101
|
Mehta N, Zhang D, Li R, Wang T, Gava A, Parthasarathy P, Gao B, Krepinsky JC. Caveolin-1 regulation of Sp1 controls production of the antifibrotic protein follistatin in kidney mesangial cells. Cell Commun Signal 2019; 17:37. [PMID: 30995923 PMCID: PMC6472091 DOI: 10.1186/s12964-019-0351-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND We previously showed that caveolin-1 (cav-1), an integral membrane protein, is required for the synthesis of matrix proteins by glomerular mesangial cells (MC). In a previous study to understand how cav-1 is involved in regulating matrix production, we had identified significant upregulation of the antifibrotic protein follistatin in cav-1 knockout MC. Follistatin inhibits the profibrotic effects of several members of the transforming growth factor beta superfamily, in particular the activins. Here, we characterize the molecular mechanism through which cav-1 regulates the expression of follistatin. METHODS Kidneys from cav-1 wild type and knockout (KO) mice were analyzed and primary cultures of MC from cav-1 wild-type and KO mice were utilized. FST promoter deletion constructs were generated to determine the region of the promoter important for mediating FST upregulation in cav-1 KO MC. siRNA-mediated down-regulation and overexpression of Sp1 in conjunction with luciferase activity assays, immunoprecipitation, western blotting and ChiP was used to assess the role of Sp1 in transcriptionally regulating FST expression. Pharmacologic kinase inhibitors and specific siRNA were used to determine the post-translational mechanism through which cav-1 affects Sp1 activity. RESULTS Our results establish that follistatin upregulation occurs at the transcript level. We identified Sp1 as the critical transcription factor regulating activation of the FST promoter in cav-1 KO MC through binding to a region within 123 bp of the transcription start site. We further determined that the lack of cav-1 increases Sp1 nuclear levels and transcriptional activity. This occurred through increased phosphoinositide 3-kinase (PI3K) activity and downstream protein kinase C (PKC) zeta-mediated phosphorylation and activation of Sp1. CONCLUSIONS These findings shed light on the transcriptional mechanism by which cav-1 represses the expression of a major antifibrotic protein, and can inform the development of novel antifibrotic treatment strategies.
Collapse
Affiliation(s)
- Neel Mehta
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Dan Zhang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Renzhong Li
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Tony Wang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Agata Gava
- Physiological Sciences Graduate Program, Health Sciences Centre, Federal University of Espirito Santo, Vitoria, Brazil
| | | | - Bo Gao
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Joan C Krepinsky
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada. .,St. Joseph's Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON, L8N 4A6, Canada.
| |
Collapse
|
102
|
Capera J, Serrano-Novillo C, Navarro-Pérez M, Cassinelli S, Felipe A. The Potassium Channel Odyssey: Mechanisms of Traffic and Membrane Arrangement. Int J Mol Sci 2019; 20:ijms20030734. [PMID: 30744118 PMCID: PMC6386995 DOI: 10.3390/ijms20030734] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/29/2022] Open
Abstract
Ion channels are transmembrane proteins that conduct specific ions across biological membranes. Ion channels are present at the onset of many cellular processes, and their malfunction triggers severe pathologies. Potassium channels (KChs) share a highly conserved signature that is necessary to conduct K⁺ through the pore region. To be functional, KChs require an exquisite regulation of their subcellular location and abundance. A wide repertoire of signatures facilitates the proper targeting of the channel, fine-tuning the balance that determines traffic and location. These signature motifs can be part of the secondary or tertiary structure of the protein and are spread throughout the entire sequence. Furthermore, the association of the pore-forming subunits with different ancillary proteins forms functional complexes. These partners can modulate traffic and activity by adding their own signatures as well as by exposing or masking the existing ones. Post-translational modifications (PTMs) add a further dimension to traffic regulation. Therefore, the fate of a KCh is not fully dependent on a gene sequence but on the balance of many other factors regulating traffic. In this review, we assemble recent evidence contributing to our understanding of the spatial expression of KChs in mammalian cells. We compile specific signatures, PTMs, and associations that govern the destination of a functional channel.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Clara Serrano-Novillo
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Silvia Cassinelli
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| |
Collapse
|
103
|
Thangavel C, Gomes CM, Zderic SA, Javed E, Addya S, Singh J, Das S, Birbe R, Den RB, Rattan S, Deshpande DA, Penn RB, Chacko S, Boopathi E. NF-κB and GATA-Binding Factor 6 Repress Transcription of Caveolins in Bladder Smooth Muscle Hypertrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:847-867. [PMID: 30707892 DOI: 10.1016/j.ajpath.2018.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 12/03/2018] [Accepted: 12/17/2018] [Indexed: 12/26/2022]
Abstract
Caveolins (CAVs) are structural proteins of caveolae that function as signaling platforms to regulate smooth muscle contