1
|
Yumura S. Wound Repair of the Cell Membrane: Lessons from Dictyostelium Cells. Cells 2024; 13:341. [PMID: 38391954 PMCID: PMC10886852 DOI: 10.3390/cells13040341] [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: 12/20/2023] [Revised: 01/30/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
The cell membrane is frequently subjected to damage, either through physical or chemical means. The swift restoration of the cell membrane's integrity is crucial to prevent the leakage of intracellular materials and the uncontrolled influx of extracellular ions. Consequently, wound repair plays a vital role in cell survival, akin to the importance of DNA repair. The mechanisms involved in wound repair encompass a series of events, including ion influx, membrane patch formation, endocytosis, exocytosis, recruitment of the actin cytoskeleton, and the elimination of damaged membrane sections. Despite the absence of a universally accepted general model, diverse molecular models have been proposed for wound repair in different organisms. Traditional wound methods not only damage the cell membrane but also impact intracellular structures, including the underlying cortical actin networks, microtubules, and organelles. In contrast, the more recent improved laserporation selectively targets the cell membrane. Studies on Dictyostelium cells utilizing this method have introduced a novel perspective on the wound repair mechanism. This review commences by detailing methods for inducing wounds and subsequently reviews recent developments in the field.
Collapse
Affiliation(s)
- Shigehiko Yumura
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan
| |
Collapse
|
2
|
Morretta E, Ruggiero D, Belvedere R, Petrella A, Bruno I, Terracciano S, Monti MC. A multidisciplinary functional proteomics-aided strategy as a tool for the profiling of a novel cytotoxic thiadiazolopyrimidone. Bioorg Chem 2023; 138:106620. [PMID: 37229937 DOI: 10.1016/j.bioorg.2023.106620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023]
Abstract
In recent years, thiadiazolopyrimidine derivatives have been acknowledged for their striking poly-pharmacological framework, thus representing an interesting scaffold for the development of new therapeutic candidates. This paper examines the synthesis and the interactome characterization of a novel bioactive thiadiazolopyrimidone (compound 1), endowed with cytotoxic activity on HeLa cancer cells. In detail, starting from a small set of synthesized thiadiazolopyrimidones, a multi-disciplinary strategy has been carried out on the most bioactive one to disclose its potential biological targets by functional proteomics, using a label-free mass spectrometry based platform coupling Drug Affinity Responsive Target Stability and targeted Limited Proteolysis-Multiple Reaction Monitoring. The identification of Annexin A6 (ANXA6) as compound 1 most reliable cellular partner paved the way to deepen the protein-ligand interaction through bio-orthogonal approaches and to prove compound 1 action on migration and invasion processes governed by ANXA6 modulation. The identification of compund 1 as the first ANXA6 protein modulator represents a relevant tool to further explore the biological role of ANXA6 in cancer, as well as to develop novel anticancer candidates.
Collapse
Affiliation(s)
- Elva Morretta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| | - Dafne Ruggiero
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| | - Raffaella Belvedere
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| | - Antonello Petrella
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| | - Ines Bruno
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| | - Stefania Terracciano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| | - Maria Chiara Monti
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy.
| |
Collapse
|
3
|
Cao J, Wan S, Chen S, Yang L. ANXA6: a key molecular player in cancer progression and drug resistance. Discov Oncol 2023; 14:53. [PMID: 37129645 PMCID: PMC10154440 DOI: 10.1007/s12672-023-00662-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023] Open
Abstract
Annexin-A6 (ANXA6), a Ca2+-dependent membrane binding protein, is the largest of all conserved annexin families and highly expressed in the plasma membrane and endosomal compartments. As a multifunctional scaffold protein, ANXA6 can interact with phospholipid membranes and various signaling proteins. These properties enable ANXA6 to participate in signal transduction, cholesterol homeostasis, intracellular/extracellular membrane transport, and repair of membrane domains, etc. Many studies have demonstrated that the expression of ANXA6 is consistently altered during tumor formation and progression. ANXA6 is currently known to mediate different patterns of tumor progression in different cancer types through multiple cancer-type specific mechanisms. ANXA6 is a potentially valuable marker in the diagnosis, progression, and treatment strategy of various cancers. This review mainly summarizes recent findings on the mechanism of tumor formation, development, and drug resistance of ANXA6. The contents reviewed herein may expand researchers' understanding of ANXA6 and contribute to developing ANXA6-based diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Jinlong Cao
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Shun Wan
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Siyu Chen
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Li Yang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Prieto-Fernández L, Menéndez ST, Otero-Rosales M, Montoro-Jiménez I, Hermida-Prado F, García-Pedrero JM, Álvarez-Teijeiro S. Pathobiological functions and clinical implications of annexin dysregulation in human cancers. Front Cell Dev Biol 2022; 10:1009908. [PMID: 36247003 PMCID: PMC9554710 DOI: 10.3389/fcell.2022.1009908] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Annexins are an extensive superfamily of structurally related calcium- and phospholipid-binding proteins, largely conserved and widely distributed among species. Twelve human annexins have been identified, referred to as Annexin A1-13 (A12 remains as of yet unassigned), whose genes are spread throughout the genome on eight different chromosomes. According to their distinct tissue distribution and subcellular localization, annexins have been functionally implicated in a variety of biological processes relevant to both physiological and pathological conditions. Dysregulation of annexin expression patterns and functions has been revealed as a common feature in multiple cancers, thereby emerging as potential biomarkers and molecular targets for clinical application. Nevertheless, translation of this knowledge to the clinic requires in-depth functional and mechanistic characterization of dysregulated annexins for each individual cancer type, since each protein exhibits varying expression levels and phenotypic specificity depending on the tumor types. This review specifically and thoroughly examines the current knowledge on annexin dysfunctions in carcinogenesis. Hence, available data on expression levels, mechanism of action and pathophysiological effects of Annexin A1-13 among different cancers will be dissected, also further discussing future perspectives for potential applications as biomarkers for early diagnosis, prognosis and molecular-targeted therapies. Special attention is devoted to head and neck cancers (HNC), a complex and heterogeneous group of aggressive malignancies, often lately diagnosed, with high mortality, and scarce therapeutic options.
Collapse
Affiliation(s)
- Llara Prieto-Fernández
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sofía T. Menéndez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - María Otero-Rosales
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Irene Montoro-Jiménez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Juana M. García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Bittel DC, Chandra G, Tirunagri LMS, Deora AB, Medikayala S, Scheffer L, Defour A, Jaiswal JK. Annexin A2 Mediates Dysferlin Accumulation and Muscle Cell Membrane Repair. Cells 2020; 9:cells9091919. [PMID: 32824910 PMCID: PMC7565960 DOI: 10.3390/cells9091919] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 01/08/2023] Open
Abstract
Muscle cell plasma membrane is frequently damaged by mechanical activity, and its repair requires the membrane protein dysferlin. We previously identified that, similar to dysferlin deficit, lack of annexin A2 (AnxA2) also impairs repair of skeletal myofibers. Here, we have studied the mechanism of AnxA2-mediated muscle cell membrane repair in cultured muscle cells. We find that injury-triggered increase in cytosolic calcium causes AnxA2 to bind dysferlin and accumulate on dysferlin-containing vesicles as well as with dysferlin at the site of membrane injury. AnxA2 accumulates on the injured plasma membrane in cholesterol-rich lipid microdomains and requires Src kinase activity and the presence of cholesterol. Lack of AnxA2 and its failure to translocate to the plasma membrane, both prevent calcium-triggered dysferlin translocation to the plasma membrane and compromise repair of the injured plasma membrane. Our studies identify that Anx2 senses calcium increase and injury-triggered change in plasma membrane cholesterol to facilitate dysferlin delivery and repair of the injured plasma membrane.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Goutam Chandra
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Laxmi M. S. Tirunagri
- Department of Cellular Biophysics, The Rockefeller University, New York, NY 10065, USA;
| | - Arun B. Deora
- Department of Cell & Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA;
| | - Sushma Medikayala
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Luana Scheffer
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Aurelia Defour
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
- Department of Genomics and Precision medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
- Correspondence: ; Tel.: +1-(202)476-6456; Fax: +1-(202)476-6014
| |
Collapse
|
6
|
Korolkova OY, Widatalla SE, Williams SD, Whalen DS, Beasley HK, Ochieng J, Grewal T, Sakwe AM. Diverse Roles of Annexin A6 in Triple-Negative Breast Cancer Diagnosis, Prognosis and EGFR-Targeted Therapies. Cells 2020; 9:E1855. [PMID: 32784650 PMCID: PMC7465958 DOI: 10.3390/cells9081855] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022] Open
Abstract
The calcium (Ca2+)-dependent membrane-binding Annexin A6 (AnxA6), is a multifunctional, predominantly intracellular scaffolding protein, now known to play relevant roles in different cancer types through diverse, often cell-type-specific mechanisms. AnxA6 is differentially expressed in various stages/subtypes of several cancers, and its expression in certain tumor cells is also induced by a variety of pharmacological drugs. Together with the secretion of AnxA6 as a component of extracellular vesicles, this suggests that AnxA6 mediates distinct tumor progression patterns via extracellular and/or intracellular activities. Although it lacks enzymatic activity, some of the AnxA6-mediated functions involving membrane, nucleotide and cholesterol binding as well as the scaffolding of specific proteins or multifactorial protein complexes, suggest its potential utility in the diagnosis, prognosis and therapeutic strategies for various cancers. In breast cancer, the low AnxA6 expression levels in the more aggressive basal-like triple-negative breast cancer (TNBC) subtype correlate with its tumor suppressor activity and the poor overall survival of basal-like TNBC patients. In this review, we highlight the potential tumor suppressor function of AnxA6 in TNBC progression and metastasis, the relevance of AnxA6 in the diagnosis and prognosis of several cancers and discuss the concept of therapy-induced expression of AnxA6 as a novel mechanism for acquired resistance of TNBC to tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Olga Y. Korolkova
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (O.Y.K.); (S.E.W.); (S.D.W.); (D.S.W.); (H.K.B.); (J.O.)
| | - Sarrah E. Widatalla
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (O.Y.K.); (S.E.W.); (S.D.W.); (D.S.W.); (H.K.B.); (J.O.)
| | - Stephen D. Williams
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (O.Y.K.); (S.E.W.); (S.D.W.); (D.S.W.); (H.K.B.); (J.O.)
| | - Diva S. Whalen
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (O.Y.K.); (S.E.W.); (S.D.W.); (D.S.W.); (H.K.B.); (J.O.)
| | - Heather K. Beasley
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (O.Y.K.); (S.E.W.); (S.D.W.); (D.S.W.); (H.K.B.); (J.O.)
| | - Josiah Ochieng
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (O.Y.K.); (S.E.W.); (S.D.W.); (D.S.W.); (H.K.B.); (J.O.)
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Amos M. Sakwe
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA; (O.Y.K.); (S.E.W.); (S.D.W.); (D.S.W.); (H.K.B.); (J.O.)
| |
Collapse
|
7
|
Larpin Y, Besançon H, Iacovache MI, Babiychuk VS, Babiychuk EB, Zuber B, Draeger A, Köffel R. Bacterial pore-forming toxin pneumolysin: Cell membrane structure and microvesicle shedding capacity determines differential survival of cell types. FASEB J 2019; 34:1665-1678. [PMID: 31914676 DOI: 10.1096/fj.201901737rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/14/2019] [Accepted: 11/06/2019] [Indexed: 12/25/2022]
Abstract
Bacterial infectious diseases can lead to death or to serious illnesses. These outcomes are partly the consequence of pore-forming toxins, which are secreted by the pathogenic bacteria (eg, pneumolysin of Streptococcus pneumoniae). Pneumolysin binds to cholesterol within the plasma membrane of host cells and assembles to form trans-membrane pores, which can lead to Ca2+ influx and cell death. Membrane repair mechanisms exist that limit the extent of damage. Immune cells which are essential to fight bacterial infections critically rely on survival mechanisms after detrimental pneumolysin attacks. This study investigated the susceptibility of different immune cell types to pneumolysin. As a model system, we used the lymphoid T-cell line Jurkat, and myeloid cell lines U937 and THP-1. We show that Jurkat T cells are highly susceptible to pneumolysin attack. In contrast, myeloid THP-1 and U937 cells are less susceptible to pneumolysin. In line with these findings, human primary T cells are shown to be more susceptible to pneumolysin attack than monocytes. Differences in susceptibility to pneumolysin are due to (I) preferential binding of pneumolysin to Jurkat T cells and (II) cell type specific plasma membrane repair capacity. Myeloid cell survival is mostly dependent on Ca2+ induced expelling of damaged plasma membrane areas as microvesicles. Thus, in myeloid cells, first-line defense cells in bacterial infections, a potent cellular repair machinery ensures cell survival after pneumolysin attack. In lymphoid cells, which are important at later stages of infections, less efficient repair mechanisms and enhanced toxin binding renders the cells more sensitive to pneumolysin.
Collapse
Affiliation(s)
- Yu Larpin
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Hervé Besançon
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Mircea-Ioan Iacovache
- Laboratory of Experimental Morphology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Victoriia S Babiychuk
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Eduard B Babiychuk
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Benoît Zuber
- Laboratory of Experimental Morphology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Annette Draeger
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - René Köffel
- Department of Cell Biology, Institute of Anatomy, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Boucher E, Goldin-Blais L, Basiren Q, Mandato CA. Actin dynamics and myosin contractility during plasma membrane repair and restoration: Does one ring really heal them all? CURRENT TOPICS IN MEMBRANES 2019; 84:17-41. [PMID: 31610862 DOI: 10.1016/bs.ctm.2019.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In order to survive daily insults, cells have evolved various mechanisms that detect, stabilize and repair damages done to their plasma membrane and cytoskeletal structures. Damage to the PM endangers wounded cells by exposing them to uncontrolled exchanges with the extracellular milieu. The processes and molecular machinery enabling PM repair are therefore at the center of the bulk of the investigations into single-cell repair program. Wounds are repaired by dynamically remodeling the composition and shape of the injured area through exocytosis-mediated release of intracellular membrane components to the wounded area, endocytosis-mediated removal of the injured area, or the shedding of the injury. The wound healing program of Xenopus oocytes and early Drosophila embryos is by contrast, mostly characterized by the rapid formation of a large membrane patch over the wound that eventually fuse with the plasma membrane which restores plasma membrane continuity and lead to the shedding of patch material into the extracellular space. Formation and contraction of actomyosin ring restores normal plasma membrane composition and organizes cytoskeletal repairs. The extend of the contributions of the cytoskeleton to the wound healing program of somatic cells have comparatively received little attention. This review offers a survey of the current knowledge on how actin dynamics, myosin-based contraction and other cytoskeletal structures affects PM and cortical cytoskeleton repair of somatic cells.
Collapse
Affiliation(s)
- Eric Boucher
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Laurence Goldin-Blais
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Quentin Basiren
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Craig A Mandato
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
Grewal T, Enrich C, Rentero C, Buechler C. Annexins in Adipose Tissue: Novel Players in Obesity. Int J Mol Sci 2019; 20:ijms20143449. [PMID: 31337068 PMCID: PMC6678658 DOI: 10.3390/ijms20143449] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity and the associated comorbidities are a growing health threat worldwide. Adipose tissue dysfunction, impaired adipokine activity, and inflammation are central to metabolic diseases related to obesity. In particular, the excess storage of lipids in adipose tissues disturbs cellular homeostasis. Amongst others, organelle function and cell signaling, often related to the altered composition of specialized membrane microdomains (lipid rafts), are affected. Within this context, the conserved family of annexins are well known to associate with membranes in a calcium (Ca2+)- and phospholipid-dependent manner in order to regulate membrane-related events, such as trafficking in endo- and exocytosis and membrane microdomain organization. These multiple activities of annexins are facilitated through their diverse interactions with a plethora of lipids and proteins, often in different cellular locations and with consequences for the activity of receptors, transporters, metabolic enzymes, and signaling complexes. While increasing evidence points at the function of annexins in lipid homeostasis and cell metabolism in various cells and organs, their role in adipose tissue, obesity and related metabolic diseases is still not well understood. Annexin A1 (AnxA1) is a potent pro-resolving mediator affecting the regulation of body weight and metabolic health. Relevant for glucose metabolism and fatty acid uptake in adipose tissue, several studies suggest AnxA2 to contribute to coordinate glucose transporter type 4 (GLUT4) translocation and to associate with the fatty acid transporter CD36. On the other hand, AnxA6 has been linked to the control of adipocyte lipolysis and adiponectin release. In addition, several other annexins are expressed in fat tissues, yet their roles in adipocytes are less well examined. The current review article summarizes studies on the expression of annexins in adipocytes and in obesity. Research efforts investigating the potential role of annexins in fat tissue relevant to health and metabolic disease are discussed.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Carlos Enrich
- Department of Biomedicine, Unit of Cell Biology, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carles Rentero
- Department of Biomedicine, Unit of Cell Biology, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany.
| |
Collapse
|
10
|
Whalen DS, Widatalla SE, Korolkova OY, Nangami GS, Beasley HK, Williams SD, Virgous C, Lehmann BD, Ochieng J, Sakwe AM. Implication of calcium activated RasGRF2 in Annexin A6-mediated breast tumor cell growth and motility. Oncotarget 2019; 10:133-151. [PMID: 30719209 PMCID: PMC6349432 DOI: 10.18632/oncotarget.26512] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 12/16/2018] [Indexed: 01/10/2023] Open
Abstract
The role of AnxA6 in breast cancer and in particular, the mechanisms underlying its contribution to tumor cell growth and/or motility remain poorly understood. In this study, we established the tumor suppressor function of AnxA6 in triple negative breast cancer (TNBC) cells by showing that loss of AnxA6 is associated with early onset and rapid growth of xenograft TNBC tumors in mice. We also identified the Ca2+ activated RasGRF2 as an effector of AnxA6 mediated TNBC cell growth and motility. Activation of Ca2+ mobilizing oncogenic receptors such as epidermal growth factor receptor (EGFR) in TNBC cells or pharmacological stimulation of Ca2+ influx led to activation, subsequent degradation and altered effector functions of RasGRF2. Inhibition of Ca2+ influx or overexpression of AnxA6 blocked the activation/degradation of RasGRF2. We also show that AnxA6 acts as a scaffold for RasGRF2 and Ras proteins and that its interaction with RasGRF2 is modulated by GTP and/or activation of Ras proteins. Meanwhile, down-regulation of RasGRF2 and treatment of cells with the EGFR-targeted tyrosine kinase inhibitor (TKI) lapatinib strongly attenuated the growth of otherwise EGFR-TKI resistant AnxA6 high TNBC cells. These data not only suggest that AnxA6 modulated Ca2+ influx and effector functions of RasGRF2 underlie at least in part, the AnxA6 mediated TNBC cell growth and/or motility, but also provide a rationale to target Ras-driven TNBC with EGFR targeted therapies in combination with inhibition of RasGRF2.
Collapse
Affiliation(s)
- Diva S Whalen
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Sarrah E Widatalla
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Olga Y Korolkova
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Gladys S Nangami
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Heather K Beasley
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Stephen D Williams
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Carlos Virgous
- Animal Care Facility, Meharry Medical College, Nashville, TN, USA
| | - Brian D Lehmann
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| | - Josiah Ochieng
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Amos M Sakwe
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
11
|
Late Endosomal/Lysosomal Cholesterol Accumulation Is a Host Cell-Protective Mechanism Inhibiting Endosomal Escape of Influenza A Virus. mBio 2018; 9:mBio.01345-18. [PMID: 30042202 PMCID: PMC6058292 DOI: 10.1128/mbio.01345-18] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
To transfer the viral genome into the host cell cytoplasm, internalized influenza A virus (IAV) particles depend on the fusion of the IAV envelope with host endosomal membranes. The antiviral host interferon (IFN) response includes the upregulation of interferon-induced transmembrane protein 3 (IFITM3), which inhibits the release of the viral content into the cytosol. Although IFITM3 induction occurs concomitantly with late endosomal/lysosomal (LE/L) cholesterol accumulation, the functional significance of this process is not well understood. Here we report that LE/L cholesterol accumulation itself plays a pivotal role in the early antiviral defense. We demonstrate that inducing LE/L cholesterol accumulation is antiviral in non-IFN-primed cells, restricting incoming IAV particles and impairing mixing of IAV/endosomal membrane lipids. Our results establish a protective function of LE/L cholesterol accumulation and suggest endosomal cholesterol balance as a possible antiviral target. With annual epidemics occurring in all parts of the world and the risk of global outbreaks, influenza A virus (IAV) infections remain a major threat to public health. Infected host cells detect viral components and mount an interferon (IFN)-mediated response to restrict virus propagation and spread of infection. Identification of cellular factors and underlying mechanisms that establish such an antiviral state can provide novel strategies for the development of antiviral drugs. The contribution of LE/L cholesterol levels, especially in the context of the IFN-induced antiviral response, has remained controversial so far. Here, we report that accumulation of cholesterol in the LE/L compartment contributes to the IFN-induced host cell defense against incoming IAV. Our results establish cholesterol accumulation in LE/L per se as a novel antiviral barrier and suggest the endosomal cholesterol balance as a putative druggable host cell factor in IAV infection.
Collapse
|
12
|
Cairns R, Alvarez-Guaita A, Martínez-Saludes I, Wason SJ, Hanh J, Nagarajan SR, Hosseini-Beheshti E, Monastyrskaya K, Hoy AJ, Buechler C, Enrich C, Rentero C, Grewal T. Role of hepatic Annexin A6 in fatty acid-induced lipid droplet formation. Exp Cell Res 2017; 358:397-410. [PMID: 28712927 DOI: 10.1016/j.yexcr.2017.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/07/2017] [Accepted: 07/12/2017] [Indexed: 01/17/2023]
Abstract
Annexin A6 (AnxA6) has been implicated in the regulation of endo-/exocytic pathways, cholesterol transport, and the formation of multifactorial signaling complexes in many different cell types. More recently, AnxA6 has also been linked to triglyceride storage in adipocytes. Here we investigated the potential role of AnxA6 in fatty acid (FA) - induced lipid droplet (LD) formation in hepatocytes. AnxA6 was associated with LD from rat liver and HuH7 hepatocytes. In oleic acid (OA) -loaded HuH7 cells, substantial amounts of AnxA6 bound to LD in a Ca2+-independent manner. Remarkably, stable or transient AnxA6 overexpression in HuH7 cells led to elevated LD numbers/size and neutral lipid staining under control conditions as well as after OA loading compared to controls. In contrast, overexpression of AnxA1, AnxA2 and AnxA8 did not impact on OA-induced lipid accumulation. On the other hand, incubation of AnxA6-depleted HuH7 cells or primary hepatocytes from AnxA6 KO-mice with OA led to reduced FA accumulation and LD numbers. Furthermore, morphological analysis of liver sections from A6-KO mice revealed significantly lower LD numbers compared to wildtype animals. Interestingly, pharmacological inhibition of cytoplasmic phospholipase A2α (cPLA2α)-dependent LD formation was ineffective in AnxA6-depleted HuH7 cells. We conclude that cPLA2α-dependent pathways contribute to the novel regulatory role of hepatic AnxA6 in LD formation.
Collapse
Affiliation(s)
- Rose Cairns
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Anna Alvarez-Guaita
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Inés Martínez-Saludes
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sundeep J Wason
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Jacky Hanh
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Shilpa R Nagarajan
- Discipline of Physiology, School of Medical Science & Bosch Institute; Sydney Medical School; Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - Elham Hosseini-Beheshti
- Discipline of Physiology, School of Medical Science & Bosch Institute; Sydney Medical School; Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - Katia Monastyrskaya
- Urology Research Laboratory, Department Clinical Research, University of Bern, 3010 Bern, Switzerland
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Science & Bosch Institute; Sydney Medical School; Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 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), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain.
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
13
|
Grewal T, Hoque M, Conway JRW, Reverter M, Wahba M, Beevi SS, Timpson P, Enrich C, Rentero C. Annexin A6-A multifunctional scaffold in cell motility. Cell Adh Migr 2017; 11:288-304. [PMID: 28060548 DOI: 10.1080/19336918.2016.1268318] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Annexin A6 (AnxA6) belongs to a highly conserved protein family characterized by their calcium (Ca2+)-dependent binding to phospholipids. Over the years, immunohistochemistry, subcellular fractionations, and live cell microscopy established that AnxA6 is predominantly found at the plasma membrane and endosomal compartments. In these locations, AnxA6 acts as a multifunctional scaffold protein, recruiting signaling proteins, modulating cholesterol and membrane transport and influencing actin dynamics. These activities enable AnxA6 to contribute to the formation of multifactorial protein complexes and membrane domains relevant in signal transduction, cholesterol homeostasis and endo-/exocytic membrane transport. Hence, AnxA6 has been implicated in many biological processes, including cell proliferation, survival, differentiation, inflammation, but also membrane repair and viral infection. More recently, we and others identified roles for AnxA6 in cancer cell migration and invasion. This review will discuss how the multiple scaffold functions may enable AnxA6 to modulate migratory cell behavior in health and disease.
Collapse
Affiliation(s)
- Thomas Grewal
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Monira Hoque
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - James R W Conway
- b The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine , University of New South Wales , Sydney , NSW , Australia
| | - Meritxell Reverter
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Mohamed Wahba
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Syed S Beevi
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Paul Timpson
- b The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine , University of New South Wales , Sydney , NSW , Australia
| | - Carlos Enrich
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Carles Rentero
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| |
Collapse
|
14
|
Krautbauer S, Haberl EM, Eisinger K, Pohl R, Rein-Fischboeck L, Rentero C, Alvarez-Guaita A, Enrich C, Grewal T, Buechler C, Neumeier M. Annexin A6 regulates adipocyte lipid storage and adiponectin release. Mol Cell Endocrinol 2017; 439:419-430. [PMID: 27702590 DOI: 10.1016/j.mce.2016.09.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 09/30/2016] [Accepted: 09/30/2016] [Indexed: 12/28/2022]
Abstract
Lipid storage and adipokine secretion are critical features of adipocytes. Annexin A6 (AnxA6) is a lipid-binding protein regulating secretory pathways and its role in adiponectin release was examined. The siRNA-mediated AnxA6 knock-down in 3T3-L1 preadipocytes impaired proliferation, and differentiation of AnxA6-depleted cells to mature adipocytes was associated with higher soluble adiponectin and increased triglyceride storage. The latter was partly attributed to reduced lipolysis. Accordingly, AnxA6 overexpression in 3T3-L1 adipocytes lowered cellular triglycerides and adiponectin secretion. Indeed, serum adiponectin was increased in AnxA6 deficient mice. Expression analysis identified AnxA6 protein to be more abundant in intra-abdominal compared to subcutaneous adipose tissues of mice and men. AnxA6 protein levels increased in white adipose tissues of obese mice and here, levels were highest in subcutaneous fat. AnxA6 protein in adipocytes was upregulated by oxidative stress which might trigger AnxA6 induction in adipose tissues and contribute to impaired fat storage and adiponectin release.
Collapse
Affiliation(s)
- Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Elisabeth M Haberl
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Rebekka Pohl
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Lisa Rein-Fischboeck
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Carles Rentero
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Anna Alvarez-Guaita
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Carlos Enrich
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, 2006, Australia
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany.
| | - Markus Neumeier
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| |
Collapse
|
15
|
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: 2.4] [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
|
16
|
Khalaj K, Aminollahi E, Bordbar A, Khalaj V. Fungal annexins: a mini review. SPRINGERPLUS 2015; 4:721. [PMID: 26636009 PMCID: PMC4656261 DOI: 10.1186/s40064-015-1519-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/09/2015] [Indexed: 01/09/2023]
Abstract
The large family of annexins is composed of more than a thousand members which are typically phospholipid-binding proteins. Annexins act in a number of signalling networks and membrane trafficking events which are fundamental to cell physiology. Annexins exert their functions mainly through their calcium-dependent membrane binding abilities; however, some calcium-independent interactions have been documented in the literature. Although mammalian and plant annexins have been well characterized, little is known about this family in fungi. This mini review summarizes the available data on fungal annexins.
Collapse
Affiliation(s)
- Kamand Khalaj
- Medicine Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Aminollahi
- Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Bordbar
- Molecular Systematics Laboratory, Parasitology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Vahid Khalaj
- Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
17
|
García-Melero A, Reverter M, Hoque M, Meneses-Salas E, Koese M, Conway JRW, Johnsen CH, Alvarez-Guaita A, Morales-Paytuvi F, Elmaghrabi YA, Pol A, Tebar F, Murray RZ, Timpson P, Enrich C, Grewal T, Rentero C. Annexin A6 and Late Endosomal Cholesterol Modulate Integrin Recycling and Cell Migration. J Biol Chem 2015; 291:1320-35. [PMID: 26578516 DOI: 10.1074/jbc.m115.683557] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Indexed: 01/01/2023] Open
Abstract
Annexins are a family of proteins that bind to phospholipids in a calcium-dependent manner. Earlier studies implicated annexin A6 (AnxA6) to inhibit secretion and participate in the organization of the extracellular matrix. We recently showed that elevated AnxA6 levels significantly reduced secretion of the extracellular matrix protein fibronectin (FN). Because FN is directly linked to the ability of cells to migrate, this prompted us to investigate the role of AnxA6 in cell migration. Up-regulation of AnxA6 in several cell models was associated with reduced cell migration in wound healing, individual cell tracking and three-dimensional migration/invasion assays. The reduced ability of AnxA6-expressing cells to migrate was associated with decreased cell surface expression of αVβ3 and α5β1 integrins, both FN receptors. Mechanistically, we found that elevated AnxA6 levels interfered with syntaxin-6 (Stx6)-dependent recycling of integrins to the cell surface. AnxA6 overexpression caused mislocalization and accumulation of Stx6 and integrins in recycling endosomes, whereas siRNA-mediated AnxA6 knockdown did not modify the trafficking of integrins. Given our recent findings that inhibition of cholesterol export from late endosomes (LEs) inhibits Stx6-dependent integrin recycling and that elevated AnxA6 levels cause LE cholesterol accumulation, we propose that AnxA6 and blockage of LE cholesterol transport are critical for endosomal function required for Stx6-mediated recycling of integrins in cell migration.
Collapse
Affiliation(s)
- Ana García-Melero
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Meritxell Reverter
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Monira Hoque
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Elsa Meneses-Salas
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Meryem Koese
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia
| | - James R W Conway
- Garvan Institute of Medical Research and Kinghorn Cancer Centre, Cancer Research Program, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Camilla H Johnsen
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Anna Alvarez-Guaita
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Frederic Morales-Paytuvi
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Yasmin A Elmaghrabi
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Albert Pol
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| | - Francesc Tebar
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| | - Rachael Z Murray
- Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4095, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research and Kinghorn Cancer Centre, Cancer Research Program, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Carlos Enrich
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales 2006, Australia,
| | - Carles Rentero
- From the Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain, and
| |
Collapse
|
18
|
Proteomic analysis of 3-MCPD and 3-MCPD dipalmitate-induced toxicity in rat kidney. Arch Toxicol 2015; 90:1437-48. [DOI: 10.1007/s00204-015-1576-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/29/2015] [Indexed: 02/02/2023]
|
19
|
Shah A, Taneyhill LA. Differential expression pattern of Annexin A6 in chick neural crest and placode cells during cranial gangliogenesis. Gene Expr Patterns 2015; 18:21-8. [PMID: 25976293 DOI: 10.1016/j.gep.2015.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 01/27/2023]
Abstract
The cranial trigeminal and epibranchial ganglia are components of the peripheral nervous system that possess an important somatosensory role. These ganglia arise from the intermixing and coalescence of two different migratory cell types, neural crest cells and neurogenic placodes cells, and thus typify the phenomena of cell migration and intercellular interactions for their creation. The underlying molecular mechanisms of ganglia formation, however, are still poorly understood. To address this, we have analyzed the spatio-temporal expression profile of Annexin A6 during chick gangliogenesis, as Annexin proteins play important, conserved roles in ganglia development and physiology. We observe Annexin A6 protein in cranial neural crest cells prior to, during and after their emergence from the neural tube. Fully migratory cranial neural crest cells, however, are devoid of Annexin A6. Interestingly, we note Annexin A6 protein in trigeminal and epibranchial placode cells as these cells ingress from the ectoderm to initiate ganglia formation. This expression is also maintained in the sensory placodes later on when they coalesce with neural crest cells to assemble the cranial ganglia. These results suggest that the dynamic expression of Annexin A6 in various embryonic cell types may allow Annexin A6 to serve distinct functions throughout embryonic development.
