1
|
Vedeler A, Tartaglia GG, Pastore A. Annexin, a Protein for All Seasons: From Calcium Dependent Membrane Metabolism to RNA Recognition. Bioessays 2025:e70019. [PMID: 40350993 DOI: 10.1002/bies.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/15/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Annexins are a protein family well known to bind to phospholipids in a calcium-dependent way. They are involved in several different crucial cellular processes such as cell division, calcium signaling, membrane repair, vesicle trafficking, and apoptosis. Although RNA binding for some members of the family was reported long ago, it was only recently that it was shown that a common feature of the family is also the ability to bind RNA, a discovery that has added significantly to our perception of the cellular role of these proteins. In the present review, we discuss the properties of annexins under an updated light and the current knowledge on the RNA binding properties of annexins. We then focus specifically on annexin A11, because this is a less characterized member of the family but, at the same time, a potentially important component of the mRNA transport machinery in neurons. We hope to offer to the reader a more complete picture of the annexins' binding properties and new tools to evaluate the multifaceted functions of this important protein family.
Collapse
Affiliation(s)
- Anni Vedeler
- Neurotargeting Group, Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Annalisa Pastore
- Elettra Sincrotrone Trieste, Basovizza, Italy
- The Wohl Institute, King's College London, London, UK
| |
Collapse
|
2
|
Prislusky MI, Lam JGT, Contreras VR, Ng M, Chamberlain M, Pathak-Sharma S, Fields M, Zhang X, Amer AO, Seveau S. The septin cytoskeleton is required for plasma membrane repair. EMBO Rep 2024; 25:3870-3895. [PMID: 38969946 PMCID: PMC11387490 DOI: 10.1038/s44319-024-00195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 07/07/2024] Open
Abstract
Plasma membrane repair is a fundamental homeostatic process of eukaryotic cells. Here, we report a new function for the conserved cytoskeletal proteins known as septins in the repair of cells perforated by pore-forming toxins or mechanical disruption. Using a silencing RNA screen, we identified known repair factors (e.g. annexin A2, ANXA2) and novel factors such as septin 7 (SEPT7) that is essential for septin assembly. Upon plasma membrane injury, the septin cytoskeleton is extensively redistributed to form submembranous domains arranged as knob and loop structures containing F-actin, myosin IIA, S100A11, and ANXA2. Formation of these domains is Ca2+-dependent and correlates with plasma membrane repair efficiency. Super-resolution microscopy revealed that septins and F-actin form intertwined filaments associated with ANXA2. Depletion of SEPT7 prevented ANXA2 recruitment and formation of submembranous actomyosin domains. However, ANXA2 depletion had no effect on domain formation. Collectively, our data support a novel septin-based mechanism for resealing damaged cells, in which the septin cytoskeleton plays a key structural role in remodeling the plasma membrane by promoting the formation of SEPT/F-actin/myosin IIA/ANXA2/S100A11 repair domains.
Collapse
Affiliation(s)
- M Isabella Prislusky
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jonathan G T Lam
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Viviana Ruiz Contreras
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- Grupo Investigaciones Biomédicas, Universidad de Sucre, Sincelejo, Sucre, Colombia
| | - Marilynn Ng
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Madeline Chamberlain
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Sarika Pathak-Sharma
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Madalyn Fields
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Xiaoli Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amal O Amer
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Stephanie Seveau
- Department of Microbial Infection & Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
3
|
Prislusky MI, Lam JG, Contreras VR, Ng M, Chamberlain M, Pathak-Sharma S, Fields M, Zhang X, Amer AO, Seveau S. The Septin Cytoskeleton is Required for Plasma Membrane Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.12.548547. [PMID: 37503091 PMCID: PMC10369955 DOI: 10.1101/2023.07.12.548547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Mammalian cells are frequently exposed to mechanical and biochemical stressors resulting in plasma membrane injuries. Repair mechanisms reseal the plasma membrane to restore homeostasis and prevent cell death. In the present work, a silencing RNA screen was performed to uncover plasma membrane repair mechanisms of cells exposed to a pore-forming toxin (listeriolysin O). This screen identified molecules previously known to repair the injured plasma membrane such as annexin A2 (ANXA2) as well as novel plasma membrane repair candidate proteins. Of the novel candidates, we focused on septin 7 (SEPT7) because the septins are an important family of conserved eukaryotic cytoskeletal proteins. Using diverse experimental approaches, we established for the first time that SEPT7 plays a general role in plasma membrane repair of cells perforated by pore-forming toxins and mechanical wounding. Remarkably, upon cell injury, the septin cytoskeleton is extensively redistributed in a Ca 2+ -dependent fashion, a hallmark of plasma membrane repair machineries. The septins reorganize into subplasmalemmal domains arranged as knob and loop (or ring) structures containing F-actin, myosin II, and annexin A2 (ANXA2) and protrude from the cell surface. Importantly, the formation of these domains correlates with the plasma membrane repair efficiency. Super-resolution microscopy shows that septins and actin are arranged in intertwined filaments associated with ANXA2. Silencing SEPT7 expression prevented the formation of the F-actin/myosin II/ANXA2 domains, however, silencing expression of ANXA2 had no observable effect on their formation. These results highlight the key structural role of the septins in remodeling the plasma membrane and in the recruitment of the repair molecule ANXA2. Collectively, our data support a novel model in which the septin cytoskeleton acts as a scaffold to promote the formation of plasma membrane repair domains containing contractile F-actin and annexin A2.
Collapse
|
4
|
Yu C, Nelson SL, Meisl G, Ghirlando R, Deshmukh L. Phase Separation and Fibrillization of Human Annexin A7 Are Mediated by Its Proline-Rich Domain. Biochemistry 2023; 62:3036-3040. [PMID: 37788367 PMCID: PMC10634317 DOI: 10.1021/acs.biochem.3c00349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/26/2023] [Indexed: 10/05/2023]
Abstract
Human annexin A7, a calcium- and phospholipid-binding protein, governs calcium homeostasis, plasma membrane repair, apoptosis, and tumor progression. A7 contains an N-terminal proline-rich domain (PRD; 180 residues, ∼24% prolines) that determines its functional specificity. Using microscopy and dye-binding assays, we show that recombinant A7 and its isolated PRD spontaneously phase separate into spherical condensates, which subsequently transform into β-sheet-rich fibrils. We demonstrate that fibrillization of A7-PRD proceeds via primary nucleation and fibril-catalyzed secondary nucleation processes, as determined by chemical kinetics, providing a mechanistic basis for its amyloid assembly. This study confirms and highlights a subclass of eukaryotic PRDs prone to forming aggregates with important physiological and pathological implications.
Collapse
Affiliation(s)
- Chenrong Yu
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| | - Spencer L. Nelson
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| | - Georg Meisl
- Department
of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Rodolfo Ghirlando
- Laboratory
of Molecular Biology, National Institute
of Diabetes and Digestive and Kidney Diseases, National
Institutes of Health, Bethesda, Maryland 20892, United States
| | - Lalit Deshmukh
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
5
|
Drescher DG, Drescher MJ, Selvakumar D, Annam NP. Analysis of Dysferlin Direct Interactions with Putative Repair Proteins Links Apoptotic Signaling to Ca 2+ Elevation via PDCD6 and FKBP8. Int J Mol Sci 2023; 24:4707. [PMID: 36902136 PMCID: PMC10002499 DOI: 10.3390/ijms24054707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Quantitative surface plasmon resonance (SPR) was utilized to determine binding strength and calcium dependence of direct interactions between dysferlin and proteins likely to mediate skeletal muscle repair, interrupted in limb girdle muscular dystrophy type 2B/R2. Dysferlin canonical C2A (cC2A) and C2F/G domains directly interacted with annexin A1, calpain-3, caveolin-3, affixin, AHNAK1, syntaxin-4, and mitsugumin-53, with cC2A the primary target and C2F lesser involved, overall demonstrating positive calcium dependence. Dysferlin C2 pairings alone showed negative calcium dependence in almost all cases. Like otoferlin, dysferlin directly interacted via its carboxy terminus with FKBP8, an anti-apoptotic outer mitochondrial membrane protein, and via its C2DE domain with apoptosis-linked gene (ALG-2/PDCD6), linking anti-apoptosis with apoptosis. Confocal Z-stack immunofluorescence confirmed co-compartmentalization of PDCD6 and FKBP8 at the sarcolemmal membrane. Our evidence supports the hypothesis that prior to injury, dysferlin C2 domains self-interact and give rise to a folded, compact structure as indicated for otoferlin. With elevation of intracellular Ca2+ in injury, dysferlin would unfold and expose the cC2A domain for interaction with annexin A1, calpain-3, mitsugumin 53, affixin, and caveolin-3, and dysferlin would realign from its interactions with PDCD6 at basal calcium levels to interact strongly with FKBP8, an intramolecular rearrangement facilitating membrane repair.
Collapse
Affiliation(s)
- Dennis G. Drescher
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marian J. Drescher
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Dakshnamurthy Selvakumar
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Neeraja P. Annam
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
6
|
Jiang Q, Lin J, Wei Q, Li C, Hou Y, Cao B, Zhang L, Ou R, Liu K, Yang T, Xiao Y, Shang H. Genetic analysis of and clinical characteristics associated with ANXA11 variants in a Chinese cohort with amyotrophic lateral sclerosis. Neurobiol Dis 2022; 175:105907. [DOI: 10.1016/j.nbd.2022.105907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
|
7
|
Wang X, Wu F, Wang H, Duan X, Huang R, Tuersuntuoheti A, Su L, Yan S, Zhao Y, Lu Y, Li K, Yao J, Luo Z, Guo L, Liu J, Chen X, Lu Y, Hu H, Li X, Bao M, Bi X, Du B, Miao S, Cai J, Wang L, Zhou H, Ying J, Song W, Zhao H. PDCD6 cooperates with C-Raf to facilitate colorectal cancer progression via Raf/MEK/ERK activation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:147. [PMID: 32746883 PMCID: PMC7398064 DOI: 10.1186/s13046-020-01632-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023]
Abstract
Background Colorectal cancer (CRC) is one of the most common malignancies, and it’s expected that the CRC burden will substantially increase in the next two decades. New biomarkers for targeted treatment and associated molecular mechanism of tumorigenesis remain to be explored. In this study, we investigated whether PDCD6 plays an oncogenic role in colorectal cancer and its underlying mechanism. Methods Programmed cell death protein 6 (PDCD6) expression in CRC samples were analyzed by immunohistochemistry and immunofluorescence. The prognosis between PDCD6 and clinical features were analyzed. The roles of PDCD6 in cellular proliferation and tumor growth were measured by using CCK8, colony formation, and tumor xenograft in nude mice. RNA-sequence (RNA-seq), Mass Spectrum (MS), Co-Immunoprecipitation (Co-IP) and Western blot were utilized to investigate the mechanism of tumor progression. Immunohistochemistry (IHC) and quantitative real-time PCR (qRT-PCR) were performed to determine the correlation of PDCD6 and MAPK pathway. Results Higher expression levels of PDCD6 in tumor tissues were associated with a poorer prognosis in patients with CRC. Furthermore, PDCD6 increased cell proliferation in vitro and tumor growth in vivo. Mechanistically, RNA-seq showed that PDCD6 could affect the activation of the MAPK signaling pathway. PDCD6 interacted with c-Raf, resulting in the activation of downstream c-Raf/MEK/ERK pathway and the upregulation of core cell proliferation genes such as MYC and JUN. Conclusions These findings reveal the oncogenic effect of PDCD6 in CRC by activating c-Raf/MEK/ERK pathway and indicate that PDCD6 might be a potential prognostic indicator and therapeutic target for patients with colorectal cancer.
Collapse
Affiliation(s)
- Xiaojuan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.,State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, TsinghuaUniversity, Beijing, 100084, China
| | - Fan Wu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Han Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Xiaoyuan Duan
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Rong Huang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Amannisa Tuersuntuoheti
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Luying Su
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Shida Yan
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuechao Zhao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Yan Lu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Kai Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Jinjie Yao
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiwen Luo
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Guo
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianmei Liu
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiao Chen
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yalan Lu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Hanjie Hu
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xingchen Li
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mandula Bao
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinyu Bi
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Boyu Du
- Department of Medical Biology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Shiying Miao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Linfang Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Haitao Zhou
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Wei Song
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Hong Zhao
- Department of Hepatobiliary Surgery and Department of Pathology, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,Key Laboratory of Gene Editing Screening and R&D of Digestive System Tumor Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
Maki M. Structures and functions of penta-EF-hand calcium-binding proteins and their interacting partners: enigmatic relationships between ALG-2 and calpain-7. Biosci Biotechnol Biochem 2019; 84:651-660. [PMID: 31814542 DOI: 10.1080/09168451.2019.1700099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The penta-EF-hand (PEF) protein family includes ALG-2 (gene name, PDCD6) and its paralogs as well as classical calpain family members. ALG-2 is a prototypic PEF protein that is widely distributed in eukaryotes and interacts with a variety of proteins in a Ca2+-dependent manner. Mammalian ALG-2 and its interacting partners have various modulatory roles including roles in cell death, signal transduction, membrane repair, ER-to-Golgi vesicular transport, and RNA processing. Some ALG-2-interacting proteins are key factors that function in the endosomal sorting complex required for transport (ESCRT) system. On the other hand, mammalian calpain-7 (CAPN7) lacks the PEF domain but contains two microtubule-interacting and trafficking (MIT) domains in tandem. CAPN7 interacts with a subset of ESCRT-III proteins through the MIT domains and regulates EGF receptor downregulation. Structures and functions of ALG-2 and those of its interacting partners as well as relationships with the calpain family are reviewed in this article.
Collapse
Affiliation(s)
- Masatoshi Maki
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
9
|
Mo HQ, Tian FJ, Li X, Zhang J, Ma XL, Zeng WH, Lin Y, Zhang Y. ANXA7 regulates trophoblast proliferation and apoptosis in preeclampsia. Am J Reprod Immunol 2019; 82:e13183. [PMID: 31446642 DOI: 10.1111/aji.13183] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/06/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022] Open
Abstract
PROBLEM Preeclampsia (PE) is a unique gestational disorder leading to maternal and neonatal morbidity and mortality. AnnexinA7 (ANXA7) is a calcium-dependent phospholipid-binding protein that promotes membrane fusion during exocytosis. However, the function of ANXA7 in placental trophoblast is poorly understood. The present study aimed to investigate a possible association between ANXA7 and human trophoblast apoptosis. METHODS We collected human placental tissues from patients with PE and normal pregnant women to elucidate the expression level of ANXA7. The ANXA7-knockdown and ANXA7-overexpressing HTR8/SVneo cells were utilized for studying the function of ANXA7 in trophoblast. The proliferation and apoptosis levels of trophoblast were examined with Western blot assay, flow cytometry, Cell Counting Kit-8 assay, and immunohistochemistry. RESULTS ANXA7 expression was significantly lower in placentas from patients with PE patients compared with that in from normal pregnant controls. Knockdown of ANXA7 induced cell apoptosis and inhibited cell proliferation in HTR-8 via by downregulating BCL2 protein levels. Overexpression of ANXA7 reduced apoptosis and promoted HTR8 proliferation. Further analyses showed that ANXA7 knockdown inhibited the activation of the JAK1/STAT3 pathway in HTR-8 cells. CONCLUSION Our findings revealed a new regulatory pathway of ANXA7/JAK1/STAT3 in trophoblast apoptosis in preeclampsia, suggesting that ANXA7 is a potential therapeutic target for preeclampsia.
Collapse
Affiliation(s)
- Hui-Qin Mo
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu-Ju Tian
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Li
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhang
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Ling Ma
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Hong Zeng
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Lin
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, RenMin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Shen TC, Chang WS, Hsia TC, Li HT, Chen WC, Tsai CW, Bau DT. Contribution of programmed cell death 6 genetic variations, gender, and smoking status to lung cancer. Onco Targets Ther 2019; 12:6237-6244. [PMID: 31496727 PMCID: PMC6693085 DOI: 10.2147/ott.s205544] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/16/2019] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Programmed cell death 6 (PDCD6) is a calcium sensor participating in T-cell receptor-, Fas-, and glucocorticoid-induced programmed cell death. At the sites of lung tumors, the expression of PDCD6 is higher than that in non-tumor tissues. However, the contribution of variant PDCD6 genotypes to lung cancer is largely unknown. The current study aimed to evaluate the contributions of the PDCD6 rs4957014 and rs3756712 genotypes to the risk of lung cancer. PATIENTS AND METHODS The contributions of PDCD6 genotypes to lung cancer risk were examined among 358 patients with lung cancer and 716 age- and gender-matched healthy controls by typical polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) methodology. RESULTS The results showed that the GG but not the GT genotype of PDCD6 rs4957014 was associated with a decreased risk of lung cancer (odds ratio (OR) =0.41, 95% confidence interval (CI) =0.23-0.72, p=0.0013). The analysis of allelic frequency distributions showed that the G allele of PDCD6 rs4957014 decreased lung cancer susceptibility (p=0.0090). There was no association between PDCD6 rs3756712 genotypes and lung cancer risk. Interestingly, the GG genotype at PDCD6 rs4957014 significantly decreased the risk of lung cancer among males (adjusted OR =0.29, 95% CI =0.14-0.57) and smokers (adjusted OR =0.34, 95% CI =0.18-0.61) but not among females and non-smokers. CONCLUSION The GG genotype of PDCD6 rs4957014 may decrease lung cancer risk in males and smokers and may serve as a practical marker for early detection and the incidence of lung cancer in Taiwan.
Collapse
Affiliation(s)
- Te-Chun Shen
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Te-Chun Hsia
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Respiratory Therapy, China Medical University, Taichung, Taiwan
| | - Hsin-Ting Li
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Chun Chen
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Respiratory Therapy, China Medical University, Taichung, Taiwan
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Da-Tian Bau
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
11
|
Koerdt SN, Ashraf APK, Gerke V. Annexins and plasma membrane repair. CURRENT TOPICS IN MEMBRANES 2019; 84:43-65. [PMID: 31610865 DOI: 10.1016/bs.ctm.2019.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Plasma membrane wound repair is a cell-autonomous process that is triggered by Ca2+ entering through the site of injury and involves membrane resealing, i.e., re-establishment of a continuous plasma membrane, as well as remodeling of the cortical actin cytoskeleton. Among other things, the injury-induced Ca2+ elevation initiates the wound site recruitment of Ca2+-regulated proteins that function in the course of repair. Annexins are a class of such Ca2+-regulated proteins. They associate with acidic phospholipids of cellular membranes in their Ca2+ bound conformation with Ca2+ sensitivities ranging from the low to high micromolar range depending on the respective annexin protein. Annexins accumulate at sites of plasma membrane injury in a temporally controlled manner and are thought to function by controlling membrane rearrangements at the wound site, most likely in conjunction with other repair proteins such as dysferlin. Their role in membrane repair, which has been evidenced in several model systems, will be discussed in this chapter.
Collapse
Affiliation(s)
- Sophia N Koerdt
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Arsila P K Ashraf
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Münster, Münster, Germany.
| |
Collapse
|
12
|
Sønder SL, Boye TL, Tölle R, Dengjel J, Maeda K, Jäättelä M, Simonsen AC, Jaiswal JK, Nylandsted J. Annexin A7 is required for ESCRT III-mediated plasma membrane repair. Sci Rep 2019; 9:6726. [PMID: 31040365 PMCID: PMC6491720 DOI: 10.1038/s41598-019-43143-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/15/2019] [Indexed: 12/21/2022] Open
Abstract
The plasma membrane of eukaryotic cells forms the essential barrier to the extracellular environment, and thus plasma membrane disruptions pose a fatal threat to cells. Here, using invasive breast cancer cells we show that the Ca2+ - and phospholipid-binding protein annexin A7 is part of the plasma membrane repair response by enabling assembly of the endosomal sorting complex required for transport (ESCRT) III. Following injury to the plasma membrane and Ca2+ flux into the cytoplasm, annexin A7 forms a complex with apoptosis linked gene-2 (ALG-2) to facilitate proper recruitment and binding of ALG-2 and ALG-2-interacting protein X (ALIX) to the damaged membrane. ALG-2 and ALIX assemble the ESCRT III complex, which helps excise and shed the damaged portion of the plasma membrane during wound healing. Our results reveal a novel function of annexin A7 – enabling plasma membrane repair by regulating ESCRT III-mediated shedding of injured plasma membrane.
Collapse
Affiliation(s)
- Stine Lauritzen Sønder
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Theresa Louise Boye
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Regine Tölle
- Department of Dermatology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Department of Biology, University of Fribourg Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Jörn Dengjel
- Department of Dermatology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Department of Biology, University of Fribourg Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Kenji Maeda
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Marja Jäättelä
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark.,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Adam Cohen Simonsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230, Odense M, Denmark
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, George Washington University School of Medicine and Health Sciences, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA
| | - Jesper Nylandsted
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark. .,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
13
|
Brito C, Cabanes D, Sarmento Mesquita F, Sousa S. Mechanisms protecting host cells against bacterial pore-forming toxins. Cell Mol Life Sci 2019; 76:1319-1339. [PMID: 30591958 PMCID: PMC6420883 DOI: 10.1007/s00018-018-2992-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022]
Abstract
Pore-forming toxins (PFTs) are key virulence determinants produced and secreted by a variety of human bacterial pathogens. They disrupt the plasma membrane (PM) by generating stable protein pores, which allow uncontrolled exchanges between the extracellular and intracellular milieus, dramatically disturbing cellular homeostasis. In recent years, many advances were made regarding the characterization of conserved repair mechanisms that allow eukaryotic cells to recover from mechanical disruption of the PM membrane. However, the specificities of the cell recovery pathways that protect host cells against PFT-induced damage remain remarkably elusive. During bacterial infections, the coordinated action of such cell recovery processes defines the outcome of infected cells and is, thus, critical for our understanding of bacterial pathogenesis. Here, we review the cellular pathways reported to be involved in the response to bacterial PFTs and discuss their impact in single-cell recovery and infection.
Collapse
Affiliation(s)
- Cláudia Brito
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Didier Cabanes
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Francisco Sarmento Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- Global Health Institute, School of Life Science, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
| |
Collapse
|
14
|
Thermodynamic Characterization of the Ca 2+-Dependent Interaction Between SOUL and ALG-2. Int J Mol Sci 2018; 19:ijms19123802. [PMID: 30501057 PMCID: PMC6321638 DOI: 10.3390/ijms19123802] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 11/17/2022] Open
Abstract
SOUL, a heme-binding protein-2 (HEBP-2), interacts with apoptosis-linked gene 2 protein (ALG-2) in a Ca2+-dependent manner. To investigate the properties of the interaction of SOUL with ALG-2, we generated several mutants of SOUL and ALG-2 and analyzed the recombinant proteins using pulldown assay and isothermal titration calorimetry. The interaction between SOUL and ALG-2 (delta3-23ALG-2) was an exothermic reaction, with 1:1 stoichiometry and high affinity (Kd = 32.4 nM) in the presence of Ca2+. The heat capacity change (ΔCp) of the reaction showed a large negative value (−390 cal/K·mol), which suggested the burial of a significant nonpolar surface area or disruption of a hydrogen bond network that was induced by the interaction (or both). One-point mutation of SOUL Phe100 or ALG-2 Trp57 resulted in complete loss of heat change, supporting the essential roles of these residues for the interaction. Nevertheless, a truncated mutant of SOUL1-143 that deleted the domain required for the interaction with ALG-2 Trp57 still showed 1:1 binding to ALG-2 with an endothermic reaction. These results provide a better understanding of the target recognition mechanism and conformational change of SOUL in the interaction with ALG-2.
Collapse
|
15
|
Shibata H. Adaptor functions of the Ca 2+-binding protein ALG-2 in protein transport from the endoplasmic reticulum. Biosci Biotechnol Biochem 2018; 83:20-32. [PMID: 30259798 DOI: 10.1080/09168451.2018.1525274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Apoptosis-linked gene 2 (ALG-2) is a Ca2+-binding protein with five repetitive EF-hand motifs, named penta-EF-hand (PEF) domain. It interacts with various target proteins and functions as a Ca2+-dependent adaptor in diverse cellular activities. In the cytoplasm, ALG-2 is predominantly localized to a specialized region of the endoplasmic reticulum (ER), called the ER exit site (ERES), through its interaction with Sec31A. Sec31A is an outer coat protein of coat protein complex II (COPII) and is recruited from the cytosol to the ERES to form COPII-coated transport vesicles. I will overview current knowledge of the physiological significance of ALG-2 in regulating ERES localization of Sec31A and the following adaptor functions of ALG-2, including bridging Sec31A and annexin A11 to stabilize Sec31A at the ERES, polymerizing the Trk-fused gene (TFG) product, and linking MAPK1-interacting and spindle stabilizing (MISS)-like (MISSL) and microtubule-associated protein 1B (MAP1B) to promote anterograde transport from the ER.
Collapse
Affiliation(s)
- Hideki Shibata
- a Department of Applied Biosciences, Graduate School of Bioagricultural Sciences , Nagoya University , Chikusa-ku , Nagoya , Japan
| |
Collapse
|
16
|
Enrich C, Rentero C, Meneses-Salas E, Tebar F, Grewal T. Annexins: Ca 2+ Effectors Determining Membrane Trafficking in the Late Endocytic Compartment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 981:351-385. [PMID: 29594868 DOI: 10.1007/978-3-319-55858-5_14] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite the discovery of annexins 40 years ago, we are just beginning to understand some of the functions of these still enigmatic proteins. Defined and characterized by their ability to bind anionic membrane lipids in a Ca2+-dependent manner, each annexin has to be considered a multifunctional protein, with a multitude of cellular locations and diverse activities. Underlying causes for this considerable functional diversity include their capability to associate with multiple cytosolic and membrane proteins. In recent years, the increasingly recognized establishment of membrane contact sites between subcellular compartments opens a new scenario for annexins as instrumental players to link Ca2+ signalling with the integration of membrane trafficking in many facets of cell physiology. In this chapter, we review and discuss current knowledge on the contribution of annexins in the biogenesis and functioning of the late endocytic compartment, affecting endo- and exocytic pathways in a variety of physiological consequences ranging from membrane repair, lysosomal exocytosis, to cell migration.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Elsa Meneses-Salas
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, Australia
| |
Collapse
|
17
|
Kanadome T, Shibata H, Kuwata K, Takahara T, Maki M. The calcium-binding protein ALG-2 promotes endoplasmic reticulum exit site localization and polymerization of Trk-fused gene (TFG) protein. FEBS J 2017; 284:56-76. [PMID: 27813252 DOI: 10.1111/febs.13949] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/28/2016] [Accepted: 11/01/2016] [Indexed: 12/27/2022]
Abstract
Apoptosis-linked gene 2 (ALG-2), which is a gene product of PDCD6, is a 22-kDa Ca2+ -binding protein. Accumulating evidence points to a role for ALG-2 as a Ca2+ -responsive adaptor protein. On binding to Ca2+ , ALG-2 undergoes a conformational change that facilitates its interaction with various proteins. It also forms a homodimer and heterodimer with peflin, a paralog of ALG-2. However, the differences in cellular roles for the ALG-2 homodimer and ALG-2/peflin heterodimer are unclear. In the present study, we found that Trk-fused gene (TFG) protein interacted with the ALG-2 homodimer. Immunostaining analysis revealed that TFG and ALG-2 partially overlapped at endoplasmic reticulum exit sites (ERES), a platform for COPII-mediated protein transport from the endoplasmic reticulum. Time-lapse live-cell imaging demonstrated that both green fluorescent protein-fused TFG and mCherry-fused ALG-2 are recruited to ERES after thapsigargin treatment, which raises intracellular Ca2+ levels. Furthermore, overexpression of ALG-2 induced the accumulation of TFG at ERES. TFG has an ALG-2-binding motif and deletion of the motif decreased TFG binding to ALG-2 and shortened its half-life at ERES, suggesting a critical role for ALG-2 in retaining TFG at ERES. We also demonstrated, by in vitro cross-linking assays, that ALG-2 promoted the polymerization of TFG in a Ca2+ -dependent manner. Collectively, the results suggest that ALG-2 acts as a Ca2+ -sensitive adaptor to concentrate and polymerize TFG at ERES, supporting a potential role for ALG-2 in COPII-dependent trafficking from the endoplasmic reticulum.
Collapse
Affiliation(s)
- Takashi Kanadome
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Japan
| | - Hideki Shibata
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Japan
| | - Keiko Kuwata
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Japan
| | - Terunao Takahara
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Japan
| | - Masatoshi Maki
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Japan
| |
Collapse
|
18
|
Tanner JJ, Frey BB, Pemberton T, Henzl MT. EF5 Is the High-Affinity Mg2+ Site in ALG-2. Biochemistry 2016; 55:5128-41. [DOI: 10.1021/acs.biochem.6b00596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- John J. Tanner
- Department
of Biochemistry, University of Missouri, Columbia, Missouri 65211, United States
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Benjamin B. Frey
- Department
of Biochemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Travis Pemberton
- Department
of Chemistry, University of Missouri, Columbia, Missouri 65211, United States
| | - Michael T. Henzl
- Department
of Biochemistry, University of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
19
|
Maki M, Takahara T, Shibata H. Multifaceted Roles of ALG-2 in Ca(2+)-Regulated Membrane Trafficking. Int J Mol Sci 2016; 17:ijms17091401. [PMID: 27571067 PMCID: PMC5037681 DOI: 10.3390/ijms17091401] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/15/2022] Open
Abstract
ALG-2 (gene name: PDCD6) is a penta-EF-hand Ca2+-binding protein and interacts with a variety of proteins in a Ca2+-dependent fashion. ALG-2 recognizes different types of identified motifs in Pro-rich regions by using different hydrophobic pockets, but other unknown modes of binding are also used for non-Pro-rich proteins. Most ALG-2-interacting proteins associate directly or indirectly with the plasma membrane or organelle membranes involving the endosomal sorting complex required for transport (ESCRT) system, coat protein complex II (COPII)-dependent ER-to-Golgi vesicular transport, and signal transduction from membrane receptors to downstream players. Binding of ALG-2 to targets may induce conformational change of the proteins. The ALG-2 dimer may also function as a Ca2+-dependent adaptor to bridge different partners and connect the subnetwork of interacting proteins.
Collapse
Affiliation(s)
- Masatoshi Maki
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| | - Terunao Takahara
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| | - Hideki Shibata
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| |
Collapse
|
20
|
Wolfmeier H, Radecke J, Schoenauer R, Koeffel R, Babiychuk VS, Drücker P, Hathaway LJ, Mitchell TJ, Zuber B, Draeger A, Babiychuk EB. Active release of pneumolysin prepores and pores by mammalian cells undergoing a Streptococcus pneumoniae attack. Biochim Biophys Acta Gen Subj 2016; 1860:2498-2509. [PMID: 27481675 DOI: 10.1016/j.bbagen.2016.07.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 07/15/2016] [Accepted: 07/24/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Streptococcus pneumoniae is a potent human pathogen. Its pore-forming exotoxin pneumolysin is instrumental for breaching the host's epithelial barrier and for the incapacitation of the immune system. METHODS AND RESULTS Using a combination of life imaging and cryo-electron microscopy we show that pneumolysin, released by cultured bacteria, is capable of permeabilizing the plasmalemma of host cells. However, such permeabilization does not lead to cell lysis since pneumolysin is actively removed by the host cells. The process of pore elimination starts with the formation of pore-bearing plasmalemmal nanotubes and proceeds by the shedding of pores that are embedded in the membrane of released microvesicles. Pneumolysin prepores are likewise removed. The protein composition of the toxin-induced microvesicles, assessed by mass spectrometry, is suggestive of a Ca(2+)-triggered mechanism encompassing the proteins of the annexin family and members of the endosomal sorting complex required for transport (ESCRT) complex. CONCLUSIONS S. pneumoniae releases sufficient amounts of pneumolysin to perforate the plasmalemma of host cells, however, the immediate cell lysis, which is frequently reported as a result of treatment with purified and artificially concentrated toxin, appears to be an unlikely event in vivo since the toxin pores are efficiently eliminated by microvesicle shedding. Therefore the dysregulation of cellular homeostasis occurring as a result of transient pore formation/elimination should be held responsible for the damaging toxin action. GENERAL SIGNIFICANCE We have achieved a comprehensive view of a general plasma membrane repair mechanism after injury by a major bacterial toxin.
Collapse
Affiliation(s)
- Heidi Wolfmeier
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Julika Radecke
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3000 Bern 9, Switzerland
| | - Roman Schoenauer
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - René Koeffel
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Viktoria S Babiychuk
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Patrick Drücker
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Lucy J Hathaway
- Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, Postfach, 3001, Bern, Switzerland
| | - Timothy J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Benoît Zuber
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland.
| | - Annette Draeger
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland
| | - Eduard B Babiychuk
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland.
| |
Collapse
|
21
|
Liu S, Wang Z, Miao J. Potential roles of annexin A7 GTPase in autophagy, senescence and apoptosis. RSC Adv 2016. [DOI: 10.1039/c6ra21736b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This review covers the roles of ANXA7 GTPase in orchestrating autophagy, senescence and apoptosis interactive networks in various cell types.
Collapse
Affiliation(s)
- ShuYan Liu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology
- School of Life Science
- Shandong University
- Jinan 250100
- China
| | - ZhaoYang Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology
- School of Life Science
- Shandong University
- Jinan 250100
- China
| | - JunYing Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology
- School of Life Science
- Shandong University
- Jinan 250100
- China
| |
Collapse
|
22
|
Henzl MT, Frey BB, Wolf AJ. ALG-2 divalent-ion affinity: Calorimetric analysis of the des23 versions reveals high-affinity site for Mg(2). Biophys Chem 2015; 209:28-40. [PMID: 26705706 DOI: 10.1016/j.bpc.2015.10.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/27/2015] [Indexed: 12/29/2022]
Affiliation(s)
- Michael T Henzl
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211.
| | - Benjamin B Frey
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211
| | - Andrew J Wolf
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211
| |
Collapse
|
23
|
Calcium signaling in membrane repair. Semin Cell Dev Biol 2015; 45:24-31. [PMID: 26519113 DOI: 10.1016/j.semcdb.2015.10.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/20/2015] [Accepted: 10/20/2015] [Indexed: 11/21/2022]
Abstract
Resealing allows cells to mend damaged membranes rapidly when plasma membrane (PM) disruptions occur. Models of PM repair mechanisms include the "lipid-patch", "endocytic removal", and "macro-vesicle shedding" models, all of which postulate a dependence on local increases in intracellular Ca(2+) at injury sites. Multiple calcium sensors, including synaptotagmin (Syt) VII, dysferlin, and apoptosis-linked gene-2 (ALG-2), are involved in PM resealing, suggesting that Ca(2+) may regulate multiple steps of the repair process. Although earlier studies focused exclusively on external Ca(2+), recent studies suggest that Ca(2+) release from intracellular stores may also be important for PM resealing. Hence, depending on injury size and the type of injury, multiple sources of Ca(2+) may be recruited to trigger and orchestrate repair processes. In this review, we discuss the mechanisms by which the resealing process is promoted by vesicular Ca(2+) channels and Ca(2+) sensors that accumulate at damage sites.
Collapse
|
24
|
Plasma membrane and cytoskeleton dynamics during single-cell wound healing. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015. [DOI: 10.1016/j.bbamcr.2015.07.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
25
|
Huang Y, Du Y, Zhang X, Bai L, Mibrahim M, Zhang J, Wei Y, Li C, Fan S, Wang H, Zhao Z, Tang J. Down-regulated expression of Annexin A7 induces apoptosis in mouse hepatocarcinoma cell line by the intrinsic mitochondrial pathway. Biomed Pharmacother 2015; 70:146-50. [PMID: 25776493 DOI: 10.1016/j.biopha.2015.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/04/2015] [Indexed: 12/21/2022] Open
Abstract
Our previous studies have shown that decreased expression of Annexin A7 elevates apoptosis in Hca-P cells, a hepatocarcinoma cell line with lymphatic metastatic potential. In this study, RNA interference technique was used to down-regulate the expression of Annexin A7, and unmanipulated Hca-P cells and transfected nonspecific-sequence Hca-P cells as control. The down-regulation of Annexin A7 declined the cell viability after cisplatin exposure. And the reduced expression of Annexin A7 decreased the expression of Bcl2, increased the expression of Cytochrome-C in the cytoplasme, and then improved the expression of Caspase-3. However there was no significant effect on the expression of Bax, Caspase-12, Fas, FasL and Caspase-8. The results indicate that the decreased expression of Annexin A7 could inhibit the proliferation, and increase the apoptosis of Hca-P cells by affecting the expression of the apoptosis associated proteins by the mitochondrial pathway.
Collapse
Affiliation(s)
- Yuhong Huang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China.
| | - Yue Du
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Xin Zhang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Lulu Bai
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Mohmmed Mibrahim
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Jun Zhang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Yuanyi Wei
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Cong Li
- Department of Pathophysiology, Dalian Medical University, Dalian, PR China
| | - Shujun Fan
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Huaxin Wang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Zhiying Zhao
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China
| | - Jianwu Tang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis of Liaoning Province, Dalian, PR China.
| |
Collapse
|
26
|
Wang J, Guo C, Liu S, Qi H, Yin Y, Liang R, Sun MZ, Greenaway FT. Annexin A11 in disease. Clin Chim Acta 2014; 431:164-8. [PMID: 24508622 DOI: 10.1016/j.cca.2014.01.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/16/2014] [Accepted: 01/18/2014] [Indexed: 12/28/2022]
Abstract
Ubiquitously expressed in many cell types, annexin A11 (Anxa11) is a member of the multigene family of Ca(2+)-regulated phospholipid-dependent and membrane-binding annexin proteins. Studies have shown that Anxa11 plays an important role in cell division, Ca(2+) signaling, vesicle trafficking and apoptosis. The deregulation and mutation of Anxa11 are involved in systemic autoimmune diseases, sarcoidosis and the development, chemoresistance and recurrence of cancers. Malfunction of Anxa11 may lead to or enhance the metastasis, invasion and drug resistance of cancers through the platelet-derived growth factor receptor (PDGFR) pathway and/or the mitogen-activated protein kinase (MAPK)/p53 pathway. In a variety of diseases, Anxa11 is most commonly reported to function through interactions with apoptosis-linked gene-2 protein (ALG-2) and/or calcyclin (S100A6). Although it has been little studied, Anxa11 is a promising biomarker for the diagnosis, treatment and prognosis of certain diseases. In this review, the associations of Anxa11 with Ca(2+)-regulated exocytosis, cytokinesis, sex differentiation, autoimmune diseases, thrombolysis and cancers are summarized and interpreted.
Collapse
Affiliation(s)
- Jiasheng Wang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Chunmei Guo
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Shuqing Liu
- Department of Biochemistry, Dalian Medical University, Dalian 116044, China
| | - Houbao Qi
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Yuling Yin
- Department of Biochemistry, Dalian Medical University, Dalian 116044, China
| | - Rui Liang
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian 116027, China
| | - Ming-Zhong Sun
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| | - Frederick T Greenaway
- Carlson School of Chemistry and Biochemistry, Clark University, Worcester, MA 01610, USA
| |
Collapse
|
27
|
Identification of a small molecule targeting annexin A7. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2092-9. [PMID: 23651924 DOI: 10.1016/j.bbamcr.2013.04.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 11/21/2022]
Abstract
Autophagy involves multiple membrane trafficking and fusion events. Annexin A7 (ANXA7) is postulated to play a role in membrane fusion during exocytosis, while the contribution of ANXA7 to autophagy is poorly understood. Our recent studies demonstrated that ABO could promote autophagy via elevation of ANXA7 and triggering ANXA7 subcellular redistribution. However, little is known about the molecular mechanisms how ANXA7 regulates autophagy. As molecular disruption of ANXA7 in mice results in several unwished phenotypes, small molecule modulators may be efficacious in defining the mechanisms of ANXA7 action. However, so far no compounds that selectively target ANXA7 have been identified. So, we hypothesize that ABO might be a potent modulator of ANXA7. We also have detected the colocalization of ANXA7 and microtubule-associated protein 1 light chain 3 (LC3), and ANXA7 was essential for LC3 accumulation in VEC autophagy. As a GTPase, whether ANXA7 affects the phosphorylation of LC3 or other proteins needs further investigation. In this study, we performed site-directed mutagenesis and found that ABO directly bound to Thr(286) of ANXA7 and inhibited its phosphorylation. By yeast two-hybrid screening, we found that ANXA7 could interact with grancalcin (GCA). ABO promoted the interaction and inhibited GCA phosphorylation, leading to the decrease of intracellular Ca(2+) concentration. At the same time, ABO inhibited the phosphorylation of LC3. Hence, by identifying ABO as an unprecedented modulator of ANXA7 as well as GCA and LC3 as interacting proteins of ANXA7, we demonstrated the possible mechanisms how ANXA7 regulates autophagy for the first time.
Collapse
|
28
|
Hoat TX, Nakayashiki H, Yang Q, Tosa Y, Mayama S. Molecular cloning of the apoptosis-related calcium-binding protein AsALG-2 in Avena sativa. MOLECULAR PLANT PATHOLOGY 2013; 14:222-9. [PMID: 23083467 PMCID: PMC6638752 DOI: 10.1111/j.1364-3703.2012.00844.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Victorin, the host-selective toxin produced by the fungus Cochliobolus victoriae, induces programmed cell death (PCD) in victorin-sensitive oat lines with characteristic features of animal apoptosis, such as mitochondrial permeability transition, chromatin condensation, nuclear DNA laddering and rRNA/mRNA degradation. In this study, we characterized a calcium-binding protein, namely AsALG-2, which might have a role in the victorin-induced PCD. AsALG-2 is homologous to the Apoptosis-Linked Gene ALG-2 identified in mammalian cells. Northern blot analysis revealed that the accumulation of AsALG-2 transcripts increased during victorin-induced PCD, but not during necrotic cell death. Salicylic acid, chitosan and chitin strongly activated the expression of general defence response genes, such as PR-10; however, neither induced cell death nor the accumulation of AsALG-2 mRNA. Pharmacological studies indicated that victorin-induced DNA laddering and AsALG-2 expression were regulated through similar pathways. The calcium channel blocker, nifedipine, moderately inhibited the accumulation of AsALG-2 mRNA during cell death. Trifluoperazine (calmodulin antagonist) and K252a (serine-threonine kinase inhibitor) reduced the victorin-induced phytoalexin accumulation, but did not prevent the victorin-induced DNA laddering or accumulation of AsALG-2 mRNA. Taken together, our investigations suggest that there is a calcium-mediated signalling pathway in animal and plant PCD in common.
Collapse
Affiliation(s)
- Trinh Xuan Hoat
- Laboratory of Plant Pathology, Graduate School of Science and Technology, Kobe University, Rokkodai, Nada-ku, Kobe, Japan.
| | | | | | | | | |
Collapse
|
29
|
Yoshibori M, Yorimitsu T, Sato K. Involvement of the penta-EF-hand protein Pef1p in the Ca2+-dependent regulation of COPII subunit assembly in Saccharomyces cerevisiae. PLoS One 2012; 7:e40765. [PMID: 22792405 PMCID: PMC3394733 DOI: 10.1371/journal.pone.0040765] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Accepted: 06/13/2012] [Indexed: 11/24/2022] Open
Abstract
Although it is well established that the coat protein complex II (COPII) mediates the transport of proteins and lipids from the endoplasmic reticulum (ER) to the Golgi apparatus, the regulation of the vesicular transport event and the mechanisms that act to counterbalance the vesicle flow between the ER and Golgi are poorly understood. In this study, we present data indicating that the penta-EF-hand Ca2+-binding protein Pef1p directly interacts with the COPII coat subunit Sec31p and regulates COPII assembly in Saccharomyces cerevisiae. ALG-2, a mammalian homolog of Pef1p, has been shown to interact with Sec31A in a Ca2+-dependent manner and to have a role in stabilizing the association of the Sec13/31 complex with the membrane. However, Pef1p displayed reversed Ca2+ dependence for Sec13/31p association; only the Ca2+-free form of Pef1p bound to the Sec13/31p complex. In addition, the influence on COPII coat assembly also appeared to be reversed; Pef1p binding acted as a kinetic inhibitor to delay Sec13/31p recruitment. Our results provide further evidence for a linkage between Ca2+-dependent signaling and ER-to-Golgi trafficking, but its mechanism of action in yeast seems to be different from the mechanism reported for its mammalian homolog ALG-2.
Collapse
Affiliation(s)
- Mariko Yoshibori
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba, Meguro-ku, Tokyo, Japan
| | - Tomohiro Yorimitsu
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba, Meguro-ku, Tokyo, Japan
| | - Ken Sato
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba, Meguro-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
30
|
Takahashi T, Suzuki H, Inuzuka T, Shibata H, Maki M. Prediction of a new ligand-binding site for type 2 motif based on the crystal structure of ALG-2 by dry and wet approaches. Int J Mol Sci 2012; 13:7532-7549. [PMID: 22837710 PMCID: PMC3397542 DOI: 10.3390/ijms13067532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/06/2012] [Accepted: 06/13/2012] [Indexed: 12/12/2022] Open
Abstract
ALG-2 is a penta-EF-hand Ca2+-binding protein and interacts with a variety of intracellular proteins. Two types of ALG-2-binding motifs have been determined: type 1, PXYPXnYP (X, variable; n = 4), in ALIX and PLSCR3; type 2, PXPGF, in Sec31A and PLSCR3. The previously solved X-ray crystal structure of the complex between ALG-2 and an ALIX peptide containing type 1 motif showed that the peptide binds to Pocket 1 and Pocket 2. Co-crystallization of ALG-2 and type 2 motif-containing peptides has not been successful. To gain insights into the molecular basis of type 2 motif recognition, we searched for a new hydrophobic cavity by computational algorithms using MetaPocket 2.0 based on 3D structures of ALG-2. The predicted hydrophobic pocket designated Pocket 3 fits with N-acetyl-ProAlaProGlyPhe-amide, a virtual penta-peptide derived from one of the two types of ALG-2-binding sites in PLSCR3 (type 2 motif), using the molecular docking software AutoDock Vina. We investigated effects of amino acid substitutions of the predicted binding sites on binding abilities by pulldown assays using glutathione-S-transferase -fused ALG-2 of wild-type and mutant proteins and lysates of cells expressing green fluorescent protein -fused PLSCR3 of wild-type and mutants. Substitution of either L52 with Ala or F148 with Ser of ALG-2 caused loss of binding abilities to PLSCR3 lacking type 1 motif but retained those to PLSCR3 lacking type 2 motif, strongly supporting the hypothesis that Pocket 3 is the binding site for type 2 motif.
Collapse
Affiliation(s)
| | | | | | | | - Masatoshi Maki
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-52-789-4088; Fax: +81-52-789-5542
| |
Collapse
|
31
|
Osugi K, Suzuki H, Nomura T, Ariumi Y, Shibata H, Maki M. Identification of the P-body component PATL1 as a novel ALG-2-interacting protein by in silico and far-Western screening of proline-rich proteins. J Biochem 2012; 151:657-66. [PMID: 22437941 DOI: 10.1093/jb/mvs029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
ALG-2 (also named PDCD6) is a 22-kDa Ca(2+)-binding protein that belongs to the penta-EF-hand family including calpain small subunit and interacts with various proteins such as ALIX and Sec31A at their specific sites containing an ALG-2-binding motif (ABM) present in their respective Pro-rich region (PRR). In this study, to search for novel ALG-2-interacting proteins, we first performed in silico screening of ABM-containing PRRs in a human protein database. After selecting 17 sequences, we expressed the PRR or full-length proteins fused with green fluorescent protein (GFP) in HEK293T cells and analysed their abilities to bind to ALG-2 by Far-Western blotting using biotinylated ALG-2 as a probe. As a result, we found 10 positive new ALG-2-binding candidates with different degrees of binding ability. For further investigation, we selected PATL1 (alternatively designated Pat1b), a component of the P-body, which is a cytoplasmic non-membranous granule composed of translation-inactive mRNAs and proteins involved in mRNA decay. Interactions between endogenous PATL1 and ALG-2 proteins were demonstrated by a co-immunoprecipitation assay using their specific antibodies. Furthermore, in immunofluorescence microscopic analyses, PATL1 as well as DCP1A, a well-known P-body marker, co-localized with a subset of ALG-2. This is the first report showing interaction of ALG-2 with a P-body component.
Collapse
Affiliation(s)
- Kanae Osugi
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa-ku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Su D, Xu H, Feng J, Gao Y, Gu L, Ying L, Katsaros D, Yu H, Xu S, Qi M. PDCD6 is an independent predictor of progression free survival in epithelial ovarian cancer. J Transl Med 2012; 10:31. [PMID: 22369209 PMCID: PMC3305474 DOI: 10.1186/1479-5876-10-31] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 02/27/2012] [Indexed: 11/24/2022] Open
Abstract
Background Programmed cell death 6 (PDCD6) beside its known proapoptotic functions may be a player in survival pathways in cancer. The purpose of this study is to further explore the roles of PDCD6 in epithelial ovarian cancer. Methods Lentiviral vector with shRNA for PDCD6 was used to investigate the effects of PDCD6 knockdown on cell growth, cell cycle, apoptosis and motility in ovarian cancer cells. Two hundred twelve epithelial ovarian cancer tissues were analyzed for mRNA expression of PDCD6 using RT-PCR. Associations of its expression with clinical pathological factors, progression free and overall survival were evaluated. Results PDCD6 is highly expressed in metastatic ovarian cancer cells and positively regulates cell migration and invasion. Significantly, the level of PDCD6 expression in epithelial ovarian cancer correlates with clinical progression. Patients with medium or high levels of PDCD6 mRNA were at higher risk for disease progression, compared to those with low levels (HR, 1.29; P = 0.024 for medium levels; and HR, 1.57; P = 0.045 for high levels) after adjusting for age, disease stage, tumor grade, histologic type and residual tumor size. Kaplan-Meier survival analysis demonstrated similar results. However, no association was found between PDCD6 expression and overall survival. Conclusions PDCD6 seems to play an important role in ovarian cancer progression and it may be an independent predictor of progression free survival in epithelial ovarian cancer. Further studies are needed to more completely elucidate the molecular mechanisms of PDCD6 involve in ovarian cancer progression.
Collapse
Affiliation(s)
- Dan Su
- Center for Genetic and Genomic Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Maki M, Suzuki H, Shibata H. Structure and function of ALG-2, a penta-EF-hand calcium-dependent adaptor protein. SCIENCE CHINA-LIFE SCIENCES 2011; 54:770-9. [PMID: 21786200 DOI: 10.1007/s11427-011-4204-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 06/05/2011] [Indexed: 12/29/2022]
Abstract
ALG-2 (a gene product of PDCD6) is a 22-kD protein containing five serially repetitive EF-hand structures and belongs to the penta-EF-hand (PEF) family, including the subunits of typical calpains. ALG-2 is the most conserved protein among the PEF family members and its homologs are widely found in eukaryotes. X-ray crystal structures of various PEF proteins including ALG-2 have common features: presence of eight α-helices and dimer formation via paired EF5s that are positioned in anti-parallel orientation. ALG-2 forms a homodimer and a heterodimer with its closest paralog peflin. Like calmodulin, a well-known four-EF-hand protein, ALG-2 interacts with various proteins in a Ca(2+)-dependent fashion, but the binding motifs are completely different. With some exceptions, ALG-2-interacting proteins commonly contain Pro-rich regions, and ALG-2 recognizes at least two distinct Pro-containing motifs: PPYP(X)nYP (X, variable; n=4 in ALIX and PLSCR3) and PXPGF (represented by Sec31A). A shorter alternatively spliced isoform, lacking two residues and designated ALG-2(ΔGF122), does not bind ALIX but maintains binding capacity to Sec31A. X-ray crystal structural analyses have revealed that binding of calcium ions induces the configuration of the side chain of R125 so that it opens Pocket 1, which accepts PPYP, but Pocket 1 remains closed in the case of ALG-2(ΔGF122). ALG-2 dimer has two ligand-binding sites, each in a monomer molecule, and appears to function as a Ca(2+)-dependent adaptor protein to either stabilize a preformed complex or to bridge two proteins on scaffolds in systems of the endosomal sorting complex required for transport (ESCRT) and ER-to-Golgi transport.
Collapse
Affiliation(s)
- Masatoshi Maki
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| | | | | |
Collapse
|
34
|
Inuzuka T, Suzuki H, Kawasaki M, Shibata H, Wakatsuki S, Maki M. Molecular basis for defect in Alix-binding by alternatively spliced isoform of ALG-2 (ALG-2DeltaGF122) and structural roles of F122 in target recognition. BMC STRUCTURAL BIOLOGY 2010; 10:25. [PMID: 20691033 PMCID: PMC2927601 DOI: 10.1186/1472-6807-10-25] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 08/06/2010] [Indexed: 11/10/2022]
Abstract
Background ALG-2 (a gene product of PDCD6) belongs to the penta-EF-hand (PEF) protein family and Ca2+-dependently interacts with various intracellular proteins including mammalian Alix, an adaptor protein in the ESCRT system. Our previous X-ray crystal structural analyses revealed that binding of Ca2+ to EF3 enables the side chain of R125 to move enough to make a primary hydrophobic pocket (Pocket 1) accessible to a short fragment of Alix. The side chain of F122, facing a secondary hydrophobic pocket (Pocket 2), interacts with the Alix peptide. An alternatively spliced shorter isoform, designated ALG-2ΔGF122, lacks Gly121Phe122 and does not bind Alix, but the structural basis of the incompetence has remained to be elucidated. Results We solved the X-ray crystal structure of the PEF domain of ALG-2ΔGF122 in the Ca2+-bound form and compared it with that of ALG-2. Deletion of the two residues shortened α-helix 5 (α5) and changed the configuration of the R125 side chain so that it partially blocked Pocket 1. A wall created by the main chain of 121-GFG-123 and facing the two pockets was destroyed. Surprisingly, however, substitution of F122 with Ala or Gly, but not with Trp, increased the Alix-binding capacity in binding assays. The F122 substitutions exhibited different effects on binding of ALG-2 to other known interacting proteins, including TSG101 (Tumor susceptibility gene 101) and annexin A11. The X-ray crystal structure of the F122A mutant revealed that removal of the bulky F122 side chain not only created an additional open space in Pocket 2 but also abolished inter-helix interactions with W95 and V98 (present in α4) and that α5 inclined away from α4 to expand Pocket 2, suggesting acquirement of more appropriate positioning of the interacting residues to accept Alix. Conclusions We found that the inability of the two-residue shorter ALG-2 isoform to bind Alix is not due to the absence of bulky side chain of F122 but due to deformation of a main-chain wall facing pockets 1 and 2. Moreover, a residue at the position of F122 contributes to target specificity and a smaller side chain is preferable for Alix binding but not favored to bind annexin A11.
Collapse
Affiliation(s)
- Tatsutoshi Inuzuka
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Vergarajauregui S, Martina JA, Puertollano R. Identification of the penta-EF-hand protein ALG-2 as a Ca2+-dependent interactor of mucolipin-1. J Biol Chem 2009; 284:36357-36366. [PMID: 19864416 DOI: 10.1074/jbc.m109.047241] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Loss of function mutations in mucolipin-1 (MCOLN1) have been linked to mucolipidosis type IV (MLIV), a recessive lysosomal storage disease characterized by severe neurological and ophthalmological abnormalities. MCOLN1 is an ion channel that regulates membrane transport along the endolysosomal pathway. It has been suggested that MCOLN1 participates in several Ca(2+)-dependent processes, including fusion of lysosomes with the plasma membrane, fusion of late endosomes and autophagosomes with lysosomes, and lysosomal biogenesis. Here, we searched for proteins that interact with MCOLN1 in a Ca(2+)-dependent manner. We found that the penta-EF-hand protein ALG-2 binds to the NH-terminal cytosolic tail of MCOLN1. The interaction is direct, strictly dependent on Ca(2+), and mediated by a patch of charged and hydrophobic residues located between MCOLN1 residues 37 and 49. We further show that MCOLN1 and ALG-2 co-localize to enlarged endosomes induced by overexpression of an ATPase-defective dominant-negative form of Vps4B (Vps4B(E235Q)). In agreement with the proposed role of MCOLN1 in the regulation of fusion/fission events, we found that overexpression of MCOLN1 caused accumulation of enlarged, aberrant endosomes that contain both early and late endosome markers. Interestingly, aggregation of abnormal endosomes was greatly reduced when the ALG-2-binding domain in MCOLN1 was mutated, suggesting that ALG-2 regulates MCOLN1 function. Overall, our data provide new insight into the molecular mechanisms that regulate MCOLN1 activity. We propose that ALG-2 acts as a Ca(2+) sensor that modulates the function of MCOLN1 along the late endosomal-lysosomal pathway.
Collapse
Affiliation(s)
- Silvia Vergarajauregui
- Laboratory of Cell Biology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Jose A Martina
- Laboratory of Cell Biology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Rosa Puertollano
- Laboratory of Cell Biology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
36
|
The mechanism of Ca2+-dependent recognition of Alix by ALG-2: insights from X-ray crystal structures. Biochem Soc Trans 2009; 37:190-4. [PMID: 19143629 DOI: 10.1042/bst0370190] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Alix [ALG-2 (apoptosis-linked gene 2)-interacting protein X] was originally identified as a protein that interacts with ALG-2, a member of the penta-EF-hand Ca(2+)-binding protein family. ALG-2 binds to its C-terminal proline-rich region that contains four tandem repeats of PXY (where X represents an uncharged amino acid). Recent X-ray crystal structural analyses of the Ca(2+)-free and Ca(2+)-bound forms of ALG-2, as well as the complex with an Alix oligopeptide, have revealed a mechanism of Ca(2+)-dependent binding of ALG-2 to its target protein. Binding of Ca(2+) to EF3 (third EF-hand) enables the side chain of Arg(125), present in the loop connecting EF3 and EF4 (fourth EF-hand), to move sufficiently to make a primary hydrophobic pocket accessible to the critical PPYP (Pro-Pro-Tyr-Pro) motif in Alix, which partially overlaps with the GPP (Gly-Pro-Pro) motif for binding to Cep55 (centrosome protein of 55 kDa). The fact that ALG-2 forms a homodimer and each monomer has one peptide-binding site indicates the possibility that ALG-2 bridges two interacting proteins, including Alix and Tsg101 (tumour susceptibility gene 101), and functions as a Ca(2+)-dependent adaptor protein.
Collapse
|
37
|
Structural basis for Ca2+ -dependent formation of ALG-2/Alix peptide complex: Ca2+/EF3-driven arginine switch mechanism. Structure 2008; 16:1562-73. [PMID: 18940611 DOI: 10.1016/j.str.2008.07.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 07/11/2008] [Accepted: 07/24/2008] [Indexed: 11/21/2022]
Abstract
ALG-2 belongs to the penta-EF-hand (PEF) protein family and interacts with various intracellular proteins, such as Alix and TSG101, that are involved in endosomal sorting and HIV budding. Through X-ray crystallography, we solved the structures of Ca(2+)-free and -bound forms of N-terminally truncated human ALG-2 (des3-20ALG-2), Zn(2+)-bound form of full-length ALG-2, and the structure of the complex between des3-23ALG-2 and the peptide corresponding to Alix799-814 in Zn(2+)-bound form. Binding of Ca(2+) to EF3 enables the side chain of Arg125, present in the loop connecting EF3 and EF4, to move enough to make a primary hydrophobic pocket accessible to the critical PPYP motif, which partially overlaps with the GPP motif for the binding of Cep55 (centrosome protein 55 kDa). Based on these results, together with the results of in vitro binding assay with mutant ALG-2 and Alix proteins, we propose a Ca(2+)/EF3-driven arginine switch mechanism for ALG-2 binding to Alix.
Collapse
|
38
|
Suzuki H, Kawasaki M, Kakiuchi T, Shibata H, Wakatsuki S, Maki M. Crystallization and X-ray diffraction analysis of N-terminally truncated human ALG-2. Acta Crystallogr Sect F Struct Biol Cryst Commun 2008; 64:974-7. [PMID: 18997320 DOI: 10.1107/s1744309108030297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 09/20/2008] [Indexed: 01/20/2023]
Abstract
ALG-2 (apoptosis-linked gene 2) is an apoptosis-linked calcium-binding protein with five EF-hand motifs in the C-terminal region. N-terminally truncated ALG-2 (des3-23ALG-2) was crystallized by the vapour-diffusion method in buffer consisting of either 50 mM MES pH 6.5, 12.5%(v/v) 2-propanol and 150 mM calcium acetate or 100 mM MES pH 6.0, 15%(v/v) ethanol and 200 mM zinc acetate. Crystals of the Ca(2+)-bound form belonged to space group P2(1)2(1)2(1), with unit-cell parameters a = 54.8, b = 154.4, c = 237.7 A, alpha = beta = gamma = 90 degrees , and diffracted to 3.1 A resolution. Crystals of the Zn(2+)-bound form belonged to space group P2(1)2(1)2(1), with unit-cell parameters a = 52.8, b = 147.5, c = 230.7 A, alpha = beta = gamma = 90 degrees , and diffracted to 3.3 A resolution. The structures of the Ca(2+)-bound form and the Zn(2+)-bound form were solved by the molecular-replacement method. Although both crystals contained eight ALG-2 molecules per asymmetric unit, the metal-ion locations and octameric arrangements were found to be significantly different.
Collapse
Affiliation(s)
- Hironori Suzuki
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Shibata H, Suzuki H, Kakiuchi T, Inuzuka T, Yoshida H, Mizuno T, Maki M. Identification of Alix-type and Non-Alix-type ALG-2-binding sites in human phospholipid scramblase 3: differential binding to an alternatively spliced isoform and amino acid-substituted mutants. J Biol Chem 2008; 283:9623-32. [PMID: 18256029 DOI: 10.1074/jbc.m800717200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ALG-2, a prototypic member of the penta-EF-hand protein family, interacts with Alix at its C-terminal Pro-rich region containing four tandem PXY repeats. Human phospholipid scramblase 3 (PLSCR3) has a similar sequence (ABS-1) in its N-terminal region. In the present study, we found that ALG-2 interacts with PLSCR3 expressed in HEK293 cells in a Ca(2+)-dependent manner by co-immunoprecipitation, pulldown with glutathione S-transferase (GST) fused ALG-2 and an overlay assay using biotin-labeled ALG-2. The GST fusion protein of an alternatively spliced isoform of ALG-2, GST-ALG-2(DeltaGF122), pulled down green fluorescent protein (GFP)-fused PLSCR3 but not GFP Alix. Deletion of a region containing ABS-1 was not sufficient to abrogate the binding. A second ALG-2-binding site (ABS-2) was essential for interaction with ALG-2(DeltaGF122). Real-time interaction analyses with a surface plasmon resonance biosensor using synthetic oligopeptides and recombinant proteins corroborated direct Ca(2+)-dependent binding of ABS-1 to ALG-2 and that of ABS-2 to ALG-2 as well as to ALG-2(DeltaGF122). The sequence of ABS-2 contains multiple prolines and two phenylalanines, among which Phe(49) was found to be critical, because its substitution with Ala or Tyr caused a loss of binding ability by pulldown assays using oligopeptide-immobilized beads. ALG-2-interacting proteins were classified into two groups based on binding ability to ALG-2(DeltaGF122): (i) isoform-non-interactive (ABS-1) types, including Alix, annexin A7, annexin A11, and TSG101 and (ii) isoform-interactive (ABS-2) types including PLSCR3, PLSCR4 and Sec31A. GST-pulldown assays using single amino acid-substituted ALG-2 mutants revealed differences in binding specificities between the two groups, suggesting structural flexibility in ALG-2-ligand complex formation.
Collapse
Affiliation(s)
- Hideki Shibata
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Dræby I, Woods YL, la Cour JM, Mollerup J, Bourdon JC, Berchtold MW. The calcium binding protein ALG-2 binds and stabilizes Scotin, a p53-inducible gene product localized at the endoplasmic reticulum membrane. Arch Biochem Biophys 2007; 467:87-94. [PMID: 17889823 PMCID: PMC2691584 DOI: 10.1016/j.abb.2007.07.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Accepted: 07/23/2007] [Indexed: 11/25/2022]
Abstract
ALG-2 (apoptosis linked gene 2 product) is a calcium binding protein for which no clear cellular function has been established. In this study we identified Scotin as a novel ALG-2 target protein containing 6 PXY and 4 PYP repeats, earlier identified in the ALG-2 binding regions of AIP1/ALIX and TSG101, respectively. An in vitro synthesized C-terminal fragment of Scotin bound specifically to immobilized recombinant ALG-2 and tagged ALG-2 and Scotin were shown by immunoprecipitation to interact in MCF7 and U2OS cell lines. Furthermore ALG-2 bound to endogenous Scotin in extracts from mouse NIH3T3 cells. Overexpression of ALG-2 led to accumulation of Scotin in MCF7 and H1299 cells. In vitro and in vivo binding of ALG-2 to Scotin was demonstrated to be strictly calcium dependent indicating a role of this interaction in calcium signaling pathways.
Collapse
Affiliation(s)
- Ingrid Dræby
- Department of Molecular Biology, University of Copenhagen, Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Yvonne L. Woods
- Department of Surgery and Molecular Oncology, European Associated Laboratory Inserm U858, University of Dundee, Ninewells Hospital, Dundee DD1 9SY, UK
| | - Jonas M. la Cour
- Department of Molecular Biology, University of Copenhagen, Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Jens Mollerup
- Department of Molecular Biology, University of Copenhagen, Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Jean-Christophe Bourdon
- Department of Surgery and Molecular Oncology, European Associated Laboratory Inserm U858, University of Dundee, Ninewells Hospital, Dundee DD1 9SY, UK
| | - Martin W. Berchtold
- Department of Molecular Biology, University of Copenhagen, Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
- Department of Surgery and Molecular Oncology, European Associated Laboratory Inserm U858, University of Dundee, Ninewells Hospital, Dundee DD1 9SY, UK
| |
Collapse
|
41
|
Mattei S, Klein G, Satre M, Aubry L. Trafficking and developmental signaling: Alix at the crossroads. Eur J Cell Biol 2007; 85:925-36. [PMID: 16766083 DOI: 10.1016/j.ejcb.2006.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Alix is a phylogenetically conserved protein that participates in mammals in programmed cell death in association with ALG-2, a penta-EF-hand calciprotein. It contains an N-terminal Bro1 domain, a coiled-coil region and a C-terminal proline-rich domain containing several SH3- and WW-binding sites that contribute to its scaffolding properties. Recent data showed that by virtue of its Bro1 domain, Alix is functionally associated to the ESCRT complexes involved in the biogenesis of the multivesicular body and sorting of transmembrane proteins within this specific endosomal compartment. In Dictyostelium, an alx null strain shows a markedly perturbed starvation-induced morphogenetic program while ALG-2 disruptants remain unaffected. This review summarizes Dictyostelium data on Alix and ALG-2 homologues and evaluates whether known functions of Alix in other organisms can account for the developmental arrest of the alx null mutant and how Dictyostelium studies can substantiate the current understanding of the function(s) of this versatile and conserved signaling molecule.
Collapse
Affiliation(s)
- Sara Mattei
- Laboratoire de Biochimie et Biophysique des Systemes Integres, DRDC/BBSI, UMR 5092 CNRS-CEA-UJF, CEA-Grenoble, 17 Rue des Martyrs, F-38054 Grenoble cedex 9, France
| | | | | | | |
Collapse
|
42
|
Structural aspects of calcium-binding proteins and their interactions with targets. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/s0167-7306(06)41004-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
43
|
Shibata H, Suzuki H, Yoshida H, Maki M. ALG-2 directly binds Sec31A and localizes at endoplasmic reticulum exit sites in a Ca2+-dependent manner. Biochem Biophys Res Commun 2006; 353:756-63. [PMID: 17196169 DOI: 10.1016/j.bbrc.2006.12.101] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Accepted: 12/14/2006] [Indexed: 10/23/2022]
Abstract
Intracellular localization of the penta-EF-hand Ca2+-binding protein ALG-2 in HeLa cells was investigated by immunofluorescent confocal microscopy using a polyclonal antibody. In addition to its presence in the nucleus, ALG-2 was found to be distributed in a punctate pattern in the cytoplasm, where it was partly co-stained with an endoplasmic reticulum (ER) exit site marker p125. In vitro GST pull down analysis demonstrated that ALG-2 and its alternatively spliced isoform interact with the COPII component Sec31A in a Ca2+-dependent manner, and a biotin-labeled ALG-2 overlay assay revealed direct binding of ALG-2 to Sec31A. Biochemical and immunofluorescent microscopic analyses showed that ALG-2 was enriched at the Sec31A-localizing membrane compartments upon stimulation with the Ca2+ ionophore A23187. In contrast, treatment of cells with the membrane-permeant Ca2+ chelator BAPTA-AM led to a dispersion of ALG-2 throughout the cells and to a significant loss of Sec31A in the perinuclear region. These findings establish Sec31A as a novel target for ALG-2 and provide a framework for studies on the roles of ALG-2 in ER-Golgi transport.
Collapse
Affiliation(s)
- Hideki Shibata
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | |
Collapse
|
44
|
Ichioka F, Takaya E, Suzuki H, Kajigaya S, Buchman VL, Shibata H, Maki M. HD-PTP and Alix share some membrane-traffic related proteins that interact with their Bro1 domains or proline-rich regions. Arch Biochem Biophys 2006; 457:142-9. [PMID: 17174262 DOI: 10.1016/j.abb.2006.11.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 11/07/2006] [Accepted: 11/09/2006] [Indexed: 11/25/2022]
Abstract
Mammalian Alix is a multifunctional adaptor protein involved in cell death, receptor endocytosis, endosomal protein sorting and cell adhesion by associating with various proteins such as ALG-2, CIN85/Rukl/SETA, endophilins, CHMP4s and TSG101. HD-PTP is a paralog of Alix and a putative protein tyrosine phosphatase (PTP) that contains a Bro1 domain, coiled-coils, a proline-rich region (PRR) in addition to a PTP domain. We investigated interactions between HD-PTP and Alix-binding proteins. In the yeast two-hybrid assay, HD-PTP showed positive interactions with CHMP4b/Shax1, TSG101, endophilin A1 and ALG-2 but not with either RabGAPLP or CIN85. We confirmed the interactions in a mammalian system by Strep-pulldown assays in which pulldown products from the lysates of HEK293T cells expressing either Strep-tagged HD-PTP alone or co-expressing with epitope-tagged proteins were analyzed by Western blotting using specific antibodies. While Alix associated with both ALG-2 and TSG101 in a Ca2+-dependent manner, HD-PTP interacted with ALG-2 Ca2+-dependently but with TSG101 Ca2+-independently.
Collapse
Affiliation(s)
- Fumitaka Ichioka
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Montaville P, Dai Y, Cheung CY, Giller K, Becker S, Michalak M, Webb SE, Miller AL, Krebs J. Nuclear translocation of the calcium-binding protein ALG-2 induced by the RNA-binding protein RBM22. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:1335-43. [PMID: 17045351 DOI: 10.1016/j.bbamcr.2006.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 08/31/2006] [Accepted: 09/01/2006] [Indexed: 11/15/2022]
Abstract
By yeast two-hybrid screening using the calcium-binding protein ALG-2 as bait a new target of ALG-2 was identified, the RNA-binding protein RBM22. In order to confirm these interactions in vivo we prepared fluorescent constructs by using the monomeric red fluorescent protein to label ALG-2 and the enhanced green fluorescent protein to label RBM22. Confocal microscopy of NIH 3T3 cells transfected with either ALG-2 or RBM22 expression constructs encoding fluorescent fusion proteins alone revealed that the majority of ALG-2 was localized in the cytoplasm whereas RBM22 was located in the nucleus. When cells were co-transfected with expression vectors encoding both fusion proteins ALG-2 was found in the nucleus indicating that RBM22 which can shuttle between the cytoplasm and the nucleus may play a role in nuclear translocation of ALG-2. Using zebrafish as a model mRNA homologues of ALG-2 and RBM22 were microinjected into the blastodisc-yolk margin of zebrafish embryos at the 1-cell stage followed by monitoring the fusion proteins during development of the zebrafish. Hereby, we observed that ALG-2 alone evenly distributed within the cell, whereas in the presence of RBM22 the two proteins co-localized within the nucleus. More than 95% of the two proteins co-localized within the same area in the nucleus suggesting a functional interaction between the Ca(2+)-signaling protein ALG-2 and the RNA-binding protein RBM22.
Collapse
Affiliation(s)
- P Montaville
- Department of NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Yamasaki A, Tani K, Yamamoto A, Kitamura N, Komada M. The Ca2+-binding protein ALG-2 is recruited to endoplasmic reticulum exit sites by Sec31A and stabilizes the localization of Sec31A. Mol Biol Cell 2006; 17:4876-87. [PMID: 16957052 PMCID: PMC1635383 DOI: 10.1091/mbc.e06-05-0444] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The formation of transport vesicles that bud from endoplasmic reticulum (ER) exit sites is dependent on the COPII coat made up of three components: the small GTPase Sar1, the Sec23/24 complex, and the Sec13/31 complex. Here, we provide evidence that apoptosis-linked gene 2 (ALG-2), a Ca(2+)-binding protein of unknown function, regulates the COPII function at ER exit sites in mammalian cells. ALG-2 bound to the Pro-rich region of Sec31A, a ubiquitously expressed mammalian orthologue of yeast Sec31, in a Ca(2+)-dependent manner and colocalized with Sec31A at ER exit sites. A Ca(2+) binding-deficient ALG-2 mutant, which did not bind Sec31A, lost the ability to localize to ER exit sites. Overexpression of the Pro-rich region of Sec31A or RNA interference-mediated Sec31A depletion also abolished the ALG-2 localization at these sites. In contrast, depletion of ALG-2 substantially reduced the level of Sec31A associated with the membrane at ER exit sites. Finally, treatment with a cell-permeable Ca(2+) chelator caused the mislocalization of ALG-2, which was accompanied by a reduced level of Sec31A at ER exit sites. We conclude that ALG-2 is recruited to ER exit sites via Ca(2+)-dependent interaction with Sec31A and in turn stabilizes the localization of Sec31A at these sites.
Collapse
Affiliation(s)
- Akinori Yamasaki
- *Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Katsuko Tani
- School of Life Science, Tokyo University of Pharmacy and Life Science, Hachioji 192-0392, Japan; and
| | - Akitsugu Yamamoto
- Department of Bio-science, Nagahama Institute of Bio-science and Technology, Nagahama 526-0829, Japan
| | - Naomi Kitamura
- *Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Masayuki Komada
- *Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| |
Collapse
|
47
|
Marko M, Prabhu Y, Müller R, Blau-Wasser R, Schleicher M, Noegel AA. The annexins of Dictyostelium. Eur J Cell Biol 2006; 85:1011-22. [PMID: 16762449 DOI: 10.1016/j.ejcb.2006.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Annexins are a highly conserved ubiquitous family of Ca2+- and phospholipid-binding proteins present in nearly all eukaryotic cells. Analysis of the Dictyostelium genome revealed the presence of two annexin genes, the annexin C1 gene (nxnA) giving rise to two isoforms of 47 and 51 kDa (previously synexin), and the annexin C2 gene (nxnB) coding for a 56-kDa protein with 33% sequence identity to annexin C1. Annexin C2 is expressed at very low and constant levels throughout development. Quantification by real-time PCR indicated that it is present in about 35-fold lower amounts compared to annexin C1. We have used a GFP-tagged annexin C2 to study its cellular distribution and dynamics. In cell fractionation studies, annexin C2 cofractionates with annexin C1 and is enriched in the 100,000 g pellet. Like annexin C1, GFP-AnxC2 stains the plasma membrane. In addition it is present in the perinuclear region and overlaps to some degree with the Golgi apparatus, whereas annexin C1 is present on intracellular membranes resembling endosomal membranes and in the nucleus. Annexin C2 is not observed in the nucleus. An annexin C1 mutant (SYN-) which shows a defect during multicellular development can be rescued by full-length annexin C1, whereas overexpression of GFP-AnxC2 did not rescue the developmental defect The data support the concept that annexins, although having a highly conserved structure, participate in different functions in a cell.
Collapse
Affiliation(s)
- Marija Marko
- Institute for Biochemistry I, Medical Faculty, and Center for Molecular Medicine Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, D-50931 Köln, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Chander A, Naidu DG, Chen XL. A ten-residue domain (Y11–A20) in the NH2-terminus modulates membrane association of annexin A7. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:775-84. [PMID: 16843057 DOI: 10.1016/j.bbalip.2006.05.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 05/05/2006] [Accepted: 05/31/2006] [Indexed: 11/30/2022]
Abstract
Annexin A7 (synexin, annexin VII) is postulated to promote membrane fusion during surfactant secretion in alveolar type II cells and catecholamine secretion in adrenal chromaffin cells. Recently, we demonstrated that the 1-29 residues in the NH(2)-terminus could, possibly by interaction with the COOH-terminus, influence the Ca(2+)-dependent membrane binding, aggregation, and fusion properties of annexin A7 (A7). In this study, we further investigated this 29-residue domain by evaluating several deletion and point mutations for membrane-associated functions of A7. In comparison to A7, the mutants lacking 1-29 residues (A7Delta(1-29)) or 1-21 residues (A7Delta(1-21)), but not those lacking 1-10 residues (A7Delta(1-10)) or 21-29 residues (A7Delta(21-29)), showed diminished membrane binding. Segmental deletion of 10-20 residues (A7Delta(10-20)) also decreased the protein binding to membranes. The Ca(2+)-dependent membrane aggregation of PLV with A7Delta(1-29) was maximally diminished but less so with A7Delta(10-20) or A7Delta(1-21) in comparison to that with A7. However, phospholipid vesicle (PVL) aggregation was unaffected with A7Delta(1-10) or A7Delta(21-29). The Ca(2+)-dependent membrane fusion of PLV was also diminished with A7Delta(10-20) and A7Delta(1-29), but not with A7Delta(1-10). Since the mode of annexin A7 association and function with biological membranes could be different, we also evaluated these proteins for functional changes with isolated lung lamellar bodies. In comparison to A7, the binding to lamellar bodies was diminished for A7Delta(1-29) and A7Delta(1-21) but not for A7Delta(1-10). The Ca(2+)-dependent fusion of isolated lamellar bodies with PLV was also diminished with A7Delta(1-29), but not with A7Delta(10-20) or A7Delta(1-21). Taken together, our studies suggest that the 10-residue domain (Y(11)-A(20)) in the NH(2)-terminus modifies the phospholipid binding and aggregation properties of annexin A7. For binding and fusion of biological membranes, the 10-29-residue domain may be required although the annexin A7 properties are primarily modulated through the Y(11)-A(20) domain.
Collapse
Affiliation(s)
- Avinash Chander
- Department of Pediatrics, Stony Brook University School of Medicine, Stony Brook, NY 11794-8111, USA.
| | | | | |
Collapse
|
49
|
Konishi H, Namikawa K, Kiyama H. Annexin III implicated in the microglial response to motor nerve injury. Glia 2006; 53:723-32. [PMID: 16506224 DOI: 10.1002/glia.20327] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
To identify proteins implicated in peripheral nerve regeneration, we performed two-dimensional polyacrylamide gel electrophoresis and subsequent mass spectrometry analysis using nerve-injured hypoglossal nuclei of rat. We have identified annexin III (ANX III/ANX A3) as an induced protein after rat hypoglossal nerve injury. ANX III is known as a Ca2+-dependent phospholipid-binding protein, but its physiological function is mostly unknown. By in situ hybridization and immunohistochemistry, we demonstrated that ANX III expression was induced specifically in activated (axotomy-stimulated) microglia after nerve injury. ANX III was the most prominent ANX expressed in microglia of the major ANX family members (ANX I-VI). Hybridization signals for other ANX mRNAs (II, IV, V, and VI) were mainly observed in neuronal cells, and no significant hybridization signal for ANX I mRNA was detected in hypoglossal nuclei. In cultured microglia, ATP treatment induced ANX III translocation to the ruffling membrane where F-actin was accumulated. Further in vitro studies revealed that ANX III was not secreted and had F-actin binding activity in a Ca2+-dependent manner. These results suggest that ANX III may be a Ca2+-dependent mediator between phospholipids and F-actin in microglia stimulated by peripheral nerve injury.
Collapse
Affiliation(s)
- Hiroyuki Konishi
- Department of Anatomy and Neurobiology, Osaka City University, Graduate School of Medicine, Asahimachi, Osaka 545-8585, Japan
| | | | | |
Collapse
|
50
|
Wiens M, Belikov SI, Kaluzhnaya OV, Schröder HC, Hamer B, Perovic-Ottstadt S, Borejko A, Luthringer B, Müller IM, Müller WEG. Axial (apical-basal) expression of pro-apoptotic and pro-survival genes in the lake baikal demosponge Lubomirskia baicalensis. DNA Cell Biol 2006; 25:152-64. [PMID: 16569194 DOI: 10.1089/dna.2006.25.152] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Like in all other Metazoa, also in sponges (Porifera) proliferation, differentiation, and death of cells are controlled by apoptotic processes, thus allowing the establishment of a Bauplan (body plan). The demosponge Lubomirskia baicalensis from the Lake Baikal is especially suitable to assess the role of the apoptotic molecules, since its grade of construction is highly elaborated into an encrusting base and branches composed of modules lined up along the apical-basal axis. The four cDNAs, ALG-2, BAK, MA-3, and Bcl-2, were isolated from this sponge species. The expression levels of these genes follow characteristic gradients. While the proapoptotic genes are highly expressed at the base of the branches and comparably low at the top, the pro-survival gene follows an opposite gradient. Parallel with the tuned expression of these genes, the activities of the apoptosis-executing enzymes caspase-8 (IETDase activity) and caspase-3 (DEVDase activity) are lowest at the top of the branch and highest at their base. This characteristic expression/activity pattern of the genes/enzymes, which had been determined in a few specimens, collected from an unpolluted, natural site, appears reversed in specimens collected from an anthropogenically polluted site. These findings indicate the involvement of apoptotic proteins in the axis formation (branches) in L. baicalensis.
Collapse
Affiliation(s)
- Matthias Wiens
- Institut für Physiologische Chemie, Abteilung Angewandte Molekularbiologie, Universität Mainz, Mainz, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|