1
|
Hebron KE, Perkins OL, Kim A, Jian X, Girald-Berlingeri SA, Lei H, Shern JF, Conner EA, Randazzo PA, Yohe ME. ASAP1 and ARF1 Regulate Myogenic Differentiation in Rhabdomyosarcoma by Modulating TAZ Activity. Mol Cancer Res 2025; 23:95-106. [PMID: 39495123 PMCID: PMC11799837 DOI: 10.1158/1541-7786.mcr-24-0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Despite aggressive, multimodal therapies, the prognosis of patients with refractory or recurrent rhabdomyosarcoma (RMS) has not improved in four decades. Because RMS resembles skeletal muscle precursor cells, differentiation-inducing therapy has been proposed for patients with advanced disease. In RAS-mutant PAX fusion-negative RMS (FN-RMS) preclinical models, MEK1/2 inhibition (MEKi) induces differentiation, slows tumor growth, and extends survival. However, the response is short-lived. A better understanding of the molecular mechanisms regulating FN-RMS differentiation could improve differentiation therapy. In this study, we identified a role in FN-RMS differentiation for ASAP1, an ADP ribosylation factor (ARF) GTPase-activating protein (GAP) with both proinvasive and tumor-suppressor functions. We found that ASAP1 knockdown inhibited differentiation in FN-RMS cells. Interestingly, knockdown of the GTPases ARF1 or ARF5, targets of ASAP1 GAP activity, also blocked differentiation of FN-RMS. We discovered that loss of ARF pathway components blocked myogenic transcription factor expression. Therefore, we examined the effects on transcriptional regulators. MEKi led to the phosphorylation and inactivation of WW domain-containing transcriptional regulator 1 (WWTR1; TAZ), a homolog of the pro-proliferative transcriptional co-activator YAP1, regulated by the Hippo pathway. However, loss of ASAP1 or ARF1 blocked this inactivation, which inhibits MEKi-induced differentiation. Finally, MEKi-induced differentiation was rescued by dual knockdown of ASAP1 and WWTR1. This study shows that ASAP1 and ARF1 are necessary for myogenic differentiation, providing a deeper understanding of differentiation in FN-RMS and illuminating an opportunity to advance differentiation therapy. Implications: ASAP1 and ARF1 regulate MEKi-induced differentiation of FN-RMS cells by modulating WWTR1 (TAZ) activity, supporting YAP1/TAZ inhibition as a FN-RMS differentiation therapy strategy.
Collapse
Affiliation(s)
- Katie E. Hebron
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Olivia L. Perkins
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Angela Kim
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Xiaoying Jian
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sofia A. Girald-Berlingeri
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Haiyan Lei
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jack F. Shern
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth A. Conner
- Center for Cancer Research Genomics Core, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Paul A. Randazzo
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Marielle E. Yohe
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
2
|
Barbagallo C, Stella M, Di Mauro S, Scamporrino A, Filippello A, Scionti F, Di Martino MT, Purrello M, Ragusa M, Purrello F, Piro S. An Uncharacterised lncRNA Coded by the ASAP1 Locus Is Downregulated in Serum of Type 2 Diabetes Mellitus Patients. Int J Mol Sci 2023; 24:13485. [PMID: 37686290 PMCID: PMC10488254 DOI: 10.3390/ijms241713485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/23/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Diabetes mellitus (DM) is a complex and multifactorial disease characterised by high blood glucose. Type 2 Diabetes (T2D), the most frequent clinical condition accounting for about 90% of all DM cases worldwide, is a chronic disease with slow development usually affecting middle-aged or elderly individuals. T2D represents a significant problem of public health today because its incidence is constantly growing among both children and adults. It is also estimated that underdiagnosis prevalence would strongly further increase the real incidence of the disease, with about half of T2D patients being undiagnosed. Therefore, it is important to increase diagnosis accuracy. The current interest in RNA molecules (both protein- and non-protein-coding) as potential biomarkers for diagnosis, prognosis, and treatment lies in the ease and low cost of isolation and quantification with basic molecular biology techniques. In the present study, we analysed the transcriptome in serum samples collected from T2D patients and unaffected individuals to identify potential RNA-based biomarkers. Microarray-based profiling and subsequent validation using Real-Time PCR identified an uncharacterised long non-coding RNA (lncRNA) transcribed from the ASAP1 locus as a potential diagnostic biomarker. ROC curve analysis showed that a molecular signature including the lncRNA and the clinicopathological parameters of T2D patients as well as unaffected individuals showed a better diagnostic performance compared with the glycated haemoglobin test (HbA1c). This result suggests that the application of this biomarker in clinical practice would help to improve the diagnosis, and therefore the clinical management, of T2D patients. The proposed biomarker would be useful in the context of predictive, preventive, and personalised medicine (3PM/PPPM).
Collapse
Affiliation(s)
- Cristina Barbagallo
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.P.)
| | - Michele Stella
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.P.)
| | - Stefania Di Mauro
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (F.P.); (S.P.)
| | - Alessandra Scamporrino
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (F.P.); (S.P.)
| | - Agnese Filippello
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (F.P.); (S.P.)
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (F.S.); (M.T.D.M.)
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (F.S.); (M.T.D.M.)
| | - Michele Purrello
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.P.)
| | - Marco Ragusa
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.P.)
| | - Francesco Purrello
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (F.P.); (S.P.)
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, Internal Medicine, Garibaldi-Nesima Hospital, University of Catania, 95122 Catania, Italy; (S.D.M.); (A.S.); (A.F.); (F.P.); (S.P.)
| |
Collapse
|
3
|
Huo X, Zhang W, Zhao G, Chen Z, Dong P, Watari H, Narayanan R, Tillmanns TD, Pfeffer LM, Yue J. FAK PROTAC Inhibits Ovarian Tumor Growth and Metastasis by Disrupting Kinase Dependent and Independent Pathways. Front Oncol 2022; 12:851065. [PMID: 35574330 PMCID: PMC9095959 DOI: 10.3389/fonc.2022.851065] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 04/04/2022] [Indexed: 12/14/2022] Open
Abstract
Focal adhesion kinase (FAK) is highly expressed in a variety of human cancers and is a target for cancer therapy. Since FAK kinase inhibitors only block the kinase activity of FAK, they are not highly effective in clinical trials. FAK also functions as a scaffold protein in a kinase-independent pathway. To effectively target FAK, it is required to block both FAK kinase-dependent and FAK-independent pathways. Thus, we tested a new generation drug FAK PROTAC for ovarian cancer therapy, which blocks both kinase and scaffold activity. We tested the efficacy of FAK PROTAC and its parent kinase inhibitor (VS-6063) in ovarian cancer cell lines in vitro by performing cell functional assays including cell proliferation, migration, invasion. We also tested in vivo activity in orthotopic ovarian cancer mouse models. In addition, we assessed whether FAK PROTAC disrupts kinase-dependent and kinase-independent pathways. We demonstrated that FAK PROTAC is highly effective as compared to its parent FAK kinase inhibitor VS-6063 in inhibiting cell proliferation, survival, migration, and invasion. FAK PROTAC not only inhibits the FAK kinase activity but also FAK scaffold function by disrupting the interaction between FAK and its interaction protein ASAP1. We further showed that FAK PROTAC effectively inhibits ovarian tumor growth and metastasis. Taken together, FAK PROTAC inhibits both FAK kinase activity and its scaffold protein activity by disrupting the interaction between FAK and ASAP1 and is highly effective in inhibiting ovarian tumor growth and metastasis.
Collapse
Affiliation(s)
- Xueyun Huo
- School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wenjing Zhang
- Department of Genetics, Genomics & Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Guannan Zhao
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Zhenwen Chen
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Peixin Dong
- Department of Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hidemichi Watari
- Department of Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ramesh Narayanan
- Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Todd D Tillmanns
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Tennessee Health Science Center, West Cancer Center, Germantown, TN, United States
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States.,Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
4
|
Gasilina A, Yoon HY, Jian X, Luo R, Randazzo PA. A lysine-rich cluster in the N-BAR domain of ARF GTPase-activating protein ASAP1 is necessary for binding and bundling actin filaments. J Biol Chem 2022; 298:101700. [PMID: 35143843 PMCID: PMC8902617 DOI: 10.1016/j.jbc.2022.101700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/05/2022] Open
Abstract
Actin filament maintenance is critical for both normal cell homeostasis and events associated with malignant transformation. The ADP-ribosylation factor GTPase-activating protein ASAP1 regulates the dynamics of filamentous actin-based structures, including stress fibers, focal adhesions, and circular dorsal ruffles. Here, we have examined the molecular basis for ASAP1 association with actin. Using a combination of structural modeling, mutagenesis, and in vitro and cell-based assays, we identify a putative-binding interface between the N-Bin-Amphiphysin-Rvs (BAR) domain of ASAP1 and actin filaments. We found that neutralization of charges and charge reversal at positions 75, 76, and 79 of ASAP1 reduced the binding of ASAP1 BAR-pleckstrin homology tandem to actin filaments and abrogated actin bundle formation in vitro. In addition, overexpression of actin-binding defective ASAP1 BAR-pleckstrin homology [K75, K76, K79] mutants prevented cellular actin remodeling in U2OS cells. Exogenous expression of [K75E, K76E, K79E] mutant of full-length ASAP1 did not rescue the reduction of cellular actin fibers consequent to knockdown of endogenous ASAP1. Taken together, our results support the hypothesis that the lysine-rich cluster in the N-BAR domain of ASAP1 is important for regulating actin filament organization.
Collapse
Affiliation(s)
- Anjelika Gasilina
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Hye-Young Yoon
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiaoying Jian
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ruibai Luo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
5
|
Chen PW, Gasilina A, Yadav MP, Randazzo PA. Control of cell signaling by Arf GTPases and their regulators: Focus on links to cancer and other GTPase families. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119171. [PMID: 34774605 DOI: 10.1016/j.bbamcr.2021.119171] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/23/2021] [Accepted: 11/04/2021] [Indexed: 12/31/2022]
Abstract
The ADP-ribosylation factors (Arfs) comprise a family of regulatory GTP binding proteins. The Arfs regulate membrane trafficking and cytoskeleton remodeling, processes critical for eukaryotes and which have been the focus of most studies on Arfs. A more limited literature describes a role in signaling and in integrating several signaling pathways to bring about specific cell behaviors. Here, we will highlight work describing function of Arf1, Arf6 and several effectors and regulators of Arfs in signaling.
Collapse
Affiliation(s)
- Pei-Wen Chen
- Department of Biology, Williams College, Williamstown, MA, United States of America
| | - Anjelika Gasilina
- National Heart, Lung and Blood Institute, NIH, Bethesda, MD, United States of America(1); Laboratory of Cellular and Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, United States of America
| | - Mukesh P Yadav
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, United States of America
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, NIH, Bethesda, MD, United States of America.
| |
Collapse
|
6
|
Wang B, Li H, Zhao X, Zhang W, Zhao G, Wu Z, Zhang R, Dong P, Watari H, Tigyi G, Li W, Yue J. A Luminacin D Analog HL142 Inhibits Ovarian Tumor Growth and Metastasis by Reversing EMT and Attenuating the TGFβ and FAK Pathways. J Cancer 2021; 12:5654-5663. [PMID: 34405025 PMCID: PMC8364639 DOI: 10.7150/jca.61066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/07/2021] [Indexed: 01/10/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) is known to contribute to tumor metastasis and chemoresistance. Reversing EMT using small molecule inhibitors to target EMT associated gene expression represents an effective strategy for cancer treatment. The purpose of this study is to test whether a new luminacin D analog HL142 reverses EMT in ovarian cancer (OC) and has the therapeutic potential for OC. We chemically synthesized HL142 and tested its functions in OC cells in vitro and its efficacy in inhibiting ovarian tumor growth and metastasis in vivo using orthotopic OC mouse models. We first demonstrate that ASAP1 is co-amplified and interacts with the focal adhesion kinase (FAK) protein in serous ovarian carcinoma. HL142 inhibits ASAP1 and its interaction protein FAK in highly invasive OVCAR8 and moderately invasive OVCAR3 cells. HL142 inhibits EMT phenotypic switch, accompanied by upregulating epithelial marker E-cadherin and cytokeratin-7 and downregulating mesenchymal markers vimentin, β-catenin, and snail2 in both cell lines. Functionally, HL142 inhibits proliferation, colony formation, migration, and invasion. HL142 also sensitizes cell responses to chemotherapy drug paclitaxel treatment and inhibits ovarian tumor growth and metastasis in orthotopic OC mouse models. We further show that HL142 attenuates the TGFβ and FAK pathways in vitro using OC cells and in vivo using orthotopic mouse models.
Collapse
Affiliation(s)
- Baojin Wang
- Department of Gynecology and Obstetrics, Third Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.,Department of Pathology and Laboratory Medicine, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Hanxuan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Xinxin Zhao
- Department of Gynecology and Obstetrics, Third Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.,Department of Pathology and Laboratory Medicine, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Wenjing Zhang
- Department of Genetics, Genomics & Informatics, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Guannan Zhao
- Department of Pathology and Laboratory Medicine, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Zhongzhi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Ruitao Zhang
- Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Peixin Dong
- Department of Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hidemichi Watari
- Department of Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Gabor Tigyi
- Center for Cancer Research, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Department of Physiology, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, College of Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| |
Collapse
|
7
|
Zhong H, Magee MJ, Huang Y, Hui Q, Gwinn M, Gandhi NR, Sun YV. Evaluation of the Host Genetic Effects of Tuberculosis-Associated Variants Among Patients With Type 1 and Type 2 Diabetes Mellitus. Open Forum Infect Dis 2020; 7:ofaa106. [PMID: 32328508 PMCID: PMC7166116 DOI: 10.1093/ofid/ofaa106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/24/2020] [Indexed: 11/17/2022] Open
Abstract
Background Understanding the link between tuberculosis (TB) and diabetes is increasingly important as public health responds to the growing global burden of noncommunicable diseases. Genetic association studies have identified numerous host genetic variants linked to TB; however, potential host genetic mechanisms linking TB and diabetes remain unexplored. Methods We used genetic and phenotypic data from the UK Biobank to evaluate the association of 6 previously reported TB-related host genetic variants (genome-wide significant associations from published studies) with diabetes. The study included 409 692 adults of European ancestry including 2177 with type 1 diabetes mellitus (T1DM) and 13 976 with type 2 diabetes mellitus (T2DM), defined by ICD-10 diagnosis codes. Results Of the 6 TB-associated single nucleotide polymorphisms (SNPs), 2 were associated with T1DM and 3 with T2DM, after adjusting for age, sex, body mass index, smoking, alcohol use, and population structure. After correction for multiple testing, SNPs rs2894257 and rs3135359 (HLA-DRA-DQA1) were associated with T1DM (rs2894257: odds ratio [OR], 1.32; 95% confidence interval [CI], 1.21–1.45; rs3135359: OR, 1.72; 95% CI, 1.57–1.88) and T2DM (rs2894257: OR, 1.11; 95% CI, 1.08–1.15; rs3135359: OR, 1.06; 95% CI, 1.025–1.096). The associations with T2DM weakened for rs2894257 and rs3135359 after further exclusion of probable T1DM cases defined by International Statistical Classification of Diseases and Related Health Problems (ICD-10) codes. SNP rs4733781 on chromosome 8 (ASAP1 gene) was associated with T2DM after exclusion of T1DM cases. Conclusions Our findings suggest that common host genetic effects may play a role in the molecular mechanism linking TB and diabetes. Future large genetic studies of TB and diabetes should focus on developing countries with high burdens of infectious and chronic diseases.
Collapse
Affiliation(s)
- Huimin Zhong
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Matthew J Magee
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Yunfeng Huang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Qin Hui
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Marta Gwinn
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Neel R Gandhi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA.,Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA.,Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA.,Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Whole genome detection of recent selection signatures in Sarabi cattle: a unique Iranian taurine breed. Genes Genomics 2019; 42:203-215. [DOI: 10.1007/s13258-019-00888-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022]
|
9
|
Gasilina A, Vitali T, Luo R, Jian X, Randazzo PA. The ArfGAP ASAP1 Controls Actin Stress Fiber Organization via Its N-BAR Domain. iScience 2019; 22:166-180. [PMID: 31785555 PMCID: PMC6889188 DOI: 10.1016/j.isci.2019.11.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/30/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022] Open
Abstract
ASAP1 is a multi-domain ArfGAP that controls cell migration, spreading, and focal adhesion dynamics. Although its GAP activity contributes to remodeling of the actin cytoskeleton, it does not fully explain all cellular functions of ASAP1. Here we find that ASAP1 regulates actin filament assembly directly through its N-BAR domain and controls stress fiber maintenance. ASAP1 depletion caused defects in stress fiber organization. Conversely, overexpression of ASAP1 enhanced actin remodeling. The BAR-PH fragment was sufficient to affect actin. ASAP1 with the BAR domain replaced with the BAR domain of the related ACAP1 did not affect actin. The BAR-PH tandem of ASAP1 bound and bundled actin filaments directly, whereas the presence of the ArfGAP and the C-terminal linker/SH3 domain reduced binding and bundling of filaments by BAR-PH. Together these data provide evidence that ASAP1 may regulate the actin cytoskeleton through direct interaction of the BAR-PH domain with actin filaments.
Collapse
Affiliation(s)
- Anjelika Gasilina
- Section on Regulation of Ras Superfamily, Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 37, Rm. 2042, Bethesda, MD 20892, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Teresa Vitali
- Section on Regulation of Ras Superfamily, Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 37, Rm. 2042, Bethesda, MD 20892, USA
| | - Ruibai Luo
- Section on Regulation of Ras Superfamily, Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 37, Rm. 2042, Bethesda, MD 20892, USA
| | - Xiaoying Jian
- Section on Regulation of Ras Superfamily, Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 37, Rm. 2042, Bethesda, MD 20892, USA
| | - Paul A Randazzo
- Section on Regulation of Ras Superfamily, Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 37, Rm. 2042, Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Schreiber C, Saraswati S, Harkins S, Gruber A, Cremers N, Thiele W, Rothley M, Plaumann D, Korn C, Armant O, Augustin HG, Sleeman JP. Loss of ASAP1 in mice impairs adipogenic and osteogenic differentiation of mesenchymal progenitor cells through dysregulation of FAK/Src and AKT signaling. PLoS Genet 2019; 15:e1008216. [PMID: 31246957 PMCID: PMC6619832 DOI: 10.1371/journal.pgen.1008216] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/10/2019] [Accepted: 05/27/2019] [Indexed: 11/18/2022] Open
Abstract
ASAP1 is a multi-domain adaptor protein that regulates cytoskeletal dynamics, receptor recycling and intracellular vesicle trafficking. Its expression is associated with poor prognosis for a variety of cancers, and promotes cell migration, invasion and metastasis. Little is known about its physiological role. In this study, we used mice with a gene-trap inactivated ASAP1 locus to study the functional role of ASAP1 in vivo, and found defects in tissues derived from mesenchymal progenitor cells. Loss of ASAP1 led to growth retardation and delayed ossification typified by enlarged hypertrophic zones in growth plates and disorganized chondro-osseous junctions. Furthermore, loss of ASAP1 led to delayed adipocyte development and reduced fat depot formation. Consistently, deletion of ASAP1 resulted in accelerated chondrogenic differentiation of mesenchymal cells in vitro, but suppressed osteo- and adipogenic differentiation. Mechanistically, we found that FAK/Src and PI3K/AKT signaling is compromised in Asap1GT/GT MEFs, leading to impaired adipogenic differentiation. Dysregulated FAK/Src and PI3K/AKT signaling is also associated with attenuated osteogenic differentiation. Together these observations suggest that ASAP1 plays a decisive role during the differentiation of mesenchymal progenitor cells. Mesenchymal progenitor cells are capable of differentiating into a number of lineages including osteoblasts, chondrocytes and adipocytes, and have therefore attracted interest for their potential application in regenerative medicine. Furthermore, defects in mesenchymal progenitor cell differentiation are considered to contribute to various diseases including metabolic syndrome, obesity and osteoporosis. In this study, we analyzed mice deficient in the multi-adaptor protein ASAP1, which has been implicated in tumor progression and metastasis. These mice display growth retardation, and a delayed development of bone and fat tissue. Consistently, mesenchymal progenitor cells deficient in ASAP1 exhibited enhanced differentiation into chondrocytes, but impaired differentiation into adipocytes and osteoblasts. Together these observations suggest that ASAP1 plays a decisive role during the differentiation of mesenchymal stem cells, which may be relevant for a number of diseases such as cancer.
Collapse
Affiliation(s)
- Caroline Schreiber
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- * E-mail:
| | - Supriya Saraswati
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Shannon Harkins
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Annette Gruber
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Natascha Cremers
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Institute for Toxicology and Genetics, KIT Campus Nord, Karlsruhe, Germany
| | - Wilko Thiele
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Institute for Toxicology and Genetics, KIT Campus Nord, Karlsruhe, Germany
| | - Melanie Rothley
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Institute for Toxicology and Genetics, KIT Campus Nord, Karlsruhe, Germany
| | - Diana Plaumann
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Institute for Toxicology and Genetics, KIT Campus Nord, Karlsruhe, Germany
| | - Claudia Korn
- German Cancer Research Center (DKFZ-ZMBH-Alliance), Heidelberg, Germany
| | - Olivier Armant
- Institute for Toxicology and Genetics, KIT Campus Nord, Karlsruhe, Germany
| | - Hellmut G. Augustin
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- German Cancer Research Center (DKFZ-ZMBH-Alliance), Heidelberg, Germany
| | - Jonathan P. Sleeman
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
- Institute for Toxicology and Genetics, KIT Campus Nord, Karlsruhe, Germany
| |
Collapse
|
11
|
Braz CU, Taylor JF, Decker JE, Bresolin T, Espigolan R, Garcia DA, Gordo DGM, Magalhães AFB, de Albuquerque LG, de Oliveira HN. Polymorphism analysis in genes associated with meat tenderness in Nelore cattle. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
12
|
Xu X, Mishra B, Qin N, Sun X, Zhang S, Yang J, Xu R. Differential Transcriptome Analysis of Early Postnatal Developing Longissimus Dorsi Muscle from Two Pig Breeds Characterized in Divergent Myofiber Traits and Fatness. Anim Biotechnol 2018; 30:63-74. [PMID: 29471750 DOI: 10.1080/10495398.2018.1437045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Meat quality traits (MQTs) are very important in the porcine industry, which are mainly determined by skeletal muscle fiber composition, extra-muscular and/or intramuscular fat content. To identify the differentially expressed candidate genes affecting the meat quality traits, first we compared the MQTs and skeletal muscle fiber characteristics in the longissimus dorsi muscle (LDM) of the Northeast Min pig (NM) and the Changbaishan wild boar (CW) with their body weight approaching 90 kg. The significant divergences in the skeletal muscle fiber phenotypes and fatness traits between the two porcine breeds established an ideal model system for further identifying potential key functional genes that dominated MQTs. Further, a transcriptome profile analysis was performed using the Illumina sequencing method in early postnatal developing LDM from the two breeds at the ages of 42 days. Comparative analysis between these two cDNA libraries showed that there were 17,653 and 22,049 unambiguous tag-mapped sense transcripts detected from NM and CW, respectively. 4522 differentially expressed genes (DEGs) were revealed between the two tissue samples, of them, 4176 genes were found as having been upregulated and 346 genes were identified as having been downregulated in the NM library. By pathway enrichment analysis, a set of significantly enriched pathways were identified for the DEGs, which are potentially involved in myofiber development, differentiation and growth, lipogenesis and lipolysis in porcine skeletal muscle. The expression levels of 30 out of the DEGs were validated by real-time quantitative reverse transcriptase PCR (qRT-PCR) and the observed result was consistent noticeably with the Illumina transcriptome profiles. The findings from this study can contribute to future investigations of skeletal muscle growth and development mechanism and to establishing molecular approaches to improve meat quality traits in pig breeding.
Collapse
Affiliation(s)
- Xiaoxing Xu
- a Department of Human Nutrition, Food, and Animal Sciences , University of Hawaii at Manoa , Honolulu , HI , USA
| | - Birendra Mishra
- a Department of Human Nutrition, Food, and Animal Sciences , University of Hawaii at Manoa , Honolulu , HI , USA
| | - Ning Qin
- b College of Animal Science and Technology , Jilin Agricultural University , Changchun , China
| | - Xue Sun
- b College of Animal Science and Technology , Jilin Agricultural University , Changchun , China
| | - Shumin Zhang
- c Institute of Pig Science , Academy of Agricultural Sciences of Jilin Province , Gongzhuling , China
| | - Jinzeng Yang
- a Department of Human Nutrition, Food, and Animal Sciences , University of Hawaii at Manoa , Honolulu , HI , USA
| | - Rifu Xu
- b College of Animal Science and Technology , Jilin Agricultural University , Changchun , China
| |
Collapse
|
13
|
Sato S, Uemoto Y, Kikuchi T, Egawa S, Kohira K, Saito T, Sakuma H, Miyashita S, Arata S, Kojima T, Suzuki K. SNP- and haplotype-based genome-wide association studies for growth, carcass, and meat quality traits in a Duroc multigenerational population. BMC Genet 2016; 17:60. [PMID: 27094516 PMCID: PMC4837538 DOI: 10.1186/s12863-016-0368-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/15/2016] [Indexed: 12/21/2022] Open
Abstract
Background The aim of the present study was to compare the power of single nucleotide polymorphism (SNP)-based genome-wide association study (GWAS) and haplotype-based GWAS for quantitative trait loci (QTL) detection, and to detect novel candidate genes affecting economically important traits in a purebred Duroc population comprising seven-generation pedigree. First, we performed a simulation analysis using real genotype data of this population to compare the power (based on the null hypothesis) of the two methods. We then performed GWAS using both methods and real phenotype data comprising 52 traits, which included growth, carcass, and meat quality traits. Results In total, 836 animals were genotyped using the Illumina PorcineSNP60 BeadChip and 14 customized SNPs from regions of known candidate genes related to the traits of interest. The power of SNP-based GWAS was greater than that of haplotype-based GWAS in a simulation analysis. In real data analysis, a larger number of significant regions was obtained by SNP-based GWAS than by haplotype-based GWAS. For SNP-based GWAS, 23 genome-wide significant SNP regions were detected for 17 traits, and 120 genome-wide suggestive SNP regions were detected for 27 traits. For haplotype-based GWAS, 6 genome-wide significant SNP regions were detected for four traits, and 11 genome-wide suggestive SNP regions were detected for eight traits. All genome-wide significant SNP regions detected by haplotype-based GWAS were located in regions also detected by SNP-based GWAS. Four regions detected by SNP-based GWAS were significantly associated with multiple traits: on Sus scrofa chromosome (SSC) 1 at 304 Mb; and on SSC7 at 35–39 Mb, 41–42 Mb, and 103 Mb. The vertnin gene (VRTN) in particular, was located on SSC7 at 103 Mb and was significantly associated with vertebrae number and carcass lengths. Mapped QTL regions contain some candidate genes involved in skeletal formation (FUBP3; far upstream element binding protein 3) and fat deposition (METTL3; methyltransferase like 3). Conclusion Our results show that a multigenerational pig population is useful for detecting QTL, which are typically segregated in a purebred population. In addition, a novel significant region could be detected by SNP-based GWAS as opposed to haplotype-based GWAS. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0368-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shuji Sato
- National Livestock Breeding Center, Nishigo, Fukushima, 961-8511, Japan.
| | - Yoshinobu Uemoto
- National Livestock Breeding Center, Nishigo, Fukushima, 961-8511, Japan
| | - Takashi Kikuchi
- National Livestock Breeding Center, Nishigo, Fukushima, 961-8511, Japan
| | - Sachiko Egawa
- Miyazaki Branch of National Livestock Breeding Center, Kobayashi, Miyazaki, 886-0004, Japan
| | - Kimiko Kohira
- National Livestock Breeding Center, Nishigo, Fukushima, 961-8511, Japan
| | - Tomomi Saito
- National Livestock Breeding Center, Nishigo, Fukushima, 961-8511, Japan
| | - Hironori Sakuma
- National Livestock Breeding Center, Nishigo, Fukushima, 961-8511, Japan
| | - Satoshi Miyashita
- Miyazaki Branch of National Livestock Breeding Center, Kobayashi, Miyazaki, 886-0004, Japan
| | - Shinji Arata
- Miyazaki Branch of National Livestock Breeding Center, Kobayashi, Miyazaki, 886-0004, Japan
| | - Takatoshi Kojima
- National Livestock Breeding Center, Nishigo, Fukushima, 961-8511, Japan
| | - Keiichi Suzuki
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, 981-8555, Japan
| |
Collapse
|
14
|
ASAP1 mediates the invasive phenotype of human laryngeal squamous cell carcinoma to affect survival prognosis. Oncol Rep 2014; 31:2676-82. [PMID: 24788532 DOI: 10.3892/or.2014.3150] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/24/2014] [Indexed: 11/05/2022] Open
Abstract
ASAP1 helps regulate cellular structures such as actin cytoskeletal remodeling and focal adhesions that have a pivotal function in tumor progression. Overexpression of ASAP1 has proven to be a malignant indicator for a variety of tumors. To further determine the potential involvement of ASAP1 in laryngeal squamous cell carcinoma (LSCC), we evaluated the expression levels of ASAP1 by quantitative real-time reverse-transcriptase polymerase chain reaction (qRT-PCR) and immunohistochemistry in tissue samples of 64 LSCC patients. We then analyzed and correlated the results with clinicopathological features. Furthermore, we used small interfering RNA (siRNA) to inhibit ASAP1 expression in vitro. The potential function of ASAP1 in invasiveness was evaluated in the Hep-2 LSCC cell line. Kaplan-Meier method was utilized to determine the association of ASAP1 expression with survival of patients. We showed that ASAP1 was upregulated in primary LSCC tumors and was correlated with lymph node metastasis and clinical tumor stage. Similarly, higher levels of ASAP1 were detected in the Hep-2 cell line compared to the 16 human bronchial epithelial (16HBE) cell line. ASAP1 expression was downregulated by lentiviral vector transfection containing siRNA in vitro. The invasive potential of these cells was found to be significantly suppressed, while expression levels of Rac1 and Cdc42 positively correlated with the inhibition of ASAP1 expression. In Kaplan-Meier overall survival curves, higher ASAP1 mRNA levels were found to be associated with a shorter progression-free survival trend. Based on these results, ASAP1 appears to contribute to the malignant mechanism of LSCC and may represent a significant prognostic marker for LSCC patients.
Collapse
|
15
|
Tizioto P, Meirelles S, Veneroni G, Tullio R, Rosa A, Alencar M, Medeiros S, Siqueira F, Feijó G, Silva L, Torres R, Regitano L. A SNP in ASAP1 gene is associated with meat quality and production traits in Nelore breed. Meat Sci 2012; 92:855-7. [DOI: 10.1016/j.meatsci.2012.05.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 03/16/2012] [Accepted: 05/18/2012] [Indexed: 11/24/2022]
|
16
|
Veneroni GB, Meirelles SL, Grossi DA, Gasparin G, Ibelli AMG, Tizioto PC, Oliveira HN, Alencar MM, Regitano LCA. Prospecting candidate SNPs for backfat in Canchim beef cattle. GENETICS AND MOLECULAR RESEARCH 2010; 9:1997-2003. [PMID: 20957603 DOI: 10.4238/vol9-4gmr788] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Canchim is a composite cattle breed developed in Brazil for beef production. One of the breeding objectives is to increase fat deposition. QTLs for fat thickness and/or marbling have been reported on BTA4 and BTA14. The IGFBP3 and DDEF1 genes, mapped to BTA4 and BTA14, respectively, affect adipogenesis. We looked for SNPs in the IGFBP3 and DDEF1 genes that could be associated with backfat thickness in Canchim beef cattle. For SNP identification, sires with the highest accuracy were ranked according to expected breeding value for fat thickness; the 12 extremes (six sires with the highest and six with the lowest expected breeding value for the trait) were chosen. Six regions of the IGFBP3 and 14 regions of the DDEF1 were sequenced using the Sanger method. Nine SNPs were identified in IGFBP3 and 76 in the DDEF1. After an initial analysis, two SNPs were selected to be genotyped for the whole population; these were DDEF1g.279401A>G and IGFBP3c.4394T>C(Trp>Arg). We found a significant effect (P ≤ 0.05) of allele substitution on backfat thickness; however, the IGFBP3 SNP did not significantly affect this trait.
Collapse
Affiliation(s)
- G B Veneroni
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos, SP, Brasil
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The Arf (ADP-ribosylation factor) GAPs (GTPase-activating proteins) are a family of proteins with a common catalytic domain that induces hydrolysis of GTP bound to Arf GTP-binding proteins. At least three groups of multidomain Arf GAPs affect the actin cytoskeleton and cellular activities, such as migration and movement, that depend on the cytoskeleton. One role of the Arf GAPs is to regulate membrane remodelling that accompanies actin polymerization. Regulation of membrane remodelling is mediated in part by the regulation of Arf proteins. However, Arf GAPs also regulate actin independently of effects on membranes or Arf. These functions include acting as upstream regulators of Rho family proteins and providing a scaffold for Rho effectors and exchange factors. With multiple functional elements, the Arf GAPs could integrate signals and biochemical activities that result in co-ordinated changes in actin and membranes necessary for a wide range of cellular functions.
Collapse
Affiliation(s)
- Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
18
|
Ha VL, Luo R, Nie Z, Randazzo PA. Contribution of AZAP-Type Arf GAPs to cancer cell migration and invasion. Adv Cancer Res 2008; 101:1-28. [PMID: 19055940 PMCID: PMC7249260 DOI: 10.1016/s0065-230x(08)00401-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Arf GAPs are a family of proteins with a common catalytic domain that induces hydrolysis of GTP bound to the small GTP-binding protein Arf. The proteins are otherwise structurally diverse. Several subtypes of Arf GAPs have been found to be targets of oncogenes and to control cell proliferation and cell migration. The latter effects are thought to be mediated by coordinating changes in actin remodeling and membrane traffic. In this chapter, we discuss Arf GAPs that have been linked to oncogenesis and the molecular mechanisms underlying the effects of these proteins in cancer cells. We also discuss the enzymology of the Arf GAPs related to possible targeted inhibition of specific subtypes of Arf GAPs.
Collapse
Affiliation(s)
- Vi Luan Ha
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
19
|
Abstract
Small G proteins play a central role in the organization of the secretory and endocytic pathways. The majority of such small G proteins are members of the Rab family, which are anchored to the bilayer by C-terminal prenyl groups. However, the recruitment of some effectors, including vesicle coat proteins, is mediated by a second class of small G proteins that is unique in having an N-terminal amphipathic helix that becomes available for membrane insertion upon GTP binding. Sar1, Arf1, and Arf6 are the best-characterized members of this ADP-ribosylation factor (Arf) family. In addition, all eukaryotes contain additional distantly related G proteins, often called Arf like, or Arls. The complete Arf family in humans has 29 members. The roles of these related G proteins are poorly understood, but recent work has shown that some are involved in membrane traffic or organizing the cytoskeleton. Here we review what is known about all the members of the Arf family, along with the known regulatory molecules that convert them between GDP- and GTP-bound states.
Collapse
|
20
|
Bharti S, Inoue H, Bharti K, Hirsch DS, Nie Z, Yoon HY, Artym V, Yamada KM, Mueller SC, Barr VA, Randazzo PA. Src-dependent phosphorylation of ASAP1 regulates podosomes. Mol Cell Biol 2007; 27:8271-83. [PMID: 17893324 PMCID: PMC2169185 DOI: 10.1128/mcb.01781-06] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Invadopodia are Src-induced cellular structures that are thought to mediate tumor invasion. ASAP1, an Arf GTPase-activating protein (GAP) containing Src homology 3 (SH3) and Bin, amphiphysin, and RVS161/167 (BAR) domains, is a substrate of Src that controls invadopodia. We have examined the structural requirements for ASAP1-dependent formation of invadopodia and related structures in NIH 3T3 fibroblasts called podosomes. We found that both predominant splice variants of ASAP1 (ASAP1a and ASAP1b) associated with invadopodia and podosomes. Podosomes were highly dynamic, with rapid turnover of both ASAP1 and actin. Reduction of ASAP1 levels by small interfering RNA blocked formation of invadopodia and podosomes. Podosomes were formed in NIH 3T3 fibroblasts in which endogenous ASAP1 was replaced with either recombinant ASAP1a or ASAP1b. ASAP1 mutants that lacked the Src binding site or GAP activity functioned as well as wild-type ASAP1 in the formation of podosomes. Recombinant ASAP1 lacking the BAR domain, the SH3 domain, or the Src phosphorylation site did not support podosome formation. Based on these results, we conclude that ASAP1 is a critical target of tyrosine kinase signaling involved in the regulation of podosomes and invadopodia and speculate that ASAP1 may function as a coincidence detector of simultaneous protein association through the ASAP1 SH3 domain and phosphorylation by Src.
Collapse
Affiliation(s)
- Sanita Bharti
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hashimoto S, Hashimoto A, Yamada A, Onodera Y, Sabe H. Assays and properties of the ArfGAPs, AMAP1 and AMAP2, in Arf6 function. Methods Enzymol 2006; 404:216-31. [PMID: 16413272 DOI: 10.1016/s0076-6879(05)04021-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The GTPase-activating protein (GAP) domain for Arfs primarily consists of a zinc-finger structure, which is not present in known GAPs for the other Ras-superfamily GTPases. More than 20 genes have been found to encode proteins bearing the ArfGAP domain in the human genome: a number that is much larger than that of the Arf isoforms. Several Arf isoforms, such as Arf1 and Arf6, indeed have been shown to each employ multiple different ArfGAPs for their regulation and function. We have found that two ArfGAPs, namely AMAP1 and AMAP2, exhibit a novel biochemical property of directly and selectively binding to GTP-Arf6 without immediate GAPing activity, while they were previously shown to exhibit efficient catalytic GAPing activities to Arf isoforms except Arf6 in vitro. Such property of AMAPs appears to be important for AMAPs-mediated recruitment of auxiliary molecules, including paxillin, cortactin, amphiphysin, and intersectin, to sites of Arf6 activation. AMAPs thus appear to act as "effectors" rather than simple GAPs in some aspects of Arf6 function. This article presents methods and protocols developed for the functional characterization of AMAPs in Arf6 function. These methods may be applied to other types of ArfGAPs to further clarify the cellular functions of ArfGAPs as well as Arfs.
Collapse
|
22
|
Abstract
Centaurin beta4 proteins are products of the DDEF1 (development and differentiation-enhancing factor 1) locus on human chromosome 8q24.1-24.2. Recent reports have indicated that this region and its products are amplified during development of several human cancers. Centaurins are GAPs (GTPase-activating proteins) that, together with GEFs (guanine nucleotide-exchange factors), regulate cyclic activation of Arfs (ADP-ribosylation factors), members of the Ras GTPase superfamily. Centaurin beta4 proteins associate with a variety of cellular signalling components implicated in control of growth, survival and movement and may act to direct assembly and/or disassembly of molecular complexes in concert with Arf, lipid and protein phosphorylation signalling pathways.
Collapse
|
23
|
Ehlers JP, Worley L, Onken MD, Harbour JW. DDEF1 is located in an amplified region of chromosome 8q and is overexpressed in uveal melanoma. Clin Cancer Res 2005; 11:3609-13. [PMID: 15897555 DOI: 10.1158/1078-0432.ccr-04-1941] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The molecular pathogenesis of uveal melanoma is poorly understood but is usually accompanied by amplification of chromosome 8q, suggesting the activation of one or more oncogenes. We recently identified a gene expression profile that distinguishes low-grade from high-grade melanomas. In this profile, a cluster of genes at chromosome 8q was overexpressed in high-grade tumors, providing an opportunity to search for potential oncogenes in this region. EXPERIMENTAL DESIGN Gene expression microarray analysis was done on 25 primary uveal melanomas. Microarray comparative genomic hybridization (CGH), quantitative PCR, and immunohistochemistry were done on a subset of these tumors. Cell motility was measured using a wound-healing assay. RESULTS In melanomas analyzed for microarray gene expression and CGH, gain of chromosome 8q correlated most strongly with expression of DDEF1, a gene located at 8q24. In contrast, the nearby MYC oncogene exhibited no significant change in expression. Confirming the microarray findings, DDEF1 mRNA levels and protein expression were significantly higher in high-grade melanomas. Furthermore, ectopic expression of DDEF1 in low-grade melanoma cells resulted in a significant increase in cell motility, a feature of high-grade metastasizing cells. CONCLUSIONS These findings suggest that DDEF1 overexpression may be a pathogenetically relevant consequence of chromosome 8q amplification, which commonly occurs in high-grade uveal melanomas. We conclude that DDEF1 may act as an oncogene in this cancer, and it may be a useful diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Justis P Ehlers
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
24
|
I STT, Nie Z, Stewart A, Najdovska M, Hall NE, He H, Randazzo PA, Lock P. ARAP3 is transiently tyrosine phosphorylated in cells attaching to fibronectin and inhibits cell spreading in a RhoGAP-dependent manner. J Cell Sci 2004; 117:6071-84. [PMID: 15546919 DOI: 10.1242/jcs.01526] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ARAP3 is a GTPase activating protein (GAP) for Rho and Arf GTPases that is implicated in phosphoinositide 3-kinase (PI 3-kinase) signalling pathways controlling lamellipodia formation and actin stress fibre assembly. We have identified ARAP3 as a phosphorylated target of protein tyrosine kinases. In cells, ARAP3 was tyrosine phosphorylated when co-expressed with Src-family kinases (SFKs), upon stimulation with growth factors and during adhesion to the extracellular matrix (ECM) substrate fibronectin. Adhesion-induced phosphorylation of ARAP3 was suppressed by selective inhibitors of Src-family kinases and PI 3-kinase and by a Src dominant interfering mutant. Inducible expression of ARAP3 in HEK293 epithelial cells resulted in increased cell rounding, membrane process formation and cell clustering on ECM substrates. In contrast, ARAP3 dramatically slowed the kinetics of cell spreading on fibronectin but had no effect on cell adhesion. These effects of ARAP3 required a functional Rho GAP domain and were associated with reduced cellular levels of active RhoA and Rac1 but did not require the sterile alpha motif (SAM) or Arf GAP domains. Mutation of two phosphorylation sites, Y1399 and Y1404, enhanced some ARAP3 activities, suggesting that ARAP3 may be negatively regulated by phosphorylation on these tyrosine residues. These results implicate ARAP3 in integrin-mediated tyrosine kinase signalling pathways controlling Rho GTPases and cell spreading.
Collapse
Affiliation(s)
- Stacey T T I
- Department of Surgery, University of Melbourne, Level 5 Clinical Sciences Building, Royal Melbourne Hospital, VIC 3050, Australia
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Randazzo PA, Hirsch DS. Arf GAPs: multifunctional proteins that regulate membrane traffic and actin remodelling. Cell Signal 2004; 16:401-13. [PMID: 14709330 DOI: 10.1016/j.cellsig.2003.09.012] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The ADP-ribosylation factor (Arf) Arf GTPase-activating proteins (GAPs) are a family of proteins that induce hydrolysis of GTP bound to Arf. A conserved domain containing a zinc finger motif mediates catalysis. The substrate, Arf.GTP, affects membrane trafficking and actin remodelling. Consistent with activity as an Arf regulator, the Arf GAPs affect both of these pathways. However, the Arf GAPs are likely to have Arf-independent activities that contribute to their cellular functions. Structures of the Arf GAPs are diverse containing catalytic, protein-protein interaction and lipid interaction domains in addition to the Arf GAP domain. Some Arf GAPs have been identified and characterized on the basis of activities other than Arf GAP. Here, we describe the Arf GAP family, enzymology of some members of the Arf GAP family and known functions of the proteins. The results discussed illustrate roles for both Arf-dependent and -independent activities in the regulation of cellular architecture.