Collapse
Affiliation(s)
- Ankita Shah
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
20
|
Iglesias JM, Cairney CJ, Ferrier RK, McDonald L, Soady K, Kendrick H, Pringle MA, Morgan RO, Martin F, Smalley MJ, Blyth K, Stein T. Annexin A8 identifies a subpopulation of transiently quiescent c-kit positive luminal progenitor cells of the ductal mammary epithelium. PLoS One 2015; 10:e0119718. [PMID: 25803307 PMCID: PMC4372349 DOI: 10.1371/journal.pone.0119718] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 02/02/2015] [Indexed: 11/18/2022] Open
Abstract
We have previously shown that Annexin A8 (ANXA8) is strongly associated with the basal-like subgroup of breast cancers, including BRCA1-associated breast cancers, and poor prognosis; while in the mouse mammary gland AnxA8 mRNA is expressed in low-proliferative isolated pubertal mouse mammary ductal epithelium and after enforced involution, but not in isolated highly proliferative terminal end buds (TEB) or during pregnancy. To better understand ANXA8's association with this breast cancer subgroup we established ANXA8's cellular distribution in the mammary gland and ANXA8's effect on cell proliferation. We show that ANXA8 expression in the mouse mammary gland was strong during pre-puberty before the expansion of the rudimentary ductal network and was limited to a distinct subpopulation of ductal luminal epithelial cells but was not detected in TEB or in alveoli during pregnancy. Similarly, during late involution its expression was found in the surviving ductal epithelium, but not in the apoptotic alveoli. Double-immunofluorescence (IF) showed that ANXA8 positive (+ve) cells were ER-alpha negative (-ve) and mostly quiescent, as defined by lack of Ki67 expression during puberty and mid-pregnancy, but not terminally differentiated with ∼15% of ANXA8 +ve cells re-entering the cell cycle at the start of pregnancy (day 4.5). RT-PCR on RNA from FACS-sorted cells and double-IF showed that ANXA8+ve cells were a subpopulation of c-kit +ve luminal progenitor cells, which have recently been identified as the cells of origin of basal-like breast cancers. Over expression of ANXA8 in the mammary epithelial cell line Kim-2 led to a G0/G1 arrest and suppressed Ki67 expression, indicating cell cycle exit. Our data therefore identify ANXA8 as a potential mediator of quiescence in the normal mouse mammary ductal epithelium, while its expression in basal-like breast cancers may be linked to ANXA8's association with their specific cells of origin.
Collapse
Affiliation(s)
- Juan Manuel Iglesias
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Synpromics Limited, Edinburgh, United Kingdom
| | - Claire J. Cairney
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Roderick K. Ferrier
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Kelly Soady
- Medical Research Council Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Howard Kendrick
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Marie-Anne Pringle
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Reginald O. Morgan
- Department of Biochemistry and Molecular Biology and the Institute of Biotechnology of Asturias (IUBA), University of Oviedo, Oviedo, Spain
| | - Finian Martin
- Conway Institute and School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Matthew J. Smalley
- European Cancer Stem Cell Research Institute, Cardiff School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Karen Blyth
- CRUK Beatson Institute, Glasgow, United Kingdom
| | - Torsten Stein
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
21
|
Alvarez-Guaita A, Vilà de Muga S, Owen DM, Williamson D, Magenau A, García-Melero A, Reverter M, Hoque M, Cairns R, Cornely R, Tebar F, Grewal T, Gaus K, Ayala-Sanmartín J, Enrich C, Rentero C. Evidence for annexin A6-dependent plasma membrane remodelling of lipid domains. Br J Pharmacol 2015; 172:1677-90. [PMID: 25409976 DOI: 10.1111/bph.13022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 11/11/2014] [Accepted: 11/14/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Annexin A6 (AnxA6) is a calcium-dependent phospholipid-binding protein that can be recruited to the plasma membrane to function as a scaffolding protein to regulate signal complex formation, endo- and exocytic pathways as well as distribution of cellular cholesterol. Here, we have investigated how AnxA6 influences the membrane order. EXPERIMENTAL APPROACH We used Laurdan and di-4-ANEPPDHQ staining in (i) artificial membranes; (ii) live cells to investigate membrane packing and ordered lipid phases; and (iii) a super-resolution imaging (photoactivated localization microscopy, PALM) and Ripley's K second-order point pattern analysis approach to assess how AnxA6 regulates plasma membrane order domains and protein clustering. KEY RESULTS In artificial membranes, purified AnxA6 induced a global increase in membrane order. However, confocal microscopy using di-4-ANEPPDHQ in live cells showed that cells expressing AnxA6, which reduces plasma membrane cholesterol levels and modifies the actin cytoskeleton meshwork, displayed a decrease in membrane order (∼15 and 30% in A431 and MEF cells respectively). PALM data from Lck10 and Src15 membrane raft/non-raft markers revealed that AnxA6 expression induced clustering of both raft and non-raft markers. Altered clustering of Lck10 and Src15 in cells expressing AnxA6 was also observed after cholesterol extraction with methyl-β-cyclodextrin or actin cytoskeleton disruption with latrunculin B. CONCLUSIONS AND IMPLICATIONS AnxA6-induced plasma membrane remodelling indicated that elevated AnxA6 expression decreased membrane order through the regulation of cellular cholesterol homeostasis and the actin cytoskeleton. This study provides the first evidence from live cells that support current models of annexins as membrane organizers.
Collapse
Affiliation(s)
- Anna Alvarez-Guaita
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hashemi Gheinani A, Burkhard FC, Rehrauer H, Aquino Fournier C, Monastyrskaya K. MicroRNA MiR-199a-5p regulates smooth muscle cell proliferation and morphology by targeting WNT2 signaling pathway. J Biol Chem 2015; 290:7067-86. [PMID: 25596533 DOI: 10.1074/jbc.m114.618694] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
MicroRNA miR-199a-5p impairs tight junction formation, leading to increased urothelial permeability in bladder pain syndrome. Now, using transcriptome analysis in urothelial TEU-2 cells, we implicate it in the regulation of cell cycle, cytoskeleton remodeling, TGF, and WNT signaling pathways. MiR-199a-5p is highly expressed in the smooth muscle layer of the bladder, and we altered its levels in bladder smooth muscle cells (SMCs) to validate the pathway analysis. Inhibition of miR-199a-5p with antimiR increased SMC proliferation, reduced cell size, and up-regulated miR-199a-5p targets, including WNT2. Overexpression of WNT2 protein or treating SMCs with recombinant WNT2 closely mimicked the miR-199a-5p inhibition, whereas down-regulation of WNT2 in antimiR-expressing SMCs with shRNA restored cell phenotype and proliferation rates. Overexpression of miR-199a-5p in the bladder SMCs significantly increased cell size and up-regulated SM22, SM α-actin, and SM myosin heavy chain mRNA and protein levels. These changes as well as increased expression of ACTG2, TGFB1I1, and CDKN1A were mediated by up-regulation of the smooth muscle-specific transcriptional activator myocardin at mRNA and protein levels. Myocardin-related transcription factor A downstream targets Id3 and MYL9 were also induced. Up-regulation of myocardin was accompanied by down-regulation of WNT-dependent inhibitory Krüppel-like transcription factor 4 in miR-199a-5p-overexpressing cells. In contrast, Krüppel-like transcription factor 4 was induced in antimiR-expressing cells following the activation of WNT2 signaling, leading to repression of myocardin-dependent genes. MiR-199a-5p plays a critical role in the WNT2-mediated regulation of proliferative and differentiation processes in the smooth muscle and may behave as a key modulator of smooth muscle hypertrophy, which is relevant for organ remodeling.
Collapse
Affiliation(s)
- Ali Hashemi Gheinani
- From the Urology Research Laboratory, Department Clinical Research, University of Bern, 3010 Bern, Switzerland
| | - Fiona C Burkhard
- Department of Urology, University Hospital, 3010 Bern, Switzerland, and
| | | | | | - Katia Monastyrskaya
- From the Urology Research Laboratory, Department Clinical Research, University of Bern, 3010 Bern, Switzerland,
| |
Collapse
|
23
|
Luo D, Fajol A, Umbach AT, Noegel AA, Laufer S, Lang F, Föller M. Influence of annexin A7 on insulin sensitivity of cellular glucose uptake. Pflugers Arch 2014; 467:641-9. [PMID: 24903239 DOI: 10.1007/s00424-014-1541-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/07/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023]
Abstract
Insulin sensitivity is decreased by prostaglandin E2 (PGE2), a major product of cyclooxygenase (COX). As shown in erythrocytes, PGE2 formation is inhibited by annexin A7. The present study defined the role of annexin A7 in glucose metabolism. Gene-targeted mice lacking annexin A7 (annexin7 (-/-)) were compared to wild-type mice (annexin7 (+/+)). The serum 6-Keto-prostaglandin-F1α (6-Keto-PGF1α) concentration was measured by ELISA and hepatic COX activity determined by an enzyme assay. Expression of COX-1, COX-2, prostaglandin E synthase, GLUT-4, and insulin receptor was determined by Western blotting. Glucose and insulin serum concentrations were analyzed following an intraperitoneal glucose load and glucose serum levels after intraperitoneal injection of insulin. Experiments were done without and with pretreatment of the mice with COX-inhibitor aspirin. The serum 6-Keto-PGF1α level and hepatic COX activity were significantly higher in annexin7 (-/-) than in annexin7 (+/+) mice. Hepatic COX-1 expression was higher in annexin7 (-/-) mice. Glucose tolerance was decreased in annexin7 (-/-) mice. Intraperitoneal insulin injection decreased the serum glucose level in both genotypes, an effect significantly less pronounced in annexin7 (-/-) mice. Glucose-induced insulin secretion was higher in annexin7 (-/-) mice. GLUT-4 expression in skeletal muscle from annexin7 (-/-) mice was reduced. Aspirin pretreatment lowered the increase in insulin concentration following glucose injection in both genotypes and virtually abrogated the differences in serum insulin between the genotypes. Aspirin pretreatment improved glucose tolerance in annexin7 (-/-) mice. In conclusion, annexin A7 influences insulin sensitivity of cellular glucose uptake and thus glucose tolerance. These effects depend on COX activity.
Collapse
Affiliation(s)
- Dong Luo
- Department of Physiology, University of Tübingen, Gmelinstr. 5, 72076, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
24
|
Hoque M, Rentero C, Cairns R, Tebar F, Enrich C, Grewal T. Annexins — Scaffolds modulating PKC localization and signaling. Cell Signal 2014; 26:1213-25. [DOI: 10.1016/j.cellsig.2014.02.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 02/22/2014] [Indexed: 12/15/2022]
|
25
|
Can T, Faas L, Ashford DA, Dowle A, Thomas J, O'Toole P, Blanco G. Proteomic analysis of laser capture microscopy purified myotendinous junction regions from muscle sections. Proteome Sci 2014; 12:25. [PMID: 25071420 PMCID: PMC4113200 DOI: 10.1186/1477-5956-12-25] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 04/25/2014] [Indexed: 12/14/2022] Open
Abstract
The myotendinous junction is a specialized structure of the muscle fibre enriched in mechanosensing complexes, including costameric proteins and core elements of the z-disc. Here, laser capture microdissection was applied to purify membrane regions from the myotendinous junctions of mouse skeletal muscles, which were then processed for proteomic analysis. Sarcolemma sections from the longitudinal axis of the muscle fibre were used as control for the specificity of the junctional preparation. Gene ontology term analysis of the combined lists indicated a statistically significant enrichment in membrane-associated proteins. The myotendinous junction preparation contained previously uncharacterized proteins, a number of z-disc costameric ligands (e.g., actinins, capZ, αB cristallin, filamin C, cypher, calsarcin, desmin, FHL1, telethonin, nebulin, titin and an enigma-like protein) and other proposed players of sarcomeric stretch sensing and signalling, such as myotilin and the three myomesin homologs. A subset were confirmed by immunofluorescence analysis as enriched at the myotendinous junction, suggesting that laser capture microdissection from muscle sections is a valid approach to identify novel myotendinous junction players potentially involved in mechanotransduction pathways.
