metastases from HER2-amplified breast cancers is not yet clear. METHODS: Because Rictor is an obligate cofactor of mTORC2, we genetically engineered Rictor ablation or overexpression in mouse and human HER2-amplified breast cancer models for modulation of mTORC2 activity. Signaling through mTORC2-dependent pathways was also manipulated using pharmacological inhibitors of mTOR, Akt, and Rac. Signaling was assessed by western analysis and biochemical pull-down assays specific for Rac-GTP and for active Rac guanine nucleotide exchange factors (GEFs). Metastases were assessed from spontaneous tumors and from intravenously delivered tumor cells. Motility and invasion of cells was assessed using Matrigel-coated transwell assays. RESULTS: We found that Rictor ablation potently impaired, while Rictor overexpression increased, metastasis in spontaneous and intravenously seeded models of HER2-overexpressing breast cancers. Additionally, migration and invasion of HER2-amplified human breast cancer cells was diminished in the absence of Rictor, or upon pharmacological mTOR kinase inhibition. Active Rac1 was required for Rictor-dependent invasion and motility, which rescued invasion/motility in Rictor depleted cells. Rictor/mTORC2-dependent dampening of the endogenous Rac1 inhibitor RhoGDI2, a factor that correlated directly with increased overall survival in HER2-amplified breast cancer patients, promoted Rac1 activity and tumor cell invasion/migration. The mTORC2 substrate Akt did not affect RhoGDI2 dampening, but partially increased Rac1 activity through the Rac-GEF Tiam1, thus partially rescuing cell invasion/motility. The mTORC2 effector protein kinase C (PKC)α did rescue Rictor-mediated RhoGDI2 downregulation, partially rescuing Rac-guanosine triphosphate (GTP) and migration/motility. CONCLUSION: These findings suggest that mTORC2 uses two coordinated pathways to activate cell invasion/motility, both of which converge on Rac1. Akt signaling activates Rac1 through the Rac-GEF Tiam1, while PKC signaling dampens expression of the endogenous Rac1 inhibitor, RhoGDI2.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 2/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement/genetics , Disease Models, Animal , Female , Gene Amplification , Heterografts , Humans , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism
20.
Biochim Biophys Acta Gen Subj ; 1861(1 Pt A): 3061-3070, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27721047

ABSTRACT

BACKGROUND: The delineation of intrinsically weak interactions between novel targets and fragment screening hits has long limited the pace of hit-to-lead evolution. Rho guanine-nucleotide dissociation inhibitor 2 (RhoGDI2) is a novel target that lacks any chemical probes for the treatment of tumor metastasis. METHODS: Protein-observed and ligand-observed NMR spectroscopy was used to characterize the weak interactions between RhoGDI2 and fragment screening hits. RESULTS: We identified three hits of RhoGDI2 using streamlined NMR fragment-based screening. The binding site residues were assigned using non-uniformly sampled Cα- and Hα-based three dimensional NMR spectra. The molecular docking to the proposed geranylgeranyl binding pocket of RhoGDI2 was guided by NMR restraints of chemical shift perturbations and ligand-observed transferred paramagnetic relaxation enhancement. We further validated the weak RhoGDI2-hit interactions using mutagenesis and structure-affinity analysis. CONCLUSIONS: Weak interactions between RhoGDI2 and fragment screening hits were delineated using an integrated NMR approach. GENERAL INTERESTS: Binders to RhoGDI2 as a potential anti-cancer target have been first reported, and their weak interactions were depicted using NMR spectroscopy. Our work highlights the powerfulness and the versatility of the integrative NMR techniques to provide valuable structural insight into the intrinsically weak interactions between RhoGDI2 and the fragment screening hits, which could hardly be conceived using other biochemical techniques.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Peptide Fragments/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Reproducibility of Results , Small Molecule Libraries/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL