1
|
Zhao R, Ning X, Lu H, Xu W, Ma J, Cheng J, Ma R. XTP8 Promotes Ovarian Cancer Progression by Activating AKT/AMPK/mTOR Pathway to Regulate EMT. Cell Biochem Biophys 2024; 82:945-957. [PMID: 38717641 PMCID: PMC11344704 DOI: 10.1007/s12013-024-01246-4] [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] [Accepted: 02/27/2024] [Indexed: 08/25/2024]
Abstract
Ovarian cancer (OC) ranks as the fifth leading cause of cancer-related death in women. The main contributors to the poor prognosis of ovarian cancer are the high rates of recurrence and metastasis. Studies have indicated a crucial role for hepatitis B virus X Ag-Transactivated Protein 8 (XTP8), a protein containing the DEP domain, in various cellular processes, including cell growth, movement, and differentiation, across several types of cancers. However, the role of XTP8 in ovarian cancer remains unclear. We observed elevated expression of XTP8 in ovarian cancer. Silencing XTP8 inhibited cell proliferation, promoted apoptosis, and yielded contrasting results in cells overexpressing XTP8. Furthermore, XTP8 facilitated ovarian cancer invasion and migration, triggering epithelial-mesenchymal transition (EMT). Mechanistically, XTP8 silencing led to reduced phosphorylation levels of AKT, increased p-AMPK levels, and decreased p-mTOR levels, while XTP8 overexpression exerted the opposite effects. Additionally, the activation of p-AMPK rescued the promoting effect of XTP8 on EMT in ovarian cancer cell lines, indicating that XTP8 acts as an oncogene by modulating the AKT/AMPK/mTOR pathway. Through transcriptome sequencing to identify downstream targets of XTP8, we found that XTP8 influences the expression of Caldesmon (CALD1) at both transcriptional and translational levels. CALD1 can be considered a downstream target of XTP8. The collaborative action of XTP8 and CALD1 activates the AKT/AMPK/mTOR pathway, regulating EMT to promote ovarian cancer progression. Inhibiting this signaling axis might represent a potential therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Ruixue Zhao
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Xin Ning
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Hongping Lu
- Hebei Utu Pharmaceutical Company Ltd, Shijiazhuang, Hebei Province, 052165, China
| | - Wei Xu
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Jiaxin Ma
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Jun Cheng
- Hebei Utu Pharmaceutical Company Ltd, Shijiazhuang, Hebei Province, 052165, China.
| | - Rong Ma
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
2
|
Fei H, Shi X, Li S, Li Y, Yin X, Wu Z, Wang W, Shi H, Li R. DEPDC1B enhances malignant phenotypes of multiple myeloma through upregulating CCNB1 and inhibiting p53 signaling pathway. Tissue Cell 2024; 86:102263. [PMID: 37979396 DOI: 10.1016/j.tice.2023.102263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/20/2023]
Abstract
The identification and investigation of key molecules involved in the pathogenesis of multiple myeloma (MM) hold paramount clinical significance. This study primarily focuses on elucidating the role of DEPDC1B within the context of MM. Our findings robustly affirm the abundant expression of DEPDC1B in MM tissues and cell lines. Notably, DEPDC1B depletion exerted inhibitory effects on MM cell proliferation and migration while concurrently facilitating apoptosis and G2 cell cycle arrest. These outcomes stand in stark contrast to the consequences of DEPDC1B overexpression. Furthermore, we identified CCNB1 as a putative downstream target, characterized by a co-expression pattern with DEPDC1B, mediating DEPDC1B's regulatory influence on MM. Additionally, our results suggest that DEPDC1B knockdown may activate the p53 pathway, thereby impeding MM progression. To corroborate these in vitro findings, we conducted in vivo experiments that further validate the regulatory role of DEPDC1B in MM and its interaction with CCNB1 and the p53 pathway. Collectively, our research underscores DEPDC1B as a potent promoter in the development of MM, representing a promising therapeutic target for MM treatment. This discovery bears significant implications for future investigations in this field.
Collapse
Affiliation(s)
- Hairong Fei
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xue Shi
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Saisai Li
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ying Li
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiangcong Yin
- Hematology Diagnosis Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zengjie Wu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wei Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hailei Shi
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ran Li
- Department of Medical Oncology, Affiliated Qingdao Central Hospital Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
3
|
Yang M, Zhang H, Gao S, Huang W. DEPDC1 and KIF4A synergistically inhibit the malignant biological behavior of osteosarcoma cells through Hippo signaling pathway. J Orthop Surg Res 2023; 18:145. [PMID: 36849972 PMCID: PMC9972622 DOI: 10.1186/s13018-023-03572-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/01/2023] [Indexed: 03/01/2023] Open
Abstract
The treatment of osteosarcoma (OS) is still mainly surgery combined with systematic chemotherapy, and gene therapy is expected to improve the survival rate of patients. This study aimed to explore the effect of DEP domain 1 protein (DEPDC1) and kinesin super-family protein 4A (KIF4A) in OS and understand its mechanism. Th expression of DEPDC1 and KIF4A in OS cells was detected by RT-PCR and western blot. The viability, proliferation, invasion and migration of OS cells and tube formation of human umbilical vein endothelial cells (HUVECs) after indicated treatment were in turn detected by CCK-8 assay, EdU staining, wound healing assay, transwell assay and tube formation assay. The interaction between DEPDC1 and KIF4A was predicted by STRING and confirmed by co-immunoprecipitation. The expression of epithelial-mesenchymal transition (EMT)-related proteins, tube formation-related proteins and Hippo signaling pathway proteins was detected by western blot. As a result, the expression of DEPDC1 and KIF4A was all increased in U2OS cells. Down-regulation of DEPDC1 suppressed the viability, proliferation, invasion and migration of U2OS cells and tube formation of HUVECs, accompanied by the increased expression of E-cadherin and decreased expression of N-cadherin, Vimentin and VEGF. DEPDC1 was confirmed to be interacted with KIF4A. Upregulation of KIF4A partially reversed the effect of DEPDC1 interference on the above biological behaviors of U2OS cells. Down-regulation of DEPDC1 promoted the expression of p-LATS1 and p-YAP in Hippo signaling pathway, which was reversed by upregulation of KIF4A. In conclusion, down-regulation of DEPDC1 inhibited the malignant biological behavior of OS cells through the activation of Hippo signaling pathway, which could be reversed by upregulation of KIF4A.
Collapse
Affiliation(s)
- Mingming Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016, People's Republic of China.
| | - Hang Zhang
- grid.452206.70000 0004 1758 417XDepartment of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016 People’s Republic of China
| | - Shichang Gao
- grid.452206.70000 0004 1758 417XDepartment of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016 People’s Republic of China
| | - Wei Huang
- grid.452206.70000 0004 1758 417XDepartment of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016 People’s Republic of China
| |
Collapse
|
4
|
Wang Y, Wu J, Luo W, Zhang H, Shi G, Shen Y, Zhu Y, Ma C, Dai B, Ye D, Zhu Y. ALPK2 acts as tumor promotor in development of bladder cancer through targeting DEPDC1A. Cell Death Dis 2021; 12:661. [PMID: 34210956 PMCID: PMC8249393 DOI: 10.1038/s41419-021-03947-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/08/2021] [Accepted: 06/16/2021] [Indexed: 12/30/2022]
Abstract
Bladder cancer is one of the most common malignant tumors in the urinary system. The development and improvement of treatment efficiency require the deepening of the understanding of its molecular mechanism. This study investigated the role of ALPK2, which is rarely studied in malignant tumors, in the development of bladder cancer. Our results showed the upregulation of ALPK2 in bladder cancer, and data mining of TCGA database showed the association between ALPK2 and pathological parameters of patients with bladder cancer. In vitro and in vivo experiments demonstrated that knockdown of ALPK2 could inhibit bladder cancer development through regulating cell proliferation, cell apoptosis, and cell migration. Additionally, DEPDC1A is identified as a potential downstream of ALPK2 with direct interaction, whose overexpression/downregulation can inhibit/promote the malignant behavioral of bladder cancer cells. Moreover, the overexpression of DEPDC1A can rescue the inhibitory effects of ALPK2 knockdown on bladder cancer. In conclusion, ALPK2 exerts a cancer-promoting role in the development of bladder cancer by regulating DEPDC1A, which may become a promising target to improve the treatment strategy of bladder cancer.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Jie Wu
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Wenjie Luo
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Hailiang Zhang
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Guohai Shi
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Yijun Shen
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Yao Zhu
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Chunguang Ma
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Bo Dai
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Dingwei Ye
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
| | - Yiping Zhu
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Say TE, Degnan SM. Molecular and behavioural evidence that interdependent photo - and chemosensory systems regulate larval settlement in a marine sponge. Mol Ecol 2019; 29:247-261. [PMID: 31791111 DOI: 10.1111/mec.15318] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/04/2019] [Accepted: 11/26/2019] [Indexed: 01/20/2023]
Abstract
Marine pelagic larvae use a hierarchy of environmental cues to identify a suitable benthic habitat on which to settle and metamorphose into the adult phase of the life cycle. Most larvae are induced to settle by biochemical cues and many species have long been known to preferentially settle in the dark. Combined, these data suggest that larval responses to light and biochemical cues may be linked, but this has yet to be explored at the molecular level. Here, we track the vertical position of larvae of the sponge Amphimedon queenslandica to show that they descend to the benthos at twilight, by which time they are competent to respond to biochemical cues, consistent with them naturally settling in the dark. We use larval settlement assays under three different light regimes, combined with transcriptomics on individual larvae, to identify candidate molecular pathways underlying larval settlement. We find that larvae do not settle in response to biochemical cues if maintained in constant light. Our transcriptome data suggest that constant light actively represses settlement via the sustained up-regulation of two putative inactivators of chemotransduction in constant light only. Our data suggest that photo- and chemosensory systems interact to regulate larval settlement via nitric oxide and cyclic guanosine monophosphate signalling in this sponge, which belongs to one of the earliest-branching animal phyla.
Collapse
Affiliation(s)
- Tahsha E Say
- School of Biological Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Sandie M Degnan
- School of Biological Sciences, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
6
|
Caron A, Briscoe DM, Richard D, Laplante M. DEPTOR at the Nexus of Cancer, Metabolism, and Immunity. Physiol Rev 2018; 98:1765-1803. [PMID: 29897294 DOI: 10.1152/physrev.00064.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
DEP domain-containing mechanistic target of rapamycin (mTOR)-interacting protein (DEPTOR) is an important modulator of mTOR, a kinase at the center of two important protein complexes named mTORC1 and mTORC2. These highly studied complexes play essential roles in regulating growth, metabolism, and immunity in response to mitogens, nutrients, and cytokines. Defects in mTOR signaling have been associated with the development of many diseases, including cancer and diabetes, and approaches aiming at modulating mTOR activity are envisioned as an attractive strategy to improve human health. DEPTOR interaction with mTOR represses its kinase activity and rewires the mTOR signaling pathway. Over the last years, several studies have revealed key roles for DEPTOR in numerous biological and pathological processes. Here, we provide the current state of the knowledge regarding the cellular and physiological functions of DEPTOR by focusing on its impact on the mTOR pathway and its role in promoting health and disease.
Collapse
Affiliation(s)
- Alexandre Caron
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| | - David M Briscoe
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| | - Denis Richard
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| | - Mathieu Laplante
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| |
Collapse
|
7
|
Moretti M, Wang L, Grognet P, Lanver D, Link H, Kahmann R. Three regulators of G protein signaling differentially affect mating, morphology and virulence in the smut fungusUstilago maydis. Mol Microbiol 2017; 105:901-921. [DOI: 10.1111/mmi.13745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Marino Moretti
- Department of Organismic Interactions; Max Planck Institute for Terrestrial Microbiology; Karl-von-Frisch-Strasse 10, Marburg D-35043 Germany
| | - Lei Wang
- Department of Organismic Interactions; Max Planck Institute for Terrestrial Microbiology; Karl-von-Frisch-Strasse 10, Marburg D-35043 Germany
| | - Pierre Grognet
- Department of Organismic Interactions; Max Planck Institute for Terrestrial Microbiology; Karl-von-Frisch-Strasse 10, Marburg D-35043 Germany
| | - Daniel Lanver
- Department of Organismic Interactions; Max Planck Institute for Terrestrial Microbiology; Karl-von-Frisch-Strasse 10, Marburg D-35043 Germany
| | - Hannes Link
- Dynamic Control of Metabolic Networks; Max Planck Institute for Terrestrial Microbiology; Karl-von-Frisch-Strasse 16, Marburg D-35043 Germany
| | - Regine Kahmann
- Department of Organismic Interactions; Max Planck Institute for Terrestrial Microbiology; Karl-von-Frisch-Strasse 10, Marburg D-35043 Germany
| |
Collapse
|
8
|
White J, Mazzeu J, Hoischen A, Jhangiani S, Gambin T, Alcino M, Penney S, Saraiva J, Hove H, Skovby F, Kayserili H, Estrella E, Vulto-van Silfhout A, Steehouwer M, Muzny D, Sutton V, Gibbs R, Lupski J, Brunner H, van Bon B, Carvalho C, Carvalho CMB. DVL1 frameshift mutations clustering in the penultimate exon cause autosomal-dominant Robinow syndrome. Am J Hum Genet 2015; 96:612-22. [PMID: 25817016 DOI: 10.1016/j.ajhg.2015.02.015] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/24/2015] [Indexed: 12/17/2022] Open
Abstract
Robinow syndrome is a genetically heterogeneous disorder characterized by mesomelic limb shortening, genital hypoplasia, and distinctive facial features and for which both autosomal-recessive and autosomal-dominant inheritance patterns have been described. Causative variants in the non-canonical signaling gene WNT5A underlie a subset of autosomal-dominant Robinow syndrome (DRS) cases, but most individuals with DRS remain without a molecular diagnosis. We performed whole-exome sequencing in four unrelated DRS-affected individuals without coding mutations in WNT5A and found heterozygous DVL1 exon 14 mutations in three of them. Targeted Sanger sequencing in additional subjects with DRS uncovered DVL1 exon 14 mutations in five individuals, including a pair of monozygotic twins. In total, six distinct frameshift mutations were found in eight subjects, and all were heterozygous truncating variants within the penultimate exon of DVL1. In five families in which samples from unaffected parents were available, the variants were demonstrated to represent de novo mutations. All variant alleles are predicted to result in a premature termination codon within the last exon, escape nonsense-mediated decay (NMD), and most likely generate a C-terminally truncated protein with a distinct -1 reading-frame terminus. Study of the transcripts extracted from affected subjects' leukocytes confirmed expression of both wild-type and variant alleles, supporting the hypothesis that mutant mRNA escapes NMD. Genomic variants identified in our study suggest that truncation of the C-terminal domain of DVL1, a protein hypothesized to have a downstream role in the Wnt-5a non-canonical pathway, is a common cause of DRS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Claudia M B Carvalho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte MG 30190-002, Brazil.
| |
Collapse
|
9
|
Sendoel A, Maida S, Zheng X, Teo Y, Stergiou L, Rossi CA, Subasic D, Pinto SM, Kinchen JM, Shi M, Boettcher S, Meyer JN, Manz MG, Bano D, Hengartner MO. DEPDC1/LET-99 participates in an evolutionarily conserved pathway for anti-tubulin drug-induced apoptosis. Nat Cell Biol 2014; 16:812-20. [PMID: 25064737 DOI: 10.1038/ncb3010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/18/2014] [Indexed: 12/18/2022]
Abstract
Microtubule-targeting chemotherapeutics induce apoptosis in cancer cells by promoting the phosphorylation and degradation of the anti-apoptotic BCL-2 family member MCL1. The signalling cascade linking microtubule disruption to MCL1 degradation remains however to be defined. Here, we establish an in vivo screening strategy in Caenorhabditis elegans to uncover genes involved in chemotherapy-induced apoptosis. Using an RNAi-based screen, we identify three genes required for vincristine-induced apoptosis. We show that the DEP domain protein LET-99 acts upstream of the heterotrimeric G protein alpha subunit GPA-11 to control activation of the stress kinase JNK-1. The human homologue of LET-99, DEPDC1, similarly regulates vincristine-induced cell death by promoting JNK-dependent degradation of the BCL-2 family protein MCL1. Collectively, these data uncover an evolutionarily conserved mediator of anti-tubulin drug-induced apoptosis and suggest that DEPDC1 levels could be an additional determinant for therapy response upstream of MCL1.
Collapse
Affiliation(s)
- Ataman Sendoel
- 1] Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland [2] Division of Hematology, University Hospital Zurich, Raemistrasse 190 CH-8091 Zurich, Switzerland [3]
| | - Simona Maida
- German Center for Neurodegenerative Diseases (DZNE) e.V. Ludwig-Erhard-Allee 2, D-53175 Bonn, Germany
| | - Xue Zheng
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland
| | - Youjin Teo
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland
| | - Lilli Stergiou
- 1] Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland [2]
| | - Carlo-Alberto Rossi
- German Center for Neurodegenerative Diseases (DZNE) e.V. Ludwig-Erhard-Allee 2, D-53175 Bonn, Germany
| | - Deni Subasic
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland
| | - Sergio M Pinto
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland
| | - Jason M Kinchen
- Center for Cell Clearance, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Moyin Shi
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland
| | - Steffen Boettcher
- Division of Hematology, University Hospital Zurich, Raemistrasse 190 CH-8091 Zurich, Switzerland
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, North Carolina 27708, USA
| | - Markus G Manz
- Division of Hematology, University Hospital Zurich, Raemistrasse 190 CH-8091 Zurich, Switzerland
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE) e.V. Ludwig-Erhard-Allee 2, D-53175 Bonn, Germany
| | - Michael O Hengartner
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190 CH-8057 Zurich, Switzerland
| |
Collapse
|
10
|
Abstract
The Dishevelled, EGL-10 and pleckstrin (DEP) domain is a globular protein domain that is present in about ten human protein families with well-defined structural features. A picture is emerging that DEP domains mainly function in the spatial and temporal control of diverse signal transduction events by recruiting proteins to the plasma membrane. DEP domains can interact with various partners at the membrane, including phospholipids and membrane receptors, and their binding is subject to regulation.
Collapse
|
11
|
Adaptive gene regulation in the Striatum of RGS9-deficient mice. PLoS One 2014; 9:e92605. [PMID: 24663062 PMCID: PMC3963927 DOI: 10.1371/journal.pone.0092605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/24/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND RGS9-deficient mice show drug-induced dyskinesia but normal locomotor activity under unchallenged conditions. RESULTS Genes related to Ca2+ signaling and their functions were regulated in RGS9-deficient mice. CONCLUSION Changes in Ca2+ signaling that compensate for RGS9 loss-of-function can explain the normal locomotor activity in RGS9-deficient mice under unchallenged conditions. SIGNIFICANCE Identified signaling components may represent novel targets in antidyskinetic therapy. The long splice variant of the regulator of G-protein signaling 9 (RGS9-2) is enriched in striatal medium spiny neurons and dampens dopamine D2 receptor signaling. Lack of RGS9-2 can promote while its overexpression prevents drug-induced dyskinesia. Other animal models of drug-induced dyskinesia rather pointed towards overactivity of dopamine receptor-mediated signaling. To evaluate changes in signaling pathways mRNA expression levels were determined and compared in wild-type and RGS9-deficient mice. Unexpectedly, expression levels of dopamine receptors were unchanged in RGS9-deficient mice, while several genes related to Ca2+ signaling and long-term depression were differentially expressed when compared to wild type animals. Detailed investigations at the protein level revealed hyperphosphorylation of DARPP32 at Thr34 and of ERK1/2 in striata of RGS9-deficient mice. Whole cell patch clamp recordings showed that spontaneous synaptic events are increased (frequency and size) in RGS9-deficient mice while long-term depression is reduced in acute brain slices. These changes are compatible with a Ca2+-induced potentiation of dopamine receptor signaling which may contribute to the drug-induced dyskinesia in RGS9-deficient mice.
Collapse
|
12
|
Bodle CR, Mackie DI, Roman DL. RGS17: an emerging therapeutic target for lung and prostate cancers. Future Med Chem 2013; 5:995-1007. [PMID: 23734683 PMCID: PMC3865709 DOI: 10.4155/fmc.13.91] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ligands for G-protein-coupled receptors (GPCRs) represent approximately 50% of currently marketed drugs. RGS proteins modulate heterotrimeric G proteins and, thus, GPCR signaling, by accelerating the intrinsic GTPase activity of the Gα subunit. Given the prevalence of GPCR targeted therapeutics and the role RGS proteins play in G protein signaling, some RGS proteins are emerging as targets in their own right. One such RGS protein is RGS17. Increased RGS17 expression in some prostate and lung cancers has been demonstrated to support cancer progression, while reduced expression of RGS17 can lead to development of chemotherapeutic resistance in ovarian cancer. High-throughput screening is a powerful tool for lead compound identification, and utilization of high-throughput technologies has led to the discovery of several RGS inhibitors, thus far. As screening technologies advance, the identification of novel lead compounds the subsequent development of targeted therapeutics appears promising.
Collapse
Affiliation(s)
- Christopher R Bodle
- The Department of Pharmaceutical Sciences & Experimental Therapeutics, University of Iowa, College of Pharmacy, Iowa City, IA, USA
- Division of Medicinal & Natural Products Chemistry, University of Iowa, College of Pharmacy, Iowa City, IA, USA
| | - Duncan I Mackie
- The Department of Pharmaceutical Sciences & Experimental Therapeutics, University of Iowa, College of Pharmacy, Iowa City, IA, USA
- Division of Medicinal & Natural Products Chemistry, University of Iowa, College of Pharmacy, Iowa City, IA, USA
- Cancer Signaling and Experimental Therapeutics Program, The Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| | - David L Roman
- The Department of Pharmaceutical Sciences & Experimental Therapeutics, University of Iowa, College of Pharmacy, Iowa City, IA, USA
- Division of Medicinal & Natural Products Chemistry, University of Iowa, College of Pharmacy, Iowa City, IA, USA
- Cancer Signaling and Experimental Therapeutics Program, The Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| |
Collapse
|
13
|
cAMP regulates DEP domain-mediated binding of the guanine nucleotide exchange factor Epac1 to phosphatidic acid at the plasma membrane. Proc Natl Acad Sci U S A 2012; 109:3814-9. [PMID: 22343288 DOI: 10.1073/pnas.1117599109] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Epac1 is a cAMP-regulated guanine nucleotide exchange factor for the small G protein Rap. Upon cAMP binding, Epac1 undergoes a conformational change that results in its release from autoinhibition. In addition, cAMP induces the translocation of Epac1 from the cytosol to the plasma membrane. This relocalization of Epac1 is required for efficient activation of plasma membrane-located Rap and for cAMP-induced cell adhesion. This translocation requires the Dishevelled, Egl-10, Pleckstrin (DEP) domain, but the molecular entity that serves as the plasma membrane anchor and the possible mechanism of regulated binding remains elusive. Here we show that Epac1 binds directly to phosphatidic acid. Similar to the cAMP-induced Epac1 translocation, this binding is regulated by cAMP and requires the DEP domain. Furthermore, depletion of phosphatidic acid by inhibition of phospholipase D1 prevents cAMP-induced translocation of Epac1 as well as the subsequent activation of Rap at the plasma membrane. Finally, mutation of a single basic residue within a polybasic stretch of the DEP domain, which abolishes translocation, also prevents binding to phosphatidic acid. From these results we conclude that cAMP induces a conformational change in Epac1 that enables DEP domain-mediated binding to phosphatidic acid, resulting in the tethering of Epac1 at the plasma membrane and subsequent activation of Rap.
Collapse
|
14
|
Li S, Tsalkova T, White MA, Mei FC, Liu T, Wang D, Woods VL, Cheng X. Mechanism of intracellular cAMP sensor Epac2 activation: cAMP-induced conformational changes identified by amide hydrogen/deuterium exchange mass spectrometry (DXMS). J Biol Chem 2011; 286:17889-97. [PMID: 21454623 DOI: 10.1074/jbc.m111.224535] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epac2, a guanine nucleotide exchange factor, regulates a wide variety of intracellular processes in response to second messenger cAMP. In this study, we have used peptide amide hydrogen/deuterium exchange mass spectrometry to probe the solution structural and conformational dynamics of full-length Epac2 in the presence and absence of cAMP. The results support a mechanism in which cAMP-induced Epac2 activation is mediated by a major hinge motion centered on the C terminus of the second cAMP binding domain. This conformational change realigns the regulatory components of Epac2 away from the catalytic core, making the later available for effector binding. Furthermore, the interface between the first and second cAMP binding domains is highly dynamic, providing an explanation of how cAMP gains access to the ligand binding sites that, in the crystal structure, are seen to be mutually occluded by the other cAMP binding domain. Moreover, cAMP also induces conformational changes at the ionic latch/hairpin structure, which is directly involved in RAP1 binding. These results suggest that in addition to relieving the steric hindrance imposed upon the catalytic lobe by the regulatory lobe, cAMP may also be an allosteric modulator directly affecting the interaction between Epac2 and RAP1. Finally, cAMP binding also induces significant conformational changes in the dishevelled/Egl/pleckstrin (DEP) domain, a conserved structural motif that, although missing from the active Epac2 crystal structure, is important for Epac subcellular targeting and in vivo functions.
Collapse
Affiliation(s)
- Sheng Li
- Department of Medicine and Biomedical Sciences Graduate program, University of California, San Diego, La Jolla, California 92093-0656, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Numerous studies indicate that reactive oxygen species (ROS) are not merely cellular by-products of respiration, but are able to modulate various signalling pathways and play certain physiological roles. Recent studies have revealed the importance of translating ROS-generation to activation/suppression of specific signalling pathways. The Wnt signalling pathway, which is essential for early development and stem cell maintenance, is also regulated by ROS. A thioredoxin-related protein, nucleoredoxin (NRX), governs ROS-stimulated Wnt signalling in a temporal manner. NRX usually interacts with Dishevelled (Dvl), an essential adaptor protein for Wnt signalling, and blocks the activation of the Wnt pathway. Oxidative stress causes dissociation of NRX from Dvl, which enables Dvl to activate the downstream Wnt signalling pathway. This study also presents the latest research findings on NRX and its related molecules.
Collapse
Affiliation(s)
- Yosuke Funato
- Laboratory of Intracellular Signaling, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | |
Collapse
|
16
|
Turm H, Maoz M, Katz V, Yin YJ, Offermanns S, Bar-Shavit R. Protease-activated receptor-1 (PAR1) acts via a novel Galpha13-dishevelled axis to stabilize beta-catenin levels. J Biol Chem 2010; 285:15137-15148. [PMID: 20223821 DOI: 10.1074/jbc.m109.072843] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have previously shown a novel link between hPar-1 (human protease-activated receptor-1) and beta-catenin stabilization. Although it is well recognized that Wnt signaling leads to beta-catenin accumulation, the role of PAR1 in the process is unknown. We provide here evidence that PAR1 induces beta-catenin stabilization independent of Wnt, Fz (Frizzled), and the co-receptor LRP5/6 (low density lipoprotein-related protein 5/6) and identify selective mediators of the PAR1-beta-catenin axis. Immunohistological analyses of hPar1-transgenic (TG) mouse mammary tissues show the expression of both Galpha(12) and Galpha(13) compared with age-matched control counterparts. However, only Galpha(13) was found to be actively involved in PAR1-induced beta-catenin stabilization. Indeed, a dominant negative form of Galpha(13) inhibited both PAR1-induced Matrigel invasion and Lef/Tcf (lymphoid enhancer factor/T cell factor) transcription activity. PAR1-Galpha(13) association is followed by the recruitment of DVL (Dishevelled), an upstream Wnt signaling protein via the DIX domain. Small interfering RNA-Dvl silencing leads to a reduction in PAR1-induced Matrigel invasion, inhibition of Lef/Tcf transcription activity, and decreased beta-catenin accumulation. It is of note that PAR1 also promotes the binding of beta-arrestin-2 to DVL, suggesting a role for beta-arrestin-2 in PAR1-induced DVL phosphorylation dynamics. Although infection of small interfering RNA-LRP5/6 or the use of the Wnt antagonists, SFRP2 (soluble Frizzled-related protein 2) or SFRP5 potently reduced Wnt3A-mediated beta-catenin accumulation, no effect was observed on PAR1-induced beta-catenin stabilization. Collectively, our data show that PAR1 mediates beta-catenin stabilization independent of Wnt. We propose here a novel cascade of PAR1-induced Galpha(13)-DVL axis in cancer and beta-catenin stabilization.
Collapse
Affiliation(s)
- Hagit Turm
- Department of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Myriam Maoz
- Department of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Vered Katz
- Department of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Yong-Jun Yin
- Department of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Steffan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Rachel Bar-Shavit
- Department of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel.
| |
Collapse
|
17
|
Slepak VZ. Structure, function, and localization of Gβ5-RGS complexes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 86:157-203. [PMID: 20374716 PMCID: PMC3312022 DOI: 10.1016/s1877-1173(09)86006-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Members of the R7 subfamily of regulator of G protein signaling (RGS) proteins (RGS6, 7, 9, and 11) exist as heterodimers with the G protein beta subunit Gβ5. These protein complexes are only found in neurons and are defined by the presence of three domains: DEP/DHEX, Gβ5/GGL, and RGS. This article summarizes published work in the following areas: (1) the functional significance of structural organization of Gβ5-R7 complexes, (2) regional distribution of Gβ5-R7 in the nervous system and regulation of R7 family expression, (3) subcellular localization of Gβ5-R7 complexes, and (4) novel binding partners of Gβ5-R7 proteins. The review points out some contradictions between observations made by different research groups and highlights the importance of using alternative experimental approaches to obtain conclusive information about Gβ5-R7 function in vivo.
Collapse
Affiliation(s)
- Vladlen Z Slepak
- Department of Molecular and Cellular Pharmacology and the Neuroscience Program, University of Miami School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
18
|
Anderson GR, Posokhova E, Martemyanov KA. The R7 RGS protein family: multi-subunit regulators of neuronal G protein signaling. Cell Biochem Biophys 2009; 54:33-46. [PMID: 19521673 PMCID: PMC2827338 DOI: 10.1007/s12013-009-9052-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 05/27/2009] [Indexed: 01/09/2023]
Abstract
G protein-coupled receptor signaling pathways mediate the transmission of signals from the extracellular environment to the generation of cellular responses, a process that is critically important for neurons and neurotransmitter action. The ability to promptly respond to rapidly changing stimulation requires timely inactivation of G proteins, a process controlled by a family of specialized proteins known as regulators of G protein signaling (RGS). The R7 group of RGS proteins (R7 RGS) has received special attention due to their pivotal roles in the regulation of a range of crucial neuronal processes such as vision, motor control, reward behavior, and nociception in mammals. Four proteins in this group, RGS6, RGS7, RGS9, and RGS11, share a common molecular organization of three modules: (i) the catalytic RGS domain, (ii) a GGL domain that recruits G beta(5), an outlying member of the G protein beta subunit family, and (iii) a DEP/DHEX domain that mediates interactions with the membrane anchor proteins R7BP and R9AP. As heterotrimeric complexes, R7 RGS proteins not only associate with and regulate a number of G protein signaling pathway components, but have also been found to form complexes with proteins that are not traditionally associated with G protein signaling. This review summarizes our current understanding of the biology of the R7 RGS complexes including their structure/functional organization, protein-protein interactions, and physiological roles.
Collapse
Affiliation(s)
- Garret R. Anderson
- From the Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455 USA
| | - Ekaterina Posokhova
- From the Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455 USA
| | - Kirill A. Martemyanov
- From the Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455 USA
| |
Collapse
|
19
|
Sandiford SL, Slepak VZ. The Gbeta5-RGS7 complex selectively inhibits muscarinic M3 receptor signaling via the interaction between the third intracellular loop of the receptor and the DEP domain of RGS7. Biochemistry 2009; 48:2282-9. [PMID: 19182865 PMCID: PMC2766429 DOI: 10.1021/bi801989c] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Regulators of G protein signaling (RGS) make up a diverse family primarily known as GTPase-activating proteins (GAPs) for heterotrimeric G proteins. In addition to the RGS domain, which is responsible for GAP activity, most RGS proteins contain other distinct structural motifs. For example, members of the R7 family of RGS proteins contain a DEP, GGL, and novel DHEX domain and are obligatory dimers with G protein beta subunit Gbeta5. Here we show that the Gbeta5-RGS7 complex can inhibit Ca2+ mobilization elicited by muscarinic acetylcholine receptor type 3 (M3R), but not by other Gq-coupled receptors such as M1, M5, histamine H1, and GNRH receptors. The isolated DEP domain of RGS7 is sufficient for the inhibition of M3R signaling, whereas the deletion of the DEP domain renders the Gbeta5-RGS7 complex ineffective. Deletion of a portion of the third intracellular loop allowed the receptor (M3R-short) to signal but rendered it insensitive to the effect of the Gbeta5-RGS7 complex. Accordingly, the recombinant DEP domain bound in vitro to the GST-fused i3 loop of the M3R. These results identify a novel molecular mechanism that can impart receptor subtype selectivity on signal transduction via Gq-coupled muscarinic receptors.
Collapse
Affiliation(s)
- Simone L. Sandiford
- Department of Molecular and Cellular Pharmacology and the Neuroscience Program, University of Miami School of Medicine, 1600 NW 10 Ave R-189, Miami FL 33136
| | - Vladlen Z. Slepak
- Department of Molecular and Cellular Pharmacology and the Neuroscience Program, University of Miami School of Medicine, 1600 NW 10 Ave R-189, Miami FL 33136
| |
Collapse
|
20
|
Abstract
Pleckstrin, the platelet and leukocyte C kinase substrate, is a prominent substrate of PKC in platelets, monocytes, macrophages, lymphocytes, and granulocytes. Pleckstrin accounts for 1% of the total protein in these cells, but it is best known for containing the 2 prototypic Pleckstrin homology, or PH, domains. Overexpressed pleckstrin can affect polyphosphoinositide second messenger-based signaling events; however, its true in vivo role has been unknown. Here, we describe mice containing a null mutation within the pleckstrin gene. Platelets lacking pleckstrin exhibit a marked defect in exocytosis of delta and alpha granules, alphaIIbbeta3 activation, actin assembly, and aggregation after exposure to the PKC stimulant, PMA. Pleckstrin-null platelets aggregate normally in response to thrombin, but they fail to aggregate in response to thrombin in the presence of PI3K inhibitors, suggesting that a PI3K-dependent signaling pathway compensates for the loss of pleckstrin. Although pleckstrin-null platelets merged their granules in response to stimulation of PKC, they failed to empty their contents into the open canalicular system. This might be attributable to impaired actin assembly present in cells lacking pleckstrin. These data show that pleckstrin regulates the fusion of granules to the cell membrane and is an essential component of PKC-mediated exocytosis.
Collapse
|
21
|
Chapter 7 Biology and Functions of the RGS9 Isoforms. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 86:205-27. [DOI: 10.1016/s1877-1173(09)86007-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
22
|
Hochbaum D, Hong K, Barila G, Ribeiro-Neto F, Altschuler DL. Epac, in synergy with cAMP-dependent protein kinase (PKA), is required for cAMP-mediated mitogenesis. J Biol Chem 2007; 283:4464-8. [PMID: 18063584 DOI: 10.1074/jbc.c700171200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
cAMP stimulates proliferation in many cell types. For many years, cAMP-dependent protein kinase (PKA) represented the only known cAMP effector. PKA, however, does not fully mimic the action of cAMP, indicating the existence of a PKA-independent component. Since cAMP-mediated activation of the G-protein Rap1 and its phosphorylation by PKA are strictly required for the effects of cAMP on mitogenesis, we hypothesized that the Rap1 activator Epac might represent the PKA-independent factor. Here we report that Epac acts synergistically with PKA in cAMP-mediated mitogenesis. We have generated a new dominant negative Epac mutant that revealed that activation of Epac is required for thyroid-stimulating hormone or cAMP stimulation of DNA synthesis. We demonstrate that Epac's action on cAMP-mediated activation of Rap1 and cAMP-mediated mitogenesis depends on the subcellular localization of Epac via its DEP domain. Disruption of the DEP-dependent subcellular targeting of Epac abolished cAMP-Epac-mediated Rap1 activation and thyroid-stimulating hormone-mediated cell proliferation, indicating that an Epac-Rap-PKA signaling unit is critical for the mitogenic action of cAMP.
Collapse
Affiliation(s)
- Daniel Hochbaum
- Department of Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | |
Collapse
|
23
|
Fang W, Pei Y, Bidochka MJ. A regulator of a G protein signalling (RGS) gene, cag8, from the insect-pathogenic fungus Metarhizium anisopliae is involved in conidiation, virulence and hydrophobin synthesis. MICROBIOLOGY-SGM 2007; 153:1017-1025. [PMID: 17379711 DOI: 10.1099/mic.0.2006/002105-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Regulators of the G protein signalling (RGS) pathway have been implicated in the control of a diverse array of cellular functions, including conidiation in filamentous fungi. However, the regulatory processes involved in conidiation in insect-pathogenic fungi are poorly understood. Since conidia are the infective propagules in these fungi, an understanding of the regulatory processes involved in conidiation is essential to the development of an effective biocontrol fungus. Here, the cloning and characterization of an RGS protein gene, cag8 (conidiation-associated gene), from the insect-pathogenic fungus Metarhizium anisopliae is reported. Phylogenetic analysis showed that CAG8 was orthologous to the RGS protein FlbA from Aspergillus nidulans. Complementation of A. nidulans DeltaflbA, which cannot conidiate, with M. anisopliae cag8 restored conidiation. Gene disruption of cag8 in M. anisopliae resulted in the lack of conidia on agar plates and on infected insects, reduced mycelial growth, decreased virulence, lysis during growth in liquid medium as well as lack of pigmentation and irregularly shaped blastospores. Transcript levels of ssgA (hydrophobin-encoding gene) were markedly reduced in a Deltacag8 strain, while pr1A (subtilisin-like protease) transcription was unaffected. These results suggest that cag8 is involved in the modulation of conidiation, virulence and hydrophobin synthesis in M. anisopliae.
Collapse
Affiliation(s)
- Weiguo Fang
- Department of Biological Sciences, Brock University, 500 Glenridge Avenue, St Catharines, ON L2S 3A1, Canada
| | - Yan Pei
- Biotechnology Research Center, Southwest University, Beibei Chongqing 400716, P. R. China
| | - Michael J Bidochka
- Department of Biological Sciences, Brock University, 500 Glenridge Avenue, St Catharines, ON L2S 3A1, Canada
| |
Collapse
|
24
|
Funato Y, Miki H. Nucleoredoxin, a novel thioredoxin family member involved in cell growth and differentiation. Antioxid Redox Signal 2007; 9:1035-57. [PMID: 17567240 DOI: 10.1089/ars.2007.1550] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Thioredoxin (TRX) family proteins are involved in various biologic processes by regulating the response to oxidative stress. Nucleoredoxin (NRX), a relatively uncharacterized member of the TRX family protein, has recently been reported to regulate the Wnt/beta-catenin pathway, which itself regulates cell fate and early development, in a redox-dependent manner. In this review, we describe the TRX family proteins and discuss in detail the similarities and differences between NRX and other TRX family proteins. Although NRX possesses a conserved TRX domain and a catalytic motif for oxidoreductase activity, its sequence homology to TRX is not as high as that of the close relatives of TRX. The sequence of NRX is more similar to that of tryparedoxin (TryX), a TRX family member originally identified in parasite trypanosomes. We also discuss the reported properties and potential physiologic roles of NRX.
Collapse
Affiliation(s)
- Yosuke Funato
- Division of Cancer Genomics, Institute of Medical Science, University of Tokyo, Japan
| | | |
Collapse
|
25
|
Narayanan V, Sandiford SL, Wang Q, Keren-Raifman T, Levay K, Slepak VZ. Intramolecular interaction between the DEP domain of RGS7 and the Gbeta5 subunit. Biochemistry 2007; 46:6859-70. [PMID: 17511476 DOI: 10.1021/bi700524w] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The R7 family of RGS proteins (RGS6, -7, -9, -11) is characterized by the presence of three domains: DEP, GGL, and RGS. The RGS domain interacts with Galpha subunits and exhibits GAP activity. The GGL domain permanently associates with Gbeta5. The DEP domain interacts with the membrane anchoring protein, R7BP. Here we provide evidence for a novel interaction within this complex: between the DEP domain and Gbeta5. GST fusion of the RGS7 DEP domain (GST-R7DEP) binds to both native and recombinant Gbeta5-RGS7, recombinant Gbetagamma complexes, and monomeric Gbeta5 and Gbeta1 subunits. Co-immunoprecipitation and FRET assays supported the GST pull-down experiments. GST-R7DEP reduced FRET between CFP-Gbeta5 and YFP-RGS7, indicating that the DEP-Gbeta5 interaction is dynamic. In transfected cells, R7BP had no effect on the Gbeta5/RGS7 pull down by GST-R7DEP. The DEP domain of RGS9 did not bind to Gbeta5. Substitution of RGS7 Glu-73 and Asp-74 for the corresponding Ser and Gly residues (ED/SG mutation) of RGS9 diminished the DEP-Gbeta5 interaction. In the absence of R7BP both the wild-type RGS7 and the ED/SG mutant attenuated muscarinic M3 receptor-mediated Ca2+ mobilization. In the presence of R7BP, wild-type RGS7 lost this inhibitory activity, whereas the ED/SG mutant remained active. Taken together, our results are consistent with the following model. The Gbeta5-RGS7 molecule can exist in two conformations: "closed" and "open", when the DEP domain and Gbeta5 subunit either do or do not interact. The closed conformation appears to be less active with respect to its effect on Gq-mediated signaling than the open conformation.
Collapse
Affiliation(s)
- Vijaya Narayanan
- Department of Molecular and Cellular Pharmacology and Neuroscience Program, University of Miami, Miami, Florida 33136, USA
| | | | | | | | | | | |
Collapse
|
26
|
Kanehira M, Harada Y, Takata R, Shuin T, Miki T, Fujioka T, Nakamura Y, Katagiri T. Involvement of upregulation of DEPDC1 (DEP domain containing 1) in bladder carcinogenesis. Oncogene 2007; 26:6448-55. [PMID: 17452976 DOI: 10.1038/sj.onc.1210466] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In an attempt to disclose mechanisms of bladder carcinogenesis and discover novel target molecules for development of treatment, we applied a cDNA microarray to screen genes that were significantly transactivated in bladder cancer cells. Among the upregulated genes, we here focused on a novel gene, (DEPDC1) DEP domain containing 1, whose overexpression was confirmed by northern blot and immunohistochemical analyses. Immunocytochemical staining analysis detected strong staining of endogenous DEPDC1 protein in the nucleus of bladder cancer cells. Since DEPDC1 expression was hardly detectable in any of 24 normal human tissues we examined except the testis, we considered this gene-product to be a novel cancer/testis antigen. Suppression of DEPDC1 expression with small-interfering RNA significantly inhibited growth of bladder cancer cells. Taken together, these findings suggest that DEPDC1 might play an essential role in the growth of bladder cancer cells, and would be a promising molecular-target for novel therapeutic drugs or cancer peptide-vaccine to bladder cancers.
Collapse
Affiliation(s)
- M Kanehira
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Bringmann H, Cowan CR, Kong J, Hyman AA. LET-99, GOA-1/GPA-16, and GPR-1/2 are required for aster-positioned cytokinesis. Curr Biol 2006; 17:185-91. [PMID: 17189697 DOI: 10.1016/j.cub.2006.11.070] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2006] [Revised: 11/08/2006] [Accepted: 11/16/2006] [Indexed: 11/25/2022]
Abstract
At anaphase, the mitotic spindle positions the cytokinesis furrow [1]. Two populations of spindle microtubules are implicated in cytokinesis: radial microtubule arrays called asters and bundled nonkinetochore microtubules called the spindle midzone [2-4]. In C. elegans embryos, these two populations of microtubules provide two consecutive signals that position the cytokinesis furrow: The first signal is positioned midway between the microtubule asters; the second signal is positioned over the spindle midzone [5]. Evidence for two cytokinesis signals came from the identification of molecules that block midzone-positioned cytokinesis [5-7]. However, no molecules that are only required for, and thus define, the molecular pathway of aster-positioned cytokinesis have been identified. With RNAi screening, we identify LET-99 and the heterotrimeric G proteins GOA-1/GPA-16 and their regulator GPR-1/2 [10-12] in aster-positioned cytokinesis. By using mechanical spindle displacement, we show that the anaphase spindle positions cortical LET-99, at the site of the presumptive cytokinesis furrow. LET-99 enrichment at the furrow depends on the G proteins. GPR-1 is locally reduced at the site of cytokinesis-furrow formation by LET-99, which prevents accumulation of GPR-1 at this site. We conclude that LET-99 and the G proteins define a molecular pathway required for aster-positioned cytokinesis.
Collapse
Affiliation(s)
- Henrik Bringmann
- Max Planck Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| | | | | | | |
Collapse
|
28
|
Abstract
The DEP domain is present in a number of signaling molecules, including Regulator of G protein Signaling (RGS) proteins, and has been implicated in membrane targeting. New findings in yeast, however, demonstrate a major role for a DEP domain in mediating the interaction of an RGS protein to the C-terminal tail of a GPCR, thus placing RGS in close proximity with its substrate G protein alpha subunit.
Collapse
Affiliation(s)
- Songhai Chen
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
29
|
Civera C, Simon B, Stier G, Sattler M, Macias MJ. Structure and dynamics of the human pleckstrin DEP domain: distinct molecular features of a novel DEP domain subfamily. Proteins 2006; 58:354-66. [PMID: 15573383 DOI: 10.1002/prot.20320] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pleckstrin1 is a major substrate for protein kinase C in platelets and leukocytes, and comprises a central DEP (disheveled, Egl-10, pleckstrin) domain, which is flanked by two PH (pleckstrin homology) domains. DEP domains display a unique alpha/beta fold and have been implicated in membrane binding utilizing different mechanisms. Using multiple sequence alignments and phylogenetic tree reconstructions, we find that 6 subfamilies of the DEP domain exist, of which pleckstrin represents a novel and distinct subfamily. To clarify structural determinants of the DEP fold and to gain further insight into the role of the DEP domain, we determined the three-dimensional structure of the pleckstrin DEP domain using heteronuclear NMR spectroscopy. Pleckstrin DEP shares main structural features with the DEP domains of disheveled and Epac, which belong to different DEP subfamilies. However, the pleckstrin DEP fold is distinct from these structures and contains an additional, short helix alpha4 inserted in the beta4-beta5 loop that exhibits increased backbone mobility as judged by NMR relaxation measurements. Based on sequence conservation, the helix alpha4 may also be present in the DEP domains of regulator of G-protein signaling (RGS) proteins, which are members of the same DEP subfamily. In pleckstrin, the DEP domain is surrounded by two PH domains. Structural analysis and charge complementarity suggest that the DEP domain may interact with the N-terminal PH domain in pleckstrin. Phosphorylation of the PH-DEP linker, which is required for pleckstrin function, could regulate such an intramolecular interaction. This suggests a role of the pleckstrin DEP domain in intramolecular domain interactions, which is distinct from the functions of other DEP domain subfamilies found so far.
Collapse
Affiliation(s)
- Concepcion Civera
- Dpto Quimica Fisica II, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
30
|
He B, Jablons DM. Wnt signaling in stem cells and lung cancer. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2006:27-58. [PMID: 17939294 DOI: 10.1007/2789_2007_043] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The Wnt signal transduction pathway plays important roles during embryo development, regulating cell proliferation and survival of immature cells. However, its improper function can lead to harmful consequences for humans, such as aberrant cell proliferation and, therefore, cancer. Increasing evidence suggests that stem cells may be the source of mutant cells that cause cancers to develop and proliferate. Wnt signaling has been shown to promote self-renewal in both gut epithelial and hematopoietic stem cells (HSCs) and to trigger critical pathways in carcinogenesis. Although the function of stem cells in solid tumor development is unclear, the Wnt pathway's role in determining the fate and self-renewal potential of cancer stem cells suggests a critical role in carcinogenesis. The development of new inhibitors, such as antibodies or small molecules, to inhibit this pathway may be of great therapeutic utility against cancer.
Collapse
Affiliation(s)
- B He
- Department of Surgery, University of California San Francisco Cancer Center, 1600 Divisadero Street, Box 1724, 94143-1724 San Francisco, USA.
| | | |
Collapse
|
31
|
Arigoni M, Bracco E, Lusche DF, Kae H, Weeks G, Bozzaro S. A novel Dictyostelium RasGEF required for chemotaxis and development. BMC Cell Biol 2005; 6:43. [PMID: 16336640 PMCID: PMC1325028 DOI: 10.1186/1471-2121-6-43] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Accepted: 12/07/2005] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ras proteins are guanine-nucleotide-binding enzymes that couple cell surface receptors to intracellular signaling pathways controlling cell proliferation and differentiation, both in lower and higher eukaryotes. They act as molecular switches by cycling between active GTP and inactive GDP-bound states, through the action of two classes of regulatory proteins: a) guanine nucleotide exchange factor (GEFs) and b) GTP-ase activating proteins (GAPs). Genome wide analysis of the lower eukaryote Dictyostelium discoideum revealed a surprisingly large number of Ras Guanine Nucleotide Exchange Factors (RasGEFs). RasGEFs promote the activation of Ras proteins by catalyzing the exchange of GDP for GTP, thus conferring to RasGEFs the role of main activator of Ras proteins. Up to date only four RasGEFs, which are all non-redundant either for growth or development, have been characterized in Dictyostelium. We report here the identification and characterization of a fifth non-redundant GEF, RasGEFM. RESULTS RasGEFM is a multi-domain protein containing six poly-proline stretches, a DEP, RasGEFN and RasGEF catalytic domain. The rasGEFM gene is differentially expressed during growth and development. Inactivation of the gene results in cells that form small, flat aggregates and fail to develop further. Expression of genes required for aggregation is delayed. Chemotaxis towards cAMP is impaired in the mutant, due to inability to inhibit lateral pseudopods. Endogenous cAMP accumulates during early development to a much lower extent than in wild type cells. Adenylyl cyclase activation in response to cAMP pulses is strongly reduced, by contrast guanylyl cyclase is stimulated to higher levels than in the wild type. The actin polymerization response to cAMP is also altered in the mutant. Cyclic AMP pulsing for several hours partially rescues the mutant. In vitro experiments suggest that RasGEFM acts downstream of the cAMP receptor but upstream of the G protein. CONCLUSION The data indicate that RasGEFM is involved in the establishment of the cAMP relay system. We propose that RasGEFM is a component of a Ras regulated pathway, which integrate signals acting as positive regulator for adenylyl cyclase and negative regulator for guanylyl cyclase. Altered guanylyl cyclase, combined with defective regulation of actin polymerization, results in altered chemotaxis.
Collapse
Affiliation(s)
- Maddalena Arigoni
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Enrico Bracco
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Daniel F Lusche
- Faculty of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Helmut Kae
- Dept. Microbiology and Immunology, University of British Columbia, Canada V6T1Z3
| | - Gerald Weeks
- Dept. Microbiology and Immunology, University of British Columbia, Canada V6T1Z3
| | - Salvatore Bozzaro
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, 10043 Orbassano, Italy
| |
Collapse
|
32
|
Inazu T, Kuroiwa A, Matsuda Y, Miyamoto K. Cloning, expression and chromosomal assignment of human pleckstrin 2. Mol Biol Rep 2005; 32:35-40. [PMID: 15865208 DOI: 10.1007/s11033-004-4747-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We report the isolation of human pleckstrin 2 cDNA. The cDNA contains a 1059 bp open reading frame encoding a polypeptide of 353 amino acid residues. The deduced amino acid sequence indicates that pleckstrin 2 contains two pleckstrin homology domains and a DEP (dishvelled, egl-10, and pleckstrin) domain and had a 95% identity with the sequence of mouse pleckstrin 2. Northern blot and a reverse transcription-coupled polymerase chain reaction analysis revealed that pleckstrin 2 mRNA is widely expressed in a variety of cell lines. The chromosomal location of the mouse pleckstrin 2 gene was on the D3 band of chromosome 12, as determined by fluorescence in situ hybridization and the human pleckstrin 2 gene was mapped to chromosome 14q24.1 by a bioinformatics analysis.
Collapse
Affiliation(s)
- Tetsuya Inazu
- Department of Biochemistry, Fukui Medical University, Matsuoka, Fukui, Japan.
| | | | | | | |
Collapse
|
33
|
He B, Barg RN, You L, Xu Z, Reguart N, Mikami I, Batra S, Rosell R, Jablons DM. Wnt Signaling in Stem Cells and Non–Small-Cell Lung Cancer. Clin Lung Cancer 2005; 7:54-60. [PMID: 16098245 DOI: 10.3816/clc.2005.n.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Evidence suggests that stem cells may be the source of mutant cells that cause cancers to develop and proliferate. Wnt signaling has been shown to promote self-renewal in gut epithelial and hematopoietic stem cells and to trigger critical pathways in carcinogenesis. In this review, we highlight the progress in understanding how the Wnt pathway contributes to stem cell maintenance and its role in lung carcinogenesis. Although the function of stem cells in solid tumor development is unclear, the Wnt pathway's role in determining the fate and self-renewal potential of cancer stem cells suggests a critical role in carcinogenesis and that developing drugs to inhibit this pathway may be of therapeutic interest.
Collapse
Affiliation(s)
- Biao He
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California-San Francisco, 1600 Divisadero Street, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Segers GC, Regier JC, Nuss DL. Evidence for a role of the regulator of G-protein signaling protein CPRGS-1 in Galpha subunit CPG-1-mediated regulation of fungal virulence, conidiation, and hydrophobin synthesis in the chestnut blight fungus Cryphonectria parasitica. EUKARYOTIC CELL 2005; 3:1454-63. [PMID: 15590820 PMCID: PMC539028 DOI: 10.1128/ec.3.6.1454-1463.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We previously reported that the chestnut blight fungus Cryphonectria parasitica expresses at least three G-protein alpha subunits and that Galpha subunit CPG-1 is essential for regulated growth, pigmentation, sporulation, and virulence. We now report the cloning and characterization of a C. parasitica regulator of G-protein signaling (RGS) protein, CPRGS-1. The phylogenetic relationship of CPRGS-1 to orthologs from other fungi was inferred and found to be generally concordant with species relationships based on 18S ribosomal sequences and on morphology. However, Hemiascomycotine RGS branch lengths in particular were longer than for their 18S sequence counterparts, which correlates with functional diversification in the signaling pathway. Deletion of cprgs-1 resulted in reduced growth, sparse aerial mycelium, and loss of pigmentation, sporulation, and virulence. Disruption of cprgs-1 was also accompanied by a severe posttranscriptional reduction in accumulation of CPG-1 and Gbeta subunit CPGB-1 and severely reduced expression of the hydrophobin-encoding gene cryparin. The changes in phenotype, cryparin expression, and CPGB-1 accumulation resulting from cprgs-1 gene deletion were also observed in a strain containing a mutationally activated copy of CPG-1 but not in strains containing constitutively activated mutant alleles of the other two identified Galpha subunits, CPG-2 and CPG-3. Furthermore, cprgs-1 transcript levels were increased in the activated CPG-1 strain but were unaltered in activated CPG-2 and CPG-3 strains. The results strongly suggest that CPRGS-1 is involved in regulation of Galpha subunit CPG-1-mediated signaling and establish a role for a RGS protein in the modulation of virulence, conidiation, and hydrophobin synthesis in a plant pathogenic fungus.
Collapse
Affiliation(s)
- Gerrit C. Segers
- Center for Biosystems Research, University of Maryland Biotechnology Institute, College Park, Maryland
| | - Jerome C. Regier
- Center for Biosystems Research, University of Maryland Biotechnology Institute, College Park, Maryland
| | - Donald. L. Nuss
- Center for Biosystems Research, University of Maryland Biotechnology Institute, College Park, Maryland
- Corresponding author. Mailing address: Center for Biosystems Research, University of Maryland Biotechnology Institute, 5115 Plant Sciences Bldg., College Park, MD 20742. Phone: (301) 405-0334. Fax: (301) 314-9075. E-mail:
| |
Collapse
|
35
|
Mazieres J, He B, You L, Xu Z, Jablons DM. Wnt signaling in lung cancer. Cancer Lett 2005; 222:1-10. [PMID: 15837535 DOI: 10.1016/j.canlet.2004.08.040] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2004] [Accepted: 08/26/2004] [Indexed: 01/21/2023]
Abstract
Wnt signaling has recently emerged as a critical pathway in lung carcinogenesis as already demonstrated in many cancers and particularly in colorectal cancer. We critically discuss in this review the individual components of the Wnt pathway and their role in lung cancer development. We propose that activation of the Wnt-mediated signal occurs in a different manner in lung cancer than in colorectal cancer. In lung cancer, mutations of APC or beta-catenin are rare and the Wnt pathway appears to be activated upstream of beta-catenin. We identified at least three mechanisms of activation: overexpression of Wnt effectors such as Dvl, activation of a non-canonical pathway involving JNK and repression of Wnt antagonists such as WIF-1. The respective relevance of each event and their likely relationship remain unclear. Nevertheless, we propose that many of the studied components of the Wnt pathway may serve as potential targets in the search for therapeutic agents and we can reasonably argue that blockade of Wnt pathway may lead to new treatment strategies in lung cancer.
Collapse
Affiliation(s)
- Julien Mazieres
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, 1600 Divisadero St., C322C, Box 1674, San Francisco, CA 94115, USA
| | | | | | | | | |
Collapse
|
36
|
Ajit SK, Young KH. Analysis of chimeric RGS proteins in yeast for the functional evaluation of protein domains and their potential use in drug target validation. Cell Signal 2004; 17:817-25. [PMID: 15763424 DOI: 10.1016/j.cellsig.2004.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2004] [Accepted: 11/01/2004] [Indexed: 11/27/2022]
Abstract
For the identification of regulators of G-protein signaling (RGS) modulators, previously, we developed a luciferase based yeast pheromone response (YPhR) assay to functionally investigate RGS4 (K.H. Young, Y. Wang, C. Bender, S. Ajit, F. Ramirez, A. Gilbert, B.W. Nieuwenhuijsen, in: D.P. Siderovski (Ed.), Meth. Enzymol. 389 Regulators of G_protein Signaling, Part A, 2004.). To extend the diversity of this assay, additional RGS proteins were evaluated for functional complementation in a RGS (sst2Delta) knockout yeast strain. For RGS proteins that did not function in their native form, a series of chimeric constructs were generated with the N terminus of RGS4 fused in frame with the partial or full-length RGS cDNA of interest. RGS4 N terminus fused to either full-length or the C terminus of RGS7 successfully complemented sst2Delta. On the contrary, the RGS7N/RGS4C chimera (N terminus of RGS7 in frame with RGS domain of RGS4) was not effective, showing that N terminus of RGS4 helps in targeting. RGS10 exists as two splice variants, differing only by 8 amino acids (aa) in the N terminus, being either 168 aa (RGS10S), or 174 aa (RGS10). While RGS10 was functional in yeast, RGS10S required the presence of the N terminus of RGS4 for its activity. Although the same RGS4 N terminus domain was present in chimeras generated, the GTPase accelerating protein (GAP) function observed was not similar, suggesting differences in the RGS domain function. In conclusion, the use of RGS4 N terminus chimeric constructs enabled us to develop a selectivity assay for different RGS proteins.
Collapse
Affiliation(s)
- Seena K Ajit
- Wyeth Research, Neuroscience Discovery Research, CN 8000, Princeton NJ 08543, USA
| | | |
Collapse
|
37
|
Nixon AB, Casey PJ. Analysis of the Regulation of Microtubule Dynamics by Interaction of RGSZ1 (RGS20) with the Neuronal Stathmin, SCG10. Methods Enzymol 2004; 390:53-64. [PMID: 15488170 DOI: 10.1016/s0076-6879(04)90004-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Regulators of G protein signaling (RGS proteins) are a diverse family of proteins that act to negatively regulate signaling by heterotrimeric G proteins; however, recent data have implied additional functions for RGS proteins. Previously, we employed the yeast two-hybrid system and identified the microtubule-destabilizing protein, superior cervical ganglia neural-specific 10 protein (SCG10), as a potential effector protein of RGSZ1. This article describes the expression and biochemical purification of both RGSZ1 and SCG10 and details the development of various in vitro assays to evaluate microtubule polymerization?depolymerization. Both turbidimetric and microscopy-based assays can be employed to study the impact that RGS proteins have on SCG10 function. The application of these in vitro assays may help identify a novel role for RGS proteins in regulating the cytoskeletal network.
Collapse
Affiliation(s)
- Andrew B Nixon
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
38
|
Abstract
Heterodimeric complexes between individual members of the R7 subfamily of regulators of G-protein signaling proteins and the Gbeta5 isoform of the heterotrimeric G-protein beta subunit family are strongly expressed in the cell nucleus in neurons and brain, as well as in the cytoplasm and plasma membrane. Native and recombinant Gbeta5 and/or R7 expression have been studied in model systems like rat pheochromocytoma PC12 cells where their nuclear localization can be studied by fluorescence microscopy and/or subcellular fractionation. Nucleic acid counterstains chosen for compatibility with the fluorescent tags on secondary antibodies can facilitate the assay of R7/Gbeta5 nuclear localization by epifluorescence or confocal laser microscopy. Subcellular fractionation allows isolation of a purified nuclear fraction that can be probed for the presence of Gbeta5 and/or R7 subunits by immunoblots or immunoprecipitation and compared to other subcellular fractions. While the function of nuclear R7/Gbeta5 complexes is unknown, comparison with the properties of other RGS proteins that localize to the cell nucleus may suggest modes of action. Models are offered in which the reversible post-translational modification of R7/Gbeta5 complexes regulates their nuclear localization and signaling activity, whether the target of such signaling activity is in the nucleus, at the plasma membrane, or both.
Collapse
Affiliation(s)
- William F Simonds
- Metabolic Diseases Branch/NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
39
|
Yang P, Qyang Y, Bartholomeusz G, Zhou X, Marcus S. The novel Rho GTPase-activating protein family protein, Rga8, provides a potential link between Cdc42/p21-activated kinase and Rho signaling pathways in the fission yeast, Schizosaccharomyces pombe. J Biol Chem 2003; 278:48821-30. [PMID: 14506270 DOI: 10.1074/jbc.m306819200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The PAK family kinase, Shk1, is an essential regulator of polarized growth in the fission yeast, Schizosaccharomyces pombe. Here we describe the characterization of a novel member of the RhoGAP family, Rga8, identified from a two-hybrid screen for proteins that interact with the Shk1 kinase domain. Although deletion of the rga8 gene in wild type S. pombe cells results in no obvious phenotypic defects under normal growth conditions, it partially suppresses the cold-sensitive growth and morphological defects of S. pombe cells carrying a hypomorphic allele of the shk1 gene. By contrast, overexpression of rga8 is lethal to shk1-defective cells and causes morphological and cytokinesis defects in wild type S. pombe cells. Consistent with a role for Rga8 as a downstream target of Shk1, we show that the Rga8 protein is directly phosphorylated by Shk1 in vitro and phosphorylated in a Shk1-dependent fashion in S. pombe cells. Fluorescence photomicroscopy of the GFP-Rga8 fusion protein indicates that Rga8 is localized to the cell ends during interphase and to the septum-forming region during cytokinesis. In S. pombe cells carrying the orb2-34 allele of shk1, Rga8 exhibits a monopolar pattern of localization, providing evidence that Shk1 contributes to the regulation of Rga8 localization. Although molecular analyses suggest that Rga8 functions as a GAP for the S. pombe Rho1 GTPase, genetic experiments suggest that Rga8 and Rho1 have a positive functional interaction and that gain of Rho1 function, like gain of Rga8 function, is lethal to Shk1-defective cells. Our results suggest that Rga8 is a Shk1 substrate that negatively regulates Shk1-dependent growth control pathway(s) in S. pombe, potentially through interaction with the Rho1 GTPase.
Collapse
Affiliation(s)
- Peirong Yang
- Department of Molecular Genetics and Program in Genes and Development, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
40
|
Hu G, Zhang Z, Wensel TG. Activation of RGS9-1GTPase acceleration by its membrane anchor, R9AP. J Biol Chem 2003; 278:14550-4. [PMID: 12560335 DOI: 10.1074/jbc.m212046200] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The GTPase-accelerating protein (GAP) complex RGS9-1.G beta(5) plays an important role in the kinetics of light responses by accelerating the GTP hydrolysis of G alpha(t) in vertebrate photoreceptors. Much, but not all, of this complex is tethered to disk membranes by the transmembrane protein R9AP. To determine the effect of the R9AP membrane complex on GAP activity, we purified recombinant R9AP and reconstituted it into lipid vesicles along with the photon receptor rhodopsin. Full-length RGS9-1.G beta(5) bound to R9AP-containing vesicles with high affinity (K(d) < 10 nm), but constructs lacking the DEP (dishevelled/EGL-10/pleckstrin) domain bound with much lower affinity, and binding of those lacking the entire N-terminal domain (i.e. the dishevelled/EGL-10/pleckstrin domain plus intervening domain) was not detectable. Formation of the membrane-bound complex with R9AP increased RGS9-1 GAP activity by a factor of 4. Vesicle titrations revealed that on the time scale of phototransduction, the entire reaction sequence from GTP uptake to GAP-catalyzed hydrolysis is a membrane-delimited process, and exchange of G alpha(t) between membrane surfaces is much slower than hydrolysis. Because in rod cells different pools exist of RGS9-1.G beta(5) that are either associated with R9AP or not, regulation of the association between R9AP and RGS9-1.G beta(5) represents a potential mechanism for the regulation of recovery kinetics.
Collapse
Affiliation(s)
- Guang Hu
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
41
|
Hunt RA, Edris W, Chanda PK, Nieuwenhuijsen B, Young KH. Snapin interacts with the N-terminus of regulator of G protein signaling 7. Biochem Biophys Res Commun 2003; 303:594-9. [PMID: 12659861 DOI: 10.1016/s0006-291x(03)00400-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The N-terminus of regulator of G protein signaling 7 (RGS7) contains a dishevelled/egl-10/pleckstrin (DEP) domain of unknown function. To gain insight into its function, we used yeast two-hybrid analysis to screen a human whole brain cDNA library in order to identify proteins that interact specifically with the N-terminus of human RGS7 (amino acid residues 1-248). From this analysis, we identified snapin, a protein associated with the SNARE complex in neurons, as an interactor with the N-terminus of RGS7. Deletion mutation analysis in yeast demonstrated that the interaction between RGS7 and snapin is specific and is mediated primarily by amino acid residues 1-69 of RGS7 (which contains the proximal portion of the DEP domain). The interaction between RGS7 and snapin was also demonstrated in mammalian cells by coimmunoprecipitation and pull-down assays. Our results suggest that RGS7 could play a role in synaptic vesicle exocytosis through its interaction with snapin.
Collapse
Affiliation(s)
- Rachel A Hunt
- Neuroscience Discovery Research, Wyeth Research, Princeton, NJ 08543-8000, USA
| | | | | | | | | |
Collapse
|
42
|
Warner DR, Pisano MM, Roberts EA, Greene RM. Identification of three novel Smad binding proteins involved in cell polarity. FEBS Lett 2003; 539:167-73. [PMID: 12650946 DOI: 10.1016/s0014-5793(03)00155-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A yeast two-hybrid screen was utilized to identify novel Smad 3 binding proteins expressed in developing mouse orofacial tissue. Three proteins (Erbin, Par-3, and Dishevelled) were identified that share several similar structural and functional characteristics. Each contains at least one PDZ domain and all have been demonstrated to play a role in the establishment and maintenance of cell polarity. In GST (glutathione S-transferase) pull-down assays, Erbin, Par-3, and Dishevelled bound strongly to the isolated MH2 domain of Smad 3, with weaker binding to a full-length Smad 3 protein. Failure of Erbin, Par-3, and Dishevelled to bind to a Smad 3 mutant protein that was missing the MH2 domain confirms that the binding site resides within the MH2 domain. Erbin, Par-3, and Dishevelled also interacted with the MH2 domains of other Smads, suggesting broad Smad binding specificity. Dishevelled and Erbin mutant proteins, in which the PDZ domain was removed, still retained their ability to bind Smad 3, albeit with lower affinity. While transforming growth factor beta (TGFbeta) has been suggested to alter cell polarity through a Smad-independent mechanism involving activation of members of the RhoA family of GTP binding proteins, the observation that Smads can directly interact with proteins involved in cell polarity, as shown in the present report, suggests an additional means by which TGFbeta could alter cell polarity via a Smad-dependent signaling mechanism.
Collapse
Affiliation(s)
- Dennis R Warner
- University of Louisville Birth Defects Center, Department of Molecular, Cellular, and Craniofacial Biology, University of Louisville School of Dentistry, 501 South Preston Street, Suite 301, Louisville, KY 40292, USA.
| | | | | | | |
Collapse
|
43
|
Hooks SB, Waldo GL, Corbitt J, Bodor ET, Krumins AM, Harden TK. RGS6, RGS7, RGS9, and RGS11 stimulate GTPase activity of Gi family G-proteins with differential selectivity and maximal activity. J Biol Chem 2003; 278:10087-93. [PMID: 12531899 DOI: 10.1074/jbc.m211382200] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Regulator of G-protein signaling (RGS) proteins are GTPase activating proteins (GAPs) of heterotrimeric G-proteins that alter the amplitude and kinetics of receptor-promoted signaling. In this study we defined the G-protein alpha-subunit selectivity of purified Sf9 cell-derived R7 proteins, a subfamily of RGS proteins (RGS6, -7, -9, and -11) containing a Ggamma-like (GGL) domain that mediates dimeric interaction with Gbeta(5). Gbeta(5)/R7 dimers stimulated steady state GTPase activity of Galpha-subunits of the G(i) family, but not of Galpha(q) or Galpha(11), when added to proteoliposomes containing M2 or M1 muscarinic receptor-coupled G-protein heterotrimers. Concentration effect curves of the Gbeta(5)/R7 proteins revealed differences in potencies and efficacies toward Galpha-subunits of the G(i) family. Although all four Gbeta(5)/R7 proteins exhibited similar potencies toward Galpha(o), Gbeta(5)/RGS9 and Gbeta(5)/RGS11 were more potent GAPs of Galpha(i1), Galpha(i2), and Galpha(i3) than were Gbeta(5)/RGS6 and Gbeta(5)/RGS7. The maximal GAP activity exhibited by Gbeta(5)/RGS11 was 2- to 4-fold higher than that of Gbeta(5)/RGS7 and Gbeta(5)/RGS9, with Gbeta(5)/RGS6 exhibiting an intermediate maximal GAP activity. Moreover, the less efficacious Gbeta(5)/RGS7 and Gbeta(5)/RGS9 inhibited Gbeta(5)/RGS11-stimulated GTPase activity of Galpha(o). Therefore, R7 family RGS proteins are G(i) family-selective GAPs with potentially important differences in activities.
Collapse
Affiliation(s)
- Shelley B Hooks
- Department of Pharmacology, University of North Carolina, Chapel Hill 27599, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Wieland T, Mittmann C. Regulators of G-protein signalling: multifunctional proteins with impact on signalling in the cardiovascular system. Pharmacol Ther 2003; 97:95-115. [PMID: 12559385 DOI: 10.1016/s0163-7258(02)00326-1] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Regulator of G-protein signalling (RGS) proteins form a superfamily of at least 25 proteins, which are highly diverse in structure, expression patterns, and function. They share a 120 amino acid homology domain (RGS domain), which exhibits GTPase accelerating activity for alpha-subunits of heterotrimeric G-proteins, and thus, are negative regulators of G-protein-mediated signalling. Based on the organisation of the Rgs genes, structural similarities, and differences in functions, they can be divided into at least six subfamilies of RGS proteins and three more families of RGS-like proteins. Many of these proteins regulate signalling processes within cells, not only via interaction with G-protein alpha-subunits, but are G-protein-regulated effectors, Gbetagamma scavenger, or scaffolding proteins in signal transduction complexes as well. The expression of at least 16 different RGS proteins in the mammalian or human myocardium have been described. A subgroup of at least eight was detected in a single atrial myocyte. The exact functions of these proteins remain mostly elusive, but RGS proteins such as RGS4 are involved in the regulation of G(i)-protein betagamma-subunit-gated K(+) channels. An up-regulation of RGS4 expression has been consistently found in human heart failure and some animal models. Evidence is increasing that the enhanced RGS4 expression counter-regulates the G(q/11)-induced signalling caused by hypertrophic stimuli. In the vascular system, RGS5 seems to be an important signalling regulator. It is expressed in vascular endothelial cells, but not in cultured smooth muscle cells. Its down-regulation, both in a model of capillary morphogenesis and in an animal model of stroke, render it a candidate gene, which may be involved in the regulation of capillary growth, angiogenesis, and in the pathophysiology of stroke.
Collapse
Affiliation(s)
- Thomas Wieland
- Institut für Pharmakologie und Toxikologie, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Maybachstrasse 14-16, D-68169 Mannheim, Germany.
| | | |
Collapse
|
45
|
Abstract
Cell polarity is an essential feature of many animal cells. It is critical for epithelial formation and function, for correct partitioning of fate-determining molecules, and for individual cells to chemotax or grow in a defined direction. For some of these processes, the position and orientation of the mitotic spindle must be coupled to cell polarity for correct positioning of daughter cells and inheritance of localised molecules. Recent work in several different systems has led to the realisation that similar mechanisms dictate the establishment of polarity and subsequent spindle positioning in many animal cells. Microtubules and conserved PAR proteins are essential mediators of cell polarity, and mitotic spindle positioning depends on heterotrimeric G protein signalling and the microtubule motor protein dynein.
Collapse
Affiliation(s)
- Julie Ahringer
- Wellcome Trust/Cancer Research UK Institute, Tennis Court Road, Cambridge CB2 1QR, UK.
| |
Collapse
|
46
|
Wharton KA. Runnin' with the Dvl: proteins that associate with Dsh/Dvl and their significance to Wnt signal transduction. Dev Biol 2003; 253:1-17. [PMID: 12490194 DOI: 10.1006/dbio.2002.0869] [Citation(s) in RCA: 241] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Wnt proteins transmit myriad intercellular signals crucial for the development and homeostasis of metazoan animals from Hydra to human. Abnormal Wnt signaling causes a growing number of diseases, including cancer and osteoporosis. Depending on the context, a given Wnt signal may denote: cell proliferation or apoptosis; cell fate determination, differentiation, or stem cell maintenance; a variety of changes in cell behavior; and/or coordinated interactions with its neighbors. Which event(s) occur in Wnt-responsive cells depends critically on the ability of Dishevelled (Dsh)/Dvl proteins to interpret distinct types of intracellular, receptor-generated stimuli and transmit them to at least two distinct sets of effector molecules, all while apparently ignoring a third type of Wnt-generated Ca(2+) signal. The three conserved domains present in Dsh/Dvl proteins uniquely function in each Wnt pathway, in part by association with 18 (and counting) Dsh/Dvl-associated proteins. The latest data suggest that Dsh/Dvl proteins organize dynamic, pathway-specific subcellular signaling complexes that ensure correct information routing, signal amplification, and dynamic control through feedback regulation. The biochemical and cell biological mechanisms by which Dsh/Dvl proteins accomplish these remarkable tasks remain obscure.
Collapse
Affiliation(s)
- Keith A Wharton
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas, 75390-9072, USA.
| |
Collapse
|
47
|
Patikoglou GA, Koelle MR. An N-terminal region of Caenorhabditis elegans RGS proteins EGL-10 and EAT-16 directs inhibition of G(alpha)o versus G(alpha)q signaling. J Biol Chem 2002; 277:47004-13. [PMID: 12354761 DOI: 10.1074/jbc.m208186200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Regulator of G protein signaling (RGS) proteins contain an RGS domain that inhibits G(alpha) signaling by activating G(alpha) GTPase activity. Certain RGS proteins also contain a Ggamma-like (GGL) domain and a poorly characterized but conserved N-terminal region. We assessed the functions of these subregions in the Caenorhabditis elegans RGS proteins EGL-10 and EAT-16, which selectively inhibit GOA-1 (G(alpha)(o)) and EGL-30 (G(alpha)(q)), respectively. Using transgenes in C. elegans, we expressed EGL-10, EAT-16, their subregions, or EGL-10/EAT-16 chimeras. The chimeras showed that the GGL/RGS region of either protein can act on either GOA-1 or EGL-30 and that a key factor determining G(alpha) target selectivity is the manner in which the N-terminal and GGL/RGS regions are linked. We also found that coexpressing N-terminal and GGL/RGS fragments of EGL-10 gave full EGL-10 activity, whereas either fragment alone gave little activity. Biochemical analysis showed that coexpressing the two fragments caused both to increase in abundance and also caused the GGL/RGS fragment to move to the membrane, where the N-terminal fragment is localized. By coimmunoprecipitation, we found that the N-terminal fragment complexes with the C-terminal fragment and its associated Gbeta subunit, GPB-2. We conclude that the N-terminal region directs inhibition of G(alpha) signaling by forming a complex with the GGL/RGS region and affecting its stability, membrane localization, and G(alpha) target specificity.
Collapse
Affiliation(s)
- Georgia A Patikoglou
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520-8024, USA
| | | |
Collapse
|
48
|
Abstract
DING proteins have a characteristic DINGGG- or closely related N-terminal sequence. One is found in human synovial fluid, and may be associated with rheumatoid arthritis. Other examples have receptor or signalling roles in various human and animal cells, or are involved in biomineralisation, and several of them bind to phytochemicals. As plant DING proteins have recently been discovered, we hypothesise that the DING protein-phytochemical association may represent one aspect of a ubiquitous receptor-linked signalling system. Several microbial proteins related to DING proteins have phosphatase activity, which may relate to biomineralisation in eukaryotic systems. Plant DING proteins and their microbial relatives may elicit allergic responses leading to arthritic disease.
Collapse
Affiliation(s)
- Anne Berna
- IBMP-Institut de Botanique, Strasbourg, France
| | | | | | | |
Collapse
|
49
|
Hu G, Wensel TG. R9AP, a membrane anchor for the photoreceptor GTPase accelerating protein, RGS9-1. Proc Natl Acad Sci U S A 2002; 99:9755-60. [PMID: 12119397 PMCID: PMC125004 DOI: 10.1073/pnas.152094799] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2002] [Indexed: 11/18/2022] Open
Abstract
The regulator of G protein signaling (RGS)-9-1.G(beta 5) complex forms the GTPase accelerating protein for G(alpha t) in vertebrate photoreceptors. Although the complex is soluble when expressed in vitro, extraction of the endogenous protein from membranes requires detergents. The detergent extracts contain a complex of RGS9-1, G(beta 5), G(alpha t), and a 25-kDa phosphoprotein, R9AP (RGS9-1-Anchor Protein). R9AP is encoded by one intronless gene in both human and mouse. Full or partial cDNA or genomic clones were obtained from mice, cattle, human, zebrafish, and Xenopus laevis. R9AP mRNA was detected only in the retina, and the protein only in photoreceptors. R9AP binds to the N-terminal domain of RGS9-1, and anchors it to the disk membrane via a C-terminal transmembrane helix.
Collapse
Affiliation(s)
- Guang Hu
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | |
Collapse
|
50
|
Siderovski DP, Snow BE, Chung S, Brothers GM, Sondek J, Betts L. Assays of complex formation between RGS protein G gamma subunit-like domains and G beta subunits. Methods Enzymol 2002; 344:702-23. [PMID: 11771422 DOI: 10.1016/s0076-6879(02)44750-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- David P Siderovski
- Department of Pharmacology, UNC Neuroscience Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | |
Collapse
|