Collapse
Affiliation(s)
- Tugba Can
- Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Laura Faas
- Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - David A Ashford
- Bioscience Technology Facility, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Adam Dowle
- Bioscience Technology Facility, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Jerry Thomas
- Bioscience Technology Facility, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Peter O'Toole
- Bioscience Technology Facility, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Gonzalo Blanco
- Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| |
Collapse
|
26
|
Voelkl J, Alesutan I, Pakladok T, Viereck R, Feger M, Mia S, Schönberger T, Noegel AA, Gawaz M, Lang F. Annexin A7 deficiency potentiates cardiac NFAT activity promoting hypertrophic signaling. Biochem Biophys Res Commun 2014; 445:244-9. [PMID: 24508799 DOI: 10.1016/j.bbrc.2014.01.186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 01/14/2023]
Abstract
Annexin A7 (Anxa7) is a cytoskeletal protein interacting with Ca(2+) signaling which in turn is a crucial factor for cardiac remodeling following cardiac injury. The present study explored whether Anxa7 participates in the regulation of cardiac stress signaling. To this end, mice lacking functional Anxa7 (anxa7(-/-)) and wild-type mice (anxa7(+/+)) were investigated following pressure overload by transverse aortic constriction (TAC). In addition, HL-1 cardiomyocytes were silenced with Anxa7 siRNA and treated with isoproterenol. Transcript levels were determined by quantitative RT-PCR, transcriptional activity by luciferase reporter assay and protein abundance by Western blotting and confocal microscopy. As a result, TAC treatment increased the mRNA and protein levels of Anxa7 in wild-type mice. Moreover, TAC increased heart weight to body weight ratio and the cardiac mRNA levels of αSka, Nppb, Col1a1, Col3a1 and Rcan1, effects more pronounced in anxa7(-/-) mice than in anxa7(+/+) mice. Silencing of Anxa7 in HL-1 cardiomyocytes significantly increased nuclear localization of Nfatc1. Furthermore, Anxa7 silencing increased NFAT-dependent transcriptional activity as well as αSka, Nppb, and Rcan1 mRNA levels both, under control conditions and following β-adrenergic stimulation by isoproterenol. These observations point to an important role of annexin A7 in the regulation of cardiac NFAT activity and hypertrophic response following cardiac stress conditions.
Collapse
MESH Headings
- Adrenergic beta-Agonists/pharmacology
- Animals
- Annexin A7/genetics
- Annexin A7/metabolism
- Aorta/pathology
- Blotting, Western
- Calcium-Binding Proteins
- Cell Line
- Cell Nucleus/metabolism
- Constriction, Pathologic
- Gene Expression/drug effects
- Hypertrophy
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Isoproterenol/pharmacology
- Male
- Mice
- Mice, 129 Strain
- Mice, Knockout
- Microscopy, Confocal
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- NFATC Transcription Factors/metabolism
- RNA Interference
- Receptors, Atrial Natriuretic Factor/genetics
- Receptors, Atrial Natriuretic Factor/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
Collapse
Affiliation(s)
- Jakob Voelkl
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | - Robert Viereck
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Martina Feger
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Sobuj Mia
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Tanja Schönberger
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | - Angelika A Noegel
- Center for Biochemistry, Institute of Biochemistry I, University of Cologne, Köln, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
27
|
Tebar F, Gelabert-Baldrich M, Hoque M, Cairns R, Rentero C, Pol A, Grewal T, Enrich C. Annexins and Endosomal Signaling. Methods Enzymol 2014; 535:55-74. [DOI: 10.1016/b978-0-12-397925-4.00004-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
28
|
Koumangoye RB, Nangami GN, Thompson PD, Agboto VK, Ochieng J, Sakwe AM. Reduced annexin A6 expression promotes the degradation of activated epidermal growth factor receptor and sensitizes invasive breast cancer cells to EGFR-targeted tyrosine kinase inhibitors. Mol Cancer 2013; 12:167. [PMID: 24354805 PMCID: PMC3922904 DOI: 10.1186/1476-4598-12-167] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 12/16/2013] [Indexed: 01/16/2023] Open
Abstract
Background The expression of annexin A6 (AnxA6) in AnxA6-deficient non-invasive tumor cells has been shown to terminate epidermal growth factor receptor (EGFR) activation and downstream signaling. However, as a scaffolding protein, AnxA6 may stabilize activated cell-surface receptors to promote cellular processes such as tumor cell motility and invasiveness. In this study, we investigated the contribution of AnxA6 in the activity of EGFR in invasive breast cancer cells and examined whether the expression status of AnxA6 influences the response of these cells to EGFR-targeted tyrosine kinase inhibitors (TKIs) and/or patient survival. Results We demonstrate that in invasive BT-549 breast cancer cells AnxA6 expression is required for sustained membrane localization of activated (phosho-Y1068) EGFR and consequently, persistent activation of MAP kinase ERK1/2 and phosphoinositide 3-kinase/Akt pathways. Depletion of AnxA6 in these cells was accompanied by rapid degradation of activated EGFR, attenuated downstream signaling and as expected enhanced anchorage-independent growth. Besides inhibition of cell motility and invasiveness, AnxA6-depleted cells were also more sensitive to the EGFR-targeted TKIs lapatinib and PD153035. We also provide evidence suggesting that reduced AnxA6 expression is associated with a better relapse-free survival but poorer distant metastasis-free and overall survival of basal-like breast cancer patients. Conclusions Together this demonstrates that the rapid degradation of activated EGFR in AnxA6-depleted invasive tumor cells underlies their sensitivity to EGFR-targeted TKIs and reduced motility. These data also suggest that AnxA6 expression status may be useful for the prediction of the survival and likelihood of basal-like breast cancer patients to respond to EGFR-targeted therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Amos M Sakwe
- Department of Biochemistry and Cancer Biology, Meharry Medical College, 1005 Dr, DB Todd Jr, Blvd, Nashville, TN 37208, USA.
| |
Collapse
|
29
|
Abstract
Influenza is caused by influenza A virus (IAV), an enveloped, negative-stranded RNA virus that derives its envelope lipids from the host cell plasma membrane. Here, we examined the functional role of cellular cholesterol in the IAV infection cycle. We show that shifting of cellular cholesterol pools via the Ca2+-regulated membrane-binding protein annexin A6 (AnxA6) affects the infectivity of progeny virus particles. Elevated levels of cellular AnxA6, which decrease plasma membrane and increase late endosomal cholesterol levels, impaired IAV replication and propagation, whereas RNA interference-mediated AnxA6 ablation increased viral progeny titers. Pharmacological accumulation of late endosomal cholesterol also diminished IAV virus propagation. Decreased IAV replication caused by upregulated AnxA6 expression could be restored either by exogenous replenishment of host cell cholesterol or by ectopic expression of the late endosomal cholesterol transporter Niemann-Pick C1 (NPC1). Virus released from AnxA6-overexpressing cells displayed significantly reduced cholesterol levels. Our results show that IAV replication depends on maintenance of the cellular cholesterol balance and identify AnxA6 as a critical factor in linking IAV to cellular cholesterol homeostasis. Influenza A virus (IAV) is a major public health concern, and yet, major host-pathogen interactions regulating IAV replication still remain poorly understood. It is known that host cell cholesterol is a critical factor in the influenza virus life cycle. The viral envelope is derived from the host cell membrane during the process of budding and, hence, equips the virus with a special lipid-protein mixture which is high in cholesterol. However, the influence of host cell cholesterol homeostasis on IAV infection is largely unknown. We show that IAV infection success critically depends on host cell cholesterol distribution. Cholesterol sequestration in the endosomal compartment impairs progeny titer and infectivity and is associated with reduced cholesterol content in the viral envelope.
Collapse
|
30
|
Pozo-Guisado E, Martin-Romero FJ. The regulation of STIM1 by phosphorylation. Commun Integr Biol 2013; 6:e26283. [PMID: 24505502 PMCID: PMC3914909 DOI: 10.4161/cib.26283] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 08/27/2013] [Indexed: 12/31/2022] Open
Abstract
Calcium ion (Ca(2+)) concentration plays a key role in cell signaling in eukaryotic cells. At the cellular level, Ca(2+) directly participates in such diverse cellular events as adhesion and migration, differentiation, contraction, secretion, synaptic transmission, fertilization, and cell death. As a consequence of these diverse actions, the cytosolic concentration of free Ca(2+) is tightly regulated by the coordinated activity of Ca(2+) channels, Ca(2+) pumps, and Ca(2+)-binding proteins. Although many of these regulators have been studied in depth, other proteins have been described recently, and naturally far less is known about their contribution to cell physiology. Within this last group of proteins, STIM1 has emerged as a major contributor to Ca(2+) signaling by means of its activity as Ca(2+) channel regulator. STIM1 is a protein resident mainly, but not exclusively, in the endoplasmic reticulum (ER), and activates a set of plasma membrane Ca(2+) channels termed store-operated calcium channels (SOCs) when the concentration of free Ca(2+) within the ER drops transiently as a result of Ca(2+) release from this compartment. Knowledge regarding the molecular architecture of STIM1 has grown considerably during the last years, and several structural domains within STIM1 have been reported to be required for the specific molecular interactions with other important players in Ca(2+) signaling, such as Ca(2+) channels and microtubules. Within the modulators of STIM1, phosphorylation has been shown to both activate and inactivate STIM1-dependent Ca(2+) entry depending on the cell type, cell cycle phase, and the specific residue that becomes modified. Here we shall review current knowledge regarding the modulation of STIM1 by phosphorylation.
Collapse
Affiliation(s)
- Eulalia Pozo-Guisado
- Department of Biochemistry and Molecular Biology; School of Life Sciences; University of Extremadura; Badajoz, Spain
| | | |
Collapse
|
31
|
Harauz G, Boggs JM. Myelin management by the 18.5-kDa and 21.5-kDa classic myelin basic protein isoforms. J Neurochem 2013; 125:334-61. [PMID: 23398367 DOI: 10.1111/jnc.12195] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 12/15/2022]
Abstract
The classic myelin basic protein (MBP) splice isoforms range in nominal molecular mass from 14 to 21.5 kDa, and arise from the gene in the oligodendrocyte lineage (Golli) in maturing oligodendrocytes. The 18.5-kDa isoform that predominates in adult myelin adheres the cytosolic surfaces of oligodendrocyte membranes together, and forms a two-dimensional molecular sieve restricting protein diffusion into compact myelin. However, this protein has additional roles including cytoskeletal assembly and membrane extension, binding to SH3-domains, participation in Fyn-mediated signaling pathways, sequestration of phosphoinositides, and maintenance of calcium homeostasis. Of the diverse post-translational modifications of this isoform, phosphorylation is the most dynamic, and modulates 18.5-kDa MBP's protein-membrane and protein-protein interactions, indicative of a rich repertoire of functions. In developing and mature myelin, phosphorylation can result in microdomain or even nuclear targeting of the protein, supporting the conclusion that 18.5-kDa MBP has significant roles beyond membrane adhesion. The full-length, early-developmental 21.5-kDa splice isoform is predominantly karyophilic due to a non-traditional P-Y nuclear localization signal, with effects such as promotion of oligodendrocyte proliferation. We discuss in vitro and recent in vivo evidence for multifunctionality of these classic basic proteins of myelin, and argue for a systematic evaluation of the temporal and spatial distributions of these protein isoforms, and their modified variants, during oligodendrocyte differentiation.