Collapse
Affiliation(s)
- Paul A Randazzo
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Building. 37 Room 4118, Bethesda, MD 20892, USA.
| | | |
Collapse
|
26
|
Oda A, Wada I, Miura K, Okawa K, Kadoya T, Kato T, Nishihara H, Maeda M, Tanaka S, Nagashima K, Nishitani C, Matsuno K, Ishino M, Machesky LM, Fujita H, Randazzo P. CrkL directs ASAP1 to peripheral focal adhesions. J Biol Chem 2003; 278:6456-60. [PMID: 12522101 DOI: 10.1074/jbc.m210817200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Searching for proteins in platelets that can interact with the N-terminal SH3 domain of CrkL (using a combination of a pull-down assay followed by mass spectrometry), we have found that human platelets express an ADP-ribosylation factor (Arf)-specific GTPase-activating protein (GAP), ASAP1, as a CrkL-binding protein. In spreading platelets, most endogenous ASAP1 is localized at peripheral focal adhesions. To determine the physiologic significance of the CrkL-ASAP1 association, we overexpressed CrkL, ASAP1, or both in combination in COS7 cells. Unlike endogenous ASAP1 in platelets, overexpressed ASAP1 showed diffuse cytoplasmic distribution. However, when co-expressed with wild-type CrkL, both endogenous and expressed ASAP1 accumulated at CrkL-induced focal adhesions. An SH2-mutated CrkL, which cannot localize at focal adhesions, failed to recruit ASAP1 into focal adhesions. Thus, CrkL appears to be a lynchpin between ASAP1 and peripheral focal adhesions.
Collapse
Affiliation(s)
- Atsushi Oda
- Laboratory of Environmental Biology, Department of Preventive Medicine, Hokkaido University School of Medicine, Sapporo 060-8638, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Scott MP, Zappacosta F, Kim EY, Annan RS, Miller WT. Identification of novel SH3 domain ligands for the Src family kinase Hck. Wiskott-Aldrich syndrome protein (WASP), WASP-interacting protein (WIP), and ELMO1. J Biol Chem 2002; 277:28238-46. [PMID: 12029088 DOI: 10.1074/jbc.m202783200] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The importance of the SH3 domain of Hck in kinase regulation, substrate phosphorylation, and ligand binding has been established. However, few in vivo ligands are known for the SH3 domain of Hck. In this study, we used mass spectrometry to identify approximately 25 potential binding partners for the SH3 domain of Hck from the monocyte cell line U937. Two major interacting proteins were the actin binding proteins Wiskott-Aldrich syndrome protein (WASP) and WASP-interacting protein (WIP). We also focused on a novel interaction between Hck and ELMO1, an 84-kDa protein that was recently identified as the mammalian ortholog of the Caenorhabditis elegans gene, ced-12. In mammalian cells, ELMO1 interacts with Dock180 as a component of the CrkII/Dock180/Rac pathway responsible for phagocytosis and cell migration. Using purified proteins, we confirmed that WASP-interacting protein and ELMO1 interact directly with the SH3 domain of Hck. We also show that Hck and ELMO1 interact in intact cells and that ELMO1 is heavily tyrosine-phosphorylated in cells that co-express Hck, suggesting that it is a substrate of Hck. The binding of ELMO1 to Hck is specifically dependent on the interaction of a polyproline motif with the SH3 domain of Hck. Our results suggest that these proteins may be novel activators/effectors of Hck.
Collapse
Affiliation(s)
- Margaret Porter Scott
- Department of Physiology and Biophysics, School of Medicine, State University of New York, Stony Brook, New York 11794-8661, USA
| | | | | | | | | |
Collapse
|
28
|
Furman C, Short SM, Subramanian RR, Zetter BR, Roberts TM. DEF-1/ASAP1 is a GTPase-activating protein (GAP) for ARF1 that enhances cell motility through a GAP-dependent mechanism. J Biol Chem 2002; 277:7962-9. [PMID: 11773070 DOI: 10.1074/jbc.m109149200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DEF-1/ASAP1 is an ADP-ribosylation factor GTPase-activating protein (ARF GAP) that localizes to focal adhesions and is involved in cytoskeletal regulation. In this paper, we use a cell-based ARF GAP assay to demonstrate that DEF-1 functions as a GAP for ARF1 and not ARF6 in vivo. This degree of substrate preference was unique to DEF-1, as other ARF GAP proteins, ACAP1, ACAP2, and ARFGAP1, were able to function on both ARF1 and ARF6. Since transient overexpression of DEF-1 has been shown to interfere with focal adhesion formation and platelet-derived growth factor-induced membrane ruffling, we investigated whether NIH 3T3 cells stably expressing DEF-1 have altered cell motility. Here we report that ectopic DEF-1 enhances cell migration toward PDGF as well as IGF-1. This chemotactic effect appears to result from a general increase in cell motility, as DEF-1-expressing cells also exhibit enhanced levels of basal and chemokinetic motility. The increase in cell motility is dependent on DEF-1 GAP activity, since a DEF-1 mutant lacking the GAP domain failed to stimulate motility. This suggests that DEF-1 alters cell motility through the deactivation of ARF1. In contrast, the inhibition of cell spreading by DEF-1 was not dependent on GAP activity, indicating that spreading and motility are altered by DEF-1 through different pathways.
Collapse
Affiliation(s)
- Craig Furman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
Several new families of ARF GTPase activating proteins (ARF GAPs) have been described recently that associate with paxillin and other cytoskeletal and signaling proteins. Important insights have been gained regarding their subcellular distribution, enzymatic specificity and protein scaffold function. Evidence suggests an important role for ARF GAPs in mediating changes in the cell's actin cytoskeleton in response to adhesion and growth factor stimulation.
Collapse
Affiliation(s)
- C E Turner
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, New York 13210, USA.
| | | | | |
Collapse
|
30
|
Premont RT, Vitale N. Purification and characterization of GIT family of ADP-ribosylation factor (ARF) GTPase-activating proteins. Methods Enzymol 2001; 329:335-43. [PMID: 11210553 DOI: 10.1016/s0076-6879(01)29095-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Affiliation(s)
- R T Premont
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
31
|
Abstract
Small GTP-binding proteins (G proteins) exist in eukaryotes from yeast to human and constitute a superfamily consisting of more than 100 members. This superfamily is structurally classified into at least five families: the Ras, Rho, Rab, Sar1/Arf, and Ran families. They regulate a wide variety of cell functions as biological timers (biotimers) that initiate and terminate specific cell functions and determine the periods of time for the continuation of the specific cell functions. They furthermore play key roles in not only temporal but also spatial determination of specific cell functions. The Ras family regulates gene expression, the Rho family regulates cytoskeletal reorganization and gene expression, the Rab and Sar1/Arf families regulate vesicle trafficking, and the Ran family regulates nucleocytoplasmic transport and microtubule organization. Many upstream regulators and downstream effectors of small G proteins have been isolated, and their modes of activation and action have gradually been elucidated. Cascades and cross-talks of small G proteins have also been clarified. In this review, functions of small G proteins and their modes of activation and action are described.