Collapse
Affiliation(s)
- George Harauz
- Department of Molecular and Cellular Biology, Biophysics Interdepartmental Group and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario, Canada.
| | | |
Collapse
|
32
|
Abed M, Balasaheb S, Towhid ST, Daniel C, Amann K, Lang F. Adhesion of annexin 7 deficient erythrocytes to endothelial cells. PLoS One 2013; 8:e56650. [PMID: 23437197 PMCID: PMC3577872 DOI: 10.1371/journal.pone.0056650] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 01/11/2013] [Indexed: 11/19/2022] Open
Abstract
Annexin 7 deficiency has previously been shown to foster suicidal death of erythrocytes or eryptosis, which is triggered by increase of intracellular Ca(2+) concentration ([Ca(2+)](i)) and characterized by cell shrinkage and cell membrane scrambling with subsequent phosphatidylserine exposure at the cell surface. Eryptosis following increase of [Ca(2+)](i) by Ca(2+) ionophore ionomycin, osmotic shock or energy depletion was more pronounced in erythrocytes from annexinA7-deficient mice (anxA7(-/-)) than in erythrocytes from wild type mice (anxA7(+/+)). As phosphatidylserine exposure is considered to mediate adhesion of erythrocytes to the vascular wall, the present study explored adhesion of erythrocytes from anx7(-/-) and anx7(+/+)-mice following increase of [Ca(2+)](i) by Ca(2+) ionophore ionomycin (1 µM for 30 min), hyperosmotic shock (addition of 550 mM sucrose for 2 hours) or energy depletion (removal of glucose for 12 hours). Phosphatidylserine exposing erythrocytes were identified by annexin V binding, cell volume estimated from forward scatter in FACS analysis and adhesion to human umbilical vein endothelial cells (HUVEC) utilizing a flow chamber. As a result, ionomycin, sucrose addition and glucose removal all triggered phosphatidylserine-exposure, decreased forward scatter and enhanced adhesion of erythrocytes to human umbilical vein endothelial cells (HUVEC), effects significantly more pronounced in anx7(-/-) than in anx7(+/+)-erythrocytes. Following ischemia, morphological renal injury was significantly higher in anx7(-/-) than in anx7(+/+)-mice. The present observations demonstrate that enhanced eryptosis of annexin7 deficient cells is paralleled by increased adhesion of erythrocytes to the vascular wall, an effect, which may impact on microcirculation during ischemia.
Collapse
Affiliation(s)
- Majed Abed
- Department of Physiology, Eberhard-Karls-University, Tuebingen, Germany
- Department of Physiology, Medicine Faculty, Al-Furat University, Deir Ezzor, Syria
| | | | | | - Christoph Daniel
- Institute of Pathology, Friedrich-Alexander-University, Erlangen-Nuernberg, Germany
| | - Kerstin Amann
- Institute of Pathology, Friedrich-Alexander-University, Erlangen-Nuernberg, Germany
| | - Florian Lang
- Department of Physiology, Eberhard-Karls-University, Tuebingen, Germany
| |
Collapse
|
33
|
Konsavage WM, Umstead TM, Wu Y, Phelps DS, Shenberger JS. Hyperoxia-induced alterations in the pulmonary proteome of juvenile rats. Exp Lung Res 2013; 39:107-17. [DOI: 10.3109/01902148.2013.763871] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Chlystun M, Campanella M, Law AL, Duchen MR, Fatimathas L, Levine TP, Gerke V, Moss SE. Regulation of mitochondrial morphogenesis by annexin A6. PLoS One 2013; 8:e53774. [PMID: 23341998 PMCID: PMC3544845 DOI: 10.1371/journal.pone.0053774] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 12/03/2012] [Indexed: 11/18/2022] Open
Abstract
Mitochondrial homeostasis is critical in meeting cellular energy demands, shaping calcium signals and determining susceptibility to apoptosis. Here we report a role for anxA6 in the regulation of mitochondrial morphogenesis, and show that in cells lacking anxA6 mitochondria are fragmented, respiration is impaired and mitochondrial membrane potential is reduced. In fibroblasts from AnxA6−/− mice, mitochondrial Ca2+ uptake is reduced and cytosolic Ca2+ transients are elevated. These observations led us to investigate possible interactions between anxA6 and proteins with roles in mitochondrial fusion and fission. We found that anxA6 associates with Drp1 and that mitochondrial fragmentation in AnxA6−/− fibroblasts was prevented by the Drp1 inhibitor mdivi-1. In normal cells elevation of intracellular Ca2+ disrupted the interaction between anxA6 and Drp1, displacing anxA6 to the plasma membrane and promoting mitochondrial fission. Our results suggest that anxA6 inhibits Drp1 activity, and that Ca2+-binding to anxA6 relieves this inhibition to permit Drp1-mediated mitochondrial fission.
Collapse
Affiliation(s)
- Marcin Chlystun
- Department of Cell Biology, University College London (UCL) Institute of Ophthalmology, London, United Kingdom
| | - Michelangelo Campanella
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
- Consortium for Mitochondrial Research (CfMR), University College London, London, United Kingdom
| | - Ah-Lai Law
- Department of Cell Biology, University College London (UCL) Institute of Ophthalmology, London, United Kingdom
| | - Michael R. Duchen
- Department of Cell and Developmental Biology, Mitochondrial Biology Group, University College London, London, United Kingdom
- Consortium for Mitochondrial Research (CfMR), University College London, London, United Kingdom
| | - Lux Fatimathas
- Department of Cell Biology, University College London (UCL) Institute of Ophthalmology, London, United Kingdom
| | - Tim P. Levine
- Department of Cell Biology, University College London (UCL) Institute of Ophthalmology, London, United Kingdom
| | - Volker Gerke
- University of Muenster, Institute of Medical Biochemistry, Muenster, Germany
| | - Stephen E. Moss
- Department of Cell Biology, University College London (UCL) Institute of Ophthalmology, London, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Wu CY, Taneyhill LA. Annexin a6 modulates chick cranial neural crest cell emigration. PLoS One 2012; 7:e44903. [PMID: 22984583 PMCID: PMC3439457 DOI: 10.1371/journal.pone.0044903] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 08/09/2012] [Indexed: 11/19/2022] Open
Abstract
The vertebrate neural crest is a population of migratory cells that originates in the dorsal aspect of the embryonic neural tube. These cells undergo an epithelial-to-mesencyhmal transition (EMT), delaminate from the neural tube and migrate extensively to generate an array of differentiated cell types. Elucidating the gene regulatory networks involved in neural crest cell induction, migration and differentiation are thus crucial to understanding vertebrate development. To this end, we have identified Annexin A6 as an important regulator of chick midbrain neural crest cell emigration. Annexin proteins comprise a family of calcium-dependent, membrane-binding molecules that mediate a variety of cellular and physiological processes including cell adhesion, migration and invasion. Our data indicate that Annexin A6 is expressed in the proper spatio-temporal pattern in the chick midbrain to play a potential role in neural crest cell ontogeny. To investigate Annexin A6 function, we have depleted or overexpressed Annexin A6 in the developing midbrain neural crest cell population. Our results show that knock-down or overexpression of Annexin A6 reduces or expands the migratory neural crest cell domain, respectively. Importantly, this phenotype is not due to any change in cell proliferation or cell death but can be correlated with changes in the size of the premigratory neural crest cell population and with markers associated with EMT. Taken together, our data indicate that Annexin A6 plays a pivotal role in modulating the formation of cranial migratory neural crest cells during vertebrate development.
Collapse
Affiliation(s)
- Chyong-Yi Wu
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Lisa A. Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
36
|
Kapustin AN, Shanahan CM. Calcium regulation of vascular smooth muscle cell-derived matrix vesicles. Trends Cardiovasc Med 2012; 22:133-7. [PMID: 22902179 DOI: 10.1016/j.tcm.2012.07.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 07/13/2012] [Accepted: 07/13/2012] [Indexed: 11/28/2022]
Abstract
Vascular calcification is a pathological process common in patients with disorders of mineral metabolism and mediated by vascular smooth muscle cells (VSMCs). A key event in the initiation of VSMC calcification is the release of mineralization-competent matrix vesicles (MVs), small membrane-bound bodies with structural features enabling them to efficiently nucleate hydroxyapatite. These bodies are similar to MVs secreted by chondrocytes during bone development and their properties include the absence of calcification inhibitors, formation of nucleation sites, and accumulation of matrix metalloproteinases such as MMP-2. The mechanisms of MV biogenesis and loading remain poorly understood; however, emerging data have demonstrated that alterations in cytosolic calcium homeostasis can trigger multiple changes in MV composition that promote their mineralization.
Collapse
Affiliation(s)
- Alexander N Kapustin
- BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | | |
Collapse
|
37
|
Koese M, Rentero C, Kota BP, Hoque M, Cairns R, Wood P, Vilà de Muga S, Reverter M, Alvarez-Guaita A, Monastyrskaya K, Hughes WE, Swarbrick A, Tebar F, Daly RJ, Enrich C, Grewal T. Annexin A6 is a scaffold for PKCα to promote EGFR inactivation. Oncogene 2012; 32:2858-72. [PMID: 22797061 DOI: 10.1038/onc.2012.303] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Protein kinase Cα (PKCα) can phosphorylate the epidermal growth factor receptor (EGFR) at threonine 654 (T654) to inhibit EGFR tyrosine phosphorylation (pY-EGFR) and the associated activation of downstream effectors. However, upregulation of PKCα in a large variety of cancers is not associated with EGFR inactivation, and factors determining the potential of PKCα to downregulate EGFR are yet unknown. Here, we show that ectopic expression of annexin A6 (AnxA6), a member of the Ca(2+) and phospholipid-binding annexins, strongly reduces pY-EGFR levels while augmenting EGFR T654 phosphorylation in EGFR overexpressing A431, head and neck and breast cancer cell lines. Reduced EGFR activation in AnxA6 expressing A431 cells is associated with reduced EGFR internalization and degradation. RNA interference (RNAi)-mediated PKCα knockdown in AnxA6 expressing A431 cells reduces T654-EGFR phosphorylation, but restores EGFR tyrosine phosphorylation, clonogenic growth and EGFR degradation. These findings correlate with AnxA6 interacting with EGFR, and elevated AnxA6 levels promoting PKCα membrane association and interaction with EGFR. Stable expression of the cytosolic N-terminal mutant AnxA6(1-175), which cannot promote PKCα membrane recruitment, does not increase T654-EGFR phosphorylation or the association of PKCα with EGFR. AnxA6 overexpression does not inhibit tyrosine phosphorylation of the T654A EGFR mutant, which cannot be phosphorylated by PKCα. Most strikingly, stable plasma membrane anchoring of AnxA6 is sufficient to recruit PKCα even in the absence of EGF or Ca(2+). In summary, AnxA6 is a new PKCα scaffold to promote PKCα-mediated EGFR inactivation through increased membrane targeting of PKCα and EGFR/PKCα complex formation.
Collapse
Affiliation(s)
- M Koese
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bandorowicz-Pikula J, Wos M, Pikula S. Do annexins participate in lipid messenger mediated intracellular signaling? A question revisited. Mol Membr Biol 2012; 29:229-42. [PMID: 22694075 DOI: 10.3109/09687688.2012.693210] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Annexins are physiologically important proteins that play a role in calcium buffering but also influence membrane structure, participate in Ca²⁺-dependent membrane repair events and in remodelling of the cytoskeleton. Thirty years ago several peptides isolated from lung perfusates, peritoneal leukocytes, neutrophiles and renal cells were proven inhibitory to the activity of phospholipase A₂. Those peptides were found to derive from structurally related proteins: annexins AnxA1 and AnxA2. These findings raised the question whether annexins may participate in regulation of the production of lipid second messengers and, therefore, modulate numerous lipid mediated signaling pathways in the cell. Recent advances in the field of annexins made also with the use of knock-out animal models revealed that these proteins are indeed important constituents of specific signaling pathways. In this review we provide evidence supporting the hypothesis that annexins, as membrane-binding proteins and organizers of the membrane lateral heterogeneity, may participate in lipid mediated signaling pathways by affecting the distribution and activity of lipid metabolizing enzymes (most of the reports point to phospholipase A₂) and of protein kinases regulating activity of these enzymes. Moreover, some experimental data suggest that annexins may directly interact with lipid metabolizing enzymes and, in a calcium-dependent or independent manner, with some of their substrates and products. On the basis of these observations, many investigators suggest that annexins are capable of linking Ca²⁺, redox and lipid signaling to coordinate vital cellular responses to the environmental stimuli.
Collapse
Affiliation(s)
- Joanna Bandorowicz-Pikula
- Laboratory of Cellular Metabolism, Department of Biochemistry, Nencki Institute of Experimental Biology, PL 02-093 Warsaw, Poland.
| | | | | |
Collapse
|
39
|
Domon M, Nasir MN, Matar G, Pikula S, Besson F, Bandorowicz-Pikula J. Annexins as organizers of cholesterol- and sphingomyelin-enriched membrane microdomains in Niemann-Pick type C disease. Cell Mol Life Sci 2012; 69:1773-85. [PMID: 22159585 PMCID: PMC11114673 DOI: 10.1007/s00018-011-0894-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 11/17/2011] [Accepted: 11/21/2011] [Indexed: 01/22/2023]
Abstract
Growing evidence suggests that membrane microdomains enriched in cholesterol and sphingomyelin are sites for numerous cellular processes, including signaling, vesicular transport, interaction with pathogens, and viral infection, etc. Recently some members of the annexin family of conserved calcium and membrane-binding proteins have been recognized as cholesterol-interacting molecules and suggested to play a role in the formation, stabilization, and dynamics of membrane microdomains to affect membrane lateral organization and to attract other proteins and signaling molecules onto their territory. Furthermore, annexins were implicated in the interactions between cytosolic and membrane molecules, in the turnover and storage of cholesterol and in various signaling pathways. In this review, we focus on the mechanisms of interaction of annexins with lipid microdomains and the role of annexins in membrane microdomains dynamics including possible participation of the domain-associated forms of annexins in the etiology of human lysosomal storage disease called Niemann-Pick type C disease, related to the abnormal storage of cholesterol in the lysosome-like intracellular compartment. The involvement of annexins and cholesterol/sphingomyelin-enriched membrane microdomains in other pathologies including cardiac dysfunctions, neurodegenerative diseases, obesity, diabetes mellitus, and cancer is likely, but is not supported by substantial experimental observations, and therefore awaits further clarification.
Collapse
Affiliation(s)
- Magdalena Domon
- Laboratory of Lipid Biochemistry, Department of Biochemistry, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093, Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
40
|
PAPAKOSTAS SPIROS, VASEMÄGI ANTI, VÄHÄ JUHAPEKKA, HIMBERG MIKAEL, PEIL LAURI, PRIMMER CRAIGR. A proteomics approach reveals divergent molecular responses to salinity in populations of European whitefish (Coregonus lavaretus). Mol Ecol 2012; 21:3516-30. [DOI: 10.1111/j.1365-294x.2012.05553.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
41
|
Reverter M, Rentero C, de Muga SV, Alvarez-Guaita A, Mulay V, Cairns R, Wood P, Monastyrskaya K, Pol A, Tebar F, Blasi J, Grewal T, Enrich C. Cholesterol transport from late endosomes to the Golgi regulates t-SNARE trafficking, assembly, and function. Mol Biol Cell 2012; 22:4108-23. [PMID: 22039070 PMCID: PMC3204072 DOI: 10.1091/mbc.e11-04-0332] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study shows that impaired cholesterol egress from late endosomes in cells with high annexin A6 levels is associated with altered soluble N-ethylmaleimide–sensitive fusion protein 23 (SNAP23) and syntaxin-4 cellular distribution and assembly and accumulation in Golgi membranes. This correlates with reduced secretion of cargo along the constitutive and SNAP23/syntaxin-4–dependent secretory pathway. Cholesterol regulates plasma membrane (PM) association and functioning of syntaxin-4 and soluble N-ethylmaleimide-sensitive fusion protein 23 (SNAP23) in the secretory pathway. However, the molecular mechanism and cellular cholesterol pools that determine the localization and assembly of these target membrane SNAP receptors (t-SNAREs) are largely unknown. We recently demonstrated that high levels of annexin A6 (AnxA6) induce accumulation of cholesterol in late endosomes, thereby reducing cholesterol in the Golgi and PM. This leads to an impaired supply of cholesterol needed for cytosolic phospholipase A2 (cPLA2) to drive Golgi vesiculation and caveolin transport to the cell surface. Using AnxA6-overexpressing cells as a model for cellular cholesterol imbalance, we identify impaired cholesterol egress from late endosomes and diminution of Golgi cholesterol as correlating with the sequestration of SNAP23/syntaxin-4 in Golgi membranes. Pharmacological accumulation of late endosomal cholesterol and cPLA2 inhibition induces a similar phenotype in control cells with low AnxA6 levels. Ectopic expression of Niemann-Pick C1 (NPC1) or exogenous cholesterol restores the location of SNAP23 and syntaxin-4 within the PM. Importantly, AnxA6-mediated mislocalization of these t-SNAREs correlates with reduced secretion of cargo via the SNAP23/syntaxin-4–dependent constitutive exocytic pathway. We thus conclude that inhibition of late endosomal export and Golgi cholesterol depletion modulate t-SNARE localization and functioning along the exocytic pathway.
Collapse
Affiliation(s)
- Meritxell Reverter
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Human annexin A6 interacts with influenza a virus protein M2 and negatively modulates infection. J Virol 2011; 86:1789-801. [PMID: 22114333 DOI: 10.1128/jvi.06003-11] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The influenza A virus M2 ion channel protein has the longest cytoplasmic tail (CT) among the three viral envelope proteins and is well conserved between different viral strains. It is accessible to the host cellular machinery after fusion with the endosomal membrane and during the trafficking, assembly, and budding processes. We hypothesized that identification of host cellular interactants of M2 CT could help us to better understand the molecular mechanisms regulating the M2-dependent stages of the virus life cycle. Using yeast two-hybrid screening with M2 CT as bait, a novel interaction with the human annexin A6 (AnxA6) protein was identified, and their physical interaction was confirmed by coimmunoprecipitation assay and a colocalization study of virus-infected human cells. We found that small interfering RNA (siRNA)-mediated knockdown of AnxA6 expression significantly increased virus production, while its overexpression could reduce the titer of virus progeny, suggesting a negative regulatory role for AnxA6 during influenza A virus infection. Further characterization revealed that AnxA6 depletion or overexpression had no effect on the early stages of the virus life cycle or on viral RNA replication but impaired the release of progeny virus, as suggested by delayed or defective budding events observed at the plasma membrane of virus-infected cells by transmission electron microscopy. Collectively, this work identifies AnxA6 as a novel cellular regulator that targets and impairs the virus budding and release stages of the influenza A virus life cycle.
Collapse
|
43
|
Abstract
Ever since their discovery as cellular counterparts of viral oncogenes more than 25 years ago, much progress has been made in understanding the complex networks of signal transduction pathways activated by oncogenic Ras mutations in human cancers. The activity of Ras is regulated by nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs), and much emphasis has been put into the biochemical and structural analysis of the Ras/GAP complex. The mechanisms by which GAPs catalyze Ras-GTP hydrolysis have been clarified and revealed that oncogenic Ras mutations confer resistance to GAPs and remain constitutively active. However, it is yet unclear how cells coordinate the large and divergent GAP protein family to promote Ras inactivation and ensure a certain biological response. Different domain arrangements in GAPs to create differential protein-protein and protein-lipid interactions are probably key factors determining the inactivation of the 3 Ras isoforms H-, K-, and N-Ras and their effector pathways. In recent years, in vitro as well as cell- and animal-based studies examining GAP activity, localization, interaction partners, and expression profiles have provided further insights into Ras inactivation and revealed characteristics of several GAPs to exert specific and distinct functions. This review aims to summarize knowledge on the cell biology of RasGAP proteins that potentially contributes to differential regulation of spatiotemporal Ras signaling.
Collapse
Affiliation(s)
- Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
44
|
Reverter M, Rentero C, de Muga SV, Alvarez-Guaita A, Mulay V, Cairns R, Wood P, Monastyrskaya K, Pol A, Tebar F, Blasi J, Grewal T, Enrich C. Cholesterol transport from late endosomes to the Golgi regulates t-SNARE trafficking, assembly, and function. Mol Biol Cell 2011. [DOI: 10.1091/mbc.e11-04-0332r] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cholesterol regulates plasma membrane (PM) association and functioning of syntaxin-4 and soluble N-ethylmaleimide-sensitive fusion protein 23 (SNAP23) in the secretory pathway. However, the molecular mechanism and cellular cholesterol pools that determine the localization and assembly of these target membrane SNAP receptors (t-SNAREs) are largely unknown. We recently demonstrated that high levels of annexin A6 (AnxA6) induce accumulation of cholesterol in late endosomes, thereby reducing cholesterol in the Golgi and PM. This leads to an impaired supply of cholesterol needed for cytosolic phospholipase A2(cPLA2) to drive Golgi vesiculation and caveolin transport to the cell surface. Using AnxA6-overexpressing cells as a model for cellular cholesterol imbalance, we identify impaired cholesterol egress from late endosomes and diminution of Golgi cholesterol as correlating with the sequestration of SNAP23/syntaxin-4 in Golgi membranes. Pharmacological accumulation of late endosomal cholesterol and cPLA2inhibition induces a similar phenotype in control cells with low AnxA6 levels. Ectopic expression of Niemann-Pick C1 (NPC1) or exogenous cholesterol restores the location of SNAP23 and syntaxin-4 within the PM. Importantly, AnxA6-mediated mislocalization of these t-SNAREs correlates with reduced secretion of cargo via the SNAP23/syntaxin-4–dependent constitutive exocytic pathway. We thus conclude that inhibition of late endosomal export and Golgi cholesterol depletion modulate t-SNARE localization and functioning along the exocytic pathway.
Collapse
Affiliation(s)
- Meritxell Reverter
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Carles Rentero
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sandra Vilà de Muga
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Anna Alvarez-Guaita
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Vishwaroop Mulay
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Rose Cairns
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Peta Wood
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Katia Monastyrskaya
- Urology Research Laboratory, Department of Clinical Research, University of Bern, 3000 Bern 9, Switzerland
| | - Albert Pol
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Francesc Tebar
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Joan Blasi
- Department of Pathology and Experimental Therapeutics, IDIBELL–University of Barcelona, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia
| | - Carlos Enrich
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
45
|
Baltgalvis KA, Jaeger MA, Fitzsimons DP, Thayer SA, Lowe DA, Ervasti JM. Transgenic overexpression of γ-cytoplasmic actin protects against eccentric contraction-induced force loss in mdx mice. Skelet Muscle 2011; 1:32. [PMID: 21995957 PMCID: PMC3214766 DOI: 10.1186/2044-5040-1-32] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2011] [Accepted: 10/13/2011] [Indexed: 11/22/2022] Open
Abstract
Background γ-cytoplasmic (γ-cyto) actin levels are elevated in dystrophin-deficient mdx mouse skeletal muscle. The purpose of this study was to determine whether further elevation of γ-cyto actin levels improve or exacerbate the dystrophic phenotype of mdx mice. Methods We transgenically overexpressed γ-cyto actin, specifically in skeletal muscle of mdx mice (mdx-TG), and compared skeletal muscle pathology and force-generating capacity between mdx and mdx-TG mice at different ages. We investigated the mechanism by which γ-cyto actin provides protection from force loss by studying the role of calcium channels and stretch-activated channels in isolated skeletal muscles and muscle fibers. Analysis of variance or independent t-tests were used to detect statistical differences between groups. Results Levels of γ-cyto actin in mdx-TG skeletal muscle were elevated 200-fold compared to mdx skeletal muscle and incorporated into thin filaments. Overexpression of γ-cyto actin had little effect on most parameters of mdx muscle pathology. However, γ-cyto actin provided statistically significant protection against force loss during eccentric contractions. Store-operated calcium entry across the sarcolemma did not differ between mdx fibers compared to wild-type fibers. Additionally, the omission of extracellular calcium or the addition of streptomycin to block stretch-activated channels did not improve the force-generating capacity of isolated extensor digitorum longus muscles from mdx mice during eccentric contractions. Conclusions The data presented in this study indicate that upregulation of γ-cyto actin in dystrophic skeletal muscle can attenuate force loss during eccentric contractions and that the mechanism is independent of activation of stretch-activated channels and the accumulation of extracellular calcium.
Collapse
Affiliation(s)
- Kristen A Baltgalvis
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Cornely R, Rentero C, Enrich C, Grewal T, Gaus K. Annexin A6 is an organizer of membrane microdomains to regulate receptor localization and signalling. IUBMB Life 2011; 63:1009-17. [PMID: 21990038 DOI: 10.1002/iub.540] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 06/16/2011] [Indexed: 12/13/2022]
Abstract
Annexin A6 (AnxA6) belongs to the conserved annexin protein family--a group of Ca(2+) -dependent membrane binding proteins. It is the largest of all annexin proteins and upon activation, binds to negatively charged phospholipids in the plasma membrane and endosomes. In addition, AnxA6 associates with cholesterol-rich membrane microdomains termed lipid rafts. Membrane cholesterol triggers Ca(2+) -independent translocation of AnxA6 to membranes and AnxA6 levels determine the number of caveolae, a form of specialized rafts at the cell surface. AnxA6 also has an F-actin binding domain and interacts with cytoskeleton components. Taken together, this suggests that AnxA6 has a scaffold function to link membrane microdomains with the organization of the cytoskeleton. Such a link facilitates AnxA6 to participate in plasma membrane repair and it would also impact on receptor signalling at the cell surface, growth factor, and lipoprotein receptor trafficking, Ca(2+) -channel activity and T cell activation. Hence, the regulation of cell surface receptors by AnxA6 may be facilitated by its unique structure that allows recruitment of interaction partners and simultaneously bridging specialized membrane domains with cortical actin surrounding activated receptors.
Collapse
Affiliation(s)
- Rhea Cornely
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | | | | | | | | |
Collapse
|
47
|
In vitro maturation of the cisternal organelle in the hippocampal neuron's axon initial segment. Mol Cell Neurosci 2011; 48:104-16. [PMID: 21708259 DOI: 10.1016/j.mcn.2011.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 06/07/2011] [Accepted: 06/09/2011] [Indexed: 11/21/2022] Open
Abstract
Regulation of Ca(2+) concentrations is essential to maintain the structure and function of the axon initial segment (AIS). The so-called cisternal organelle of the AIS is a structure involved in this regulation, although little is known as to how this organelle matures and is stabilized. Here we describe how the cisternal organelle develops in cultured hippocampal neurons and the interactions that facilitate its stabilization in the AIS. We also characterize the developmental expression of molecules involved in Ca(2+) regulation in the AIS. Our results indicate that synaptopodin (synpo) positive elements considered to be associated to the cisternal organelle are present in the AIS after six days in vitro. There are largely overlapping microdomains containing the inositol 1,4,5-triphosphate receptor 1 (IP(3)R1) and the Ca(2+) binding protein annexin 6, suggesting that the regulation of Ca(2+) concentrations in the AIS is sensitive to IP(3) and subject to regulation by annexin 6. The expression of synpo, IP(3)R1 and annexin 6 in the AIS is independent of the neuron activity, as it was unaffected by tetrodotoxin blockage of action potentials and it was resistant to detergent extraction, indicating that these proteins interact with scaffolding and/or cytoskeleton proteins. The presence of ankyrin G seems to be required for the acquisition and maintenance of the cisternal organelle, while the integrity of the actin cytoskeleton must be maintained for the expression IP(3)R1 and annexin 6 to persist in the AIS.
Collapse
|
48
|
Kapustin AN, Davies JD, Reynolds JL, McNair R, Jones GT, Sidibe A, Schurgers LJ, Skepper JN, Proudfoot D, Mayr M, Shanahan CM. Calcium regulates key components of vascular smooth muscle cell-derived matrix vesicles to enhance mineralization. Circ Res 2011; 109:e1-12. [PMID: 21566214 DOI: 10.1161/circresaha.110.238808] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
RATIONALE Matrix vesicles (MVs) are specialized structures that initiate mineral nucleation during physiological skeletogenesis. Similar vesicular structures are deposited at sites of pathological vascular calcification, and studies in vitro have shown that elevated levels of extracellular calcium (Ca) can induce mineralization of vascular smooth muscle cell (VSMC)-derived MVs. OBJECTIVES To determine the mechanisms that promote mineralization of VSMC-MVs in response to calcium stress. METHODS AND RESULTS Transmission electron microscopy showed that both nonmineralized and mineralized MVs were abundantly deposited in the extracellular matrix at sites of calcification. Using cultured human VSMCs, we showed that MV mineralization is calcium dependent and can be inhibited by BAPTA-AM. MVs released by VSMCs in response to extracellular calcium lacked the key mineralization inhibitor matrix Gla protein and showed enhanced matrix metalloproteinase-2 activity. Proteomics revealed that VSMC-MVs share similarities with chondrocyte-derived MVs, including enrichment of the calcium-binding proteins annexins (Anx) A2, A5, and A6. Biotin cross-linking and flow cytometry demonstrated that in response to calcium, AnxA6 shuttled to the plasma membrane and was selectively enriched in MVs. AnxA6 was also abundant at sites of vascular calcification in vivo, and small interfering RNA depletion of AnxA6 reduced VSMC mineralization. Flow cytometry showed that in addition to AnxA6, calcium induced phosphatidylserine exposure on the MV surface, thus providing hydroxyapatite nucleation sites. CONCLUSIONS In contrast to the coordinated signaling response observed in chondrocyte MVs, mineralization of VSMC-MVs is a pathological response to disturbed intracellular calcium homeostasis that leads to inhibitor depletion and the formation of AnxA6/phosphatidylserine nucleation complexes.
Collapse
Affiliation(s)
- Alexander N Kapustin
- British Heart Foundation Centre, Cardiovascular Division, Kings College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Potez S, Luginbühl M, Monastyrskaya K, Hostettler A, Draeger A, Babiychuk EB. Tailored protection against plasmalemmal injury by annexins with different Ca2+ sensitivities. J Biol Chem 2011; 286:17982-91. [PMID: 21454475 DOI: 10.1074/jbc.m110.187625] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The annexins, a family of Ca(2+)- and lipid-binding proteins, are involved in a range of intracellular processes. Recent findings have implicated annexin A1 in the resealing of plasmalemmal injuries. Here, we demonstrate that another member of the annexin protein family, annexin A6, is also involved in the repair of plasmalemmal lesions induced by a bacterial pore-forming toxin, streptolysin O. An injury-induced elevation in the intracellular concentration of Ca(2+) ([Ca(2+)](i)) triggers plasmalemmal repair. The highly Ca(2+)-sensitive annexin A6 responds faster than annexin A1 to [Ca(2+)](i) elevation. Correspondingly, a limited plasmalemmal injury can be promptly countered by annexin A6 even without the participation of annexin A1. However, its high Ca(2+) sensitivity makes annexin A6 highly amenable to an unproductive binding to the uninjured plasmalemma; during an extensive injury accompanied by a massive elevation in [Ca(2+)](i), its active pool is severely depleted. In contrast, annexin A1 with a much lower Ca(2+) sensitivity is ineffective at the early stages of injury; however, it remains available for the repair even at high [Ca(2+)](i). Our findings highlight the role of the annexins in the process of plasmalemmal repair; a number of annexins with different Ca(2+)-sensitivities provide a cell with the means to react promptly to a limited injury in its early stages and, at the same time, to withstand a sustained injury accompanied by the continuous formation of plasmalemmal lesions.
Collapse
Affiliation(s)
- Sarah Potez
- Department of Cell Biology, Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
50
|
Podszywalow-Bartnicka P, Kosiorek M, Piwocka K, Sikora E, Zablocki K, Pikula S. Role of annexin A6 isoforms in catecholamine secretion by PC12 cells: distinct influence on calcium response. J Cell Biochem 2011; 111:168-78. [PMID: 20506562 DOI: 10.1002/jcb.22685] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Noradrenaline and adrenaline are secreted by adrenal medulla chromaffin cells via exocytosis. Exocytosis of catecholamines occurs after cell stimulation with various endogenous activators such as nicotine or after depolarization of the plasma membrane and is regulated by calcium ions. Cytosolic [Ca(2+)] increases in response to cell excitation and triggers a signal-initiated secretion. Annexins are known to participate in the regulation of membrane dynamics and are also considered to be involved in vesicular trafficking. Some experimental evidence suggests that annexins may participate in Ca(2+)-regulated catecholamine secretion. In this report the effect of annexin A6 (AnxA6) isoforms 1 and 2 on catecholamine secretion has been described. Overexpression of AnxA6 isoforms and AnxA6 knock-down in PC12 cells were accompanied by almost complete inhibition or a 20% enhancement of dopamine secretion, respectively. AnxA6-1 and AnxA6-2 overexpression reduced Delta[Ca(2+)](c) upon depolarization by 32% and 58%, respectively, while AnxA6 knock-down increased Delta[Ca(2+)](c) by 44%. The mechanism of AnxA6 action on Ca(2+) signalling is not well understood. Experimental evidence suggests that two AnxA6 isoforms interact with different targets engaged in regulation of calcium homeostasis in PC12 cells.
Collapse
|