Collapse
Affiliation(s)
- Y Takai
- Department of Molecular Biology, Osaka University Graduate School of Medicine/Faculty of Medicine, Suita, Japan.
| | | | | |
Collapse
|
32
|
Jackson TR, Brown FD, Nie Z, Miura K, Foroni L, Sun J, Hsu VW, Donaldson JG, Randazzo PA. ACAPs are arf6 GTPase-activating proteins that function in the cell periphery. J Cell Biol 2000; 151:627-38. [PMID: 11062263 PMCID: PMC2185579 DOI: 10.1083/jcb.151.3.627] [Citation(s) in RCA: 155] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2000] [Accepted: 09/18/2000] [Indexed: 11/22/2022] Open
Abstract
The GTP-binding protein ADP-ribosylation factor 6 (Arf6) regulates endosomal membrane trafficking and the actin cytoskeleton in the cell periphery. GTPase-activating proteins (GAPs) are critical regulators of Arf function, controlling the return of Arf to the inactive GDP-bound state. Here, we report the identification and characterization of two Arf6 GAPs, ACAP1 and ACAP2. Together with two previously described Arf GAPs, ASAP1 and PAP, they can be grouped into a protein family defined by several common structural motifs including coiled coil, pleckstrin homology, Arf GAP, and three complete ankyrin-repeat domains. All contain phosphoinositide-dependent GAP activity. ACAP1 and ACAP2 are widely expressed and occur together in the various cultured cell lines we examined. Similar to ASAP1, ACAP1 and ACAP2 were recruited to and, when overexpressed, inhibited the formation of platelet-derived growth factor (PDGF)-induced dorsal membrane ruffles in NIH 3T3 fibroblasts. However, in contrast with ASAP1, ACAP1 and ACAP2 functioned as Arf6 GAPs. In vitro, ACAP1 and ACAP2 preferred Arf6 as a substrate, rather than Arf1 and Arf5, more so than did ASAP1. In HeLa cells, overexpression of either ACAP blocked the formation of Arf6-dependent protrusions. In addition, ACAP1 and ACAP2 were recruited to peripheral, tubular membranes, where activation of Arf6 occurs to allow membrane recycling back to the plasma membrane. ASAP1 did not inhibit Arf6-dependent protrusions and was not recruited by Arf6 to tubular membranes. The additional effects of ASAP1 on PDGF-induced ruffling in fibroblasts suggest that multiple Arf GAPs function coordinately in the cell periphery.
Collapse
Affiliation(s)
- T R Jackson
- Laboratory of Cellular Oncology, Division of Basic Sciences, National Cancer Institute, Bethesda, Maryland 20892-4255, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Jackson TR, Kearns BG, Theibert AB. Cytohesins and centaurins: mediators of PI 3-kinase-regulated Arf signaling. Trends Biochem Sci 2000; 25:489-95. [PMID: 11050434 DOI: 10.1016/s0968-0004(00)01644-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Receptor-activated phosphoinositide (PI) 3-kinases produce PtdIns(3, 4,5)P(3) and its metabolite PtdIns(3,4)P(2) that function as second messengers in membrane recruitment and activation of target proteins. The cytohesin and centaurin protein families are potential targets for PtdIns(3,4,5)P(3) that also regulate and interact with Arf GTPases. Consequently, these families are poised to transduce PI 3-kinase activation into coordinated control of Arf-dependent pathways. Proposed downstream events in PI 3-kinase-regulated Arf cascades include modulation of vesicular trafficking and the actin cytoskeleton.
Collapse
Affiliation(s)
- T R Jackson
- Dept of Haematology, Royal Free and University College Medical School, Royal Free Campus, Rowland Hill St, NW3 2PF, London, UK.
| | | | | |
Collapse
|
34
|
Premont RT, Claing A, Vitale N, Perry SJ, Lefkowitz RJ. The GIT family of ADP-ribosylation factor GTPase-activating proteins. Functional diversity of GIT2 through alternative splicing. J Biol Chem 2000; 275:22373-80. [PMID: 10896954 DOI: 10.1074/jbc.275.29.22373] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We recently characterized a novel protein, GIT1, that interacts with G protein-coupled receptor kinases and possesses ADP-ribosylation factor (ARF) GTPase-activating protein activity. A second ubiquitously expressed member of the GIT protein family, GIT2, has been identified in data base searches. GIT2 undergoes extensive alternative splicing and exists in at least 10 and potentially as many as 33 distinct forms. The longest form of GIT2 is colinear with GIT1 and shares the same domain structure, whereas one major splice variant prominent in immune tissues completely lacks the carboxyl-terminal domain. The other 32 potential variants arise from the independent alternative splicing of five internal regions in the center of the molecule but share both the amino-terminal ARF GTPase-activating protein domain and carboxyl-terminal domain. Both the long and short carboxyl-terminal variants of GIT2 are active as GTPase-activating proteins for ARF1, and both also interact with G protein-coupled receptor kinase 2 and with p21-activated kinase-interacting exchange factors complexed with p21-activated kinase but not with paxillin. Cellular overexpression of the longest variant of GIT2 leads to inhibition of beta(2)-adrenergic receptor sequestration, whereas the shortest splice variant appears inactive. Although GIT2 shares many properties with GIT1, it also exhibits both structural and functional diversity due to tissue-specific alternative splicing.
Collapse
Affiliation(s)
- R T Premont
- Departments of Medicine and Biochemistry, Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | |
Collapse
|
35
|
Vitale N, Patton WA, Moss J, Vaughan M, Lefkowitz RJ, Premont RT. GIT proteins, A novel family of phosphatidylinositol 3,4, 5-trisphosphate-stimulated GTPase-activating proteins for ARF6. J Biol Chem 2000; 275:13901-6. [PMID: 10788515 DOI: 10.1074/jbc.275.18.13901] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ADP-ribosylation factor (ARF) proteins are key players in numerous vesicular trafficking events ranging from the formation and fusion of vesicles in the Golgi apparatus to exocytosis and endocytosis. To complete their GTPase cycle, ARFs require a guanine nucleotide-exchange protein to catalyze replacement of GDP by GTP and a GTPase-activating protein (GAP) to accelerate hydrolysis of bound GTP. Recently numerous guanine nucleotide-exchange proteins and GAP proteins have been identified and partially characterized. Every ARF GAP protein identified to date contains a characteristic zinc finger motif. GIT1 and GIT2, two members of a new family of G protein-coupled receptor kinase-interacting proteins, also contain a putative zinc finger motif and display ARF GAP activity. Truncation of the amino-terminal region containing the zinc finger motif prevented GAP activity of GIT1. One zinc molecule was found associated per molecule of purified recombinant ARF-GAP1, GIT1, and GIT2 proteins, suggesting the zinc finger motifs of ARF GAPs are functional and should play an important role in their GAP activity. Unlike ARF-GAP1, GIT1 and GIT2 stimulate hydrolysis of GTP bound to ARF6. Accordingly we found that the phospholipid dependence of the GAP activity of ARF-GAP1 and GIT proteins was quite different, as the GIT proteins are stimulated by phosphatidylinositol 3,4, 5-trisphosphate whereas ARF-GAP1 is stimulated by phosphatidylinositol 4,5-bisphosphate and diacylglycerol. These results suggest that although the mechanism of GTP hydrolysis is probably very similar in these two families of ARF GAPs, GIT proteins might specifically regulate the activity of ARF6 in cells in coordination with phosphatidylinositol 3-kinase signaling pathways.
Collapse
Affiliation(s)
- N Vitale
- INSERM U-338, Centre de Neurochimie, 5 rue Blaise Pascal, 67084 Strasbourg Cedex, France
| | | | | | | | | | | |
Collapse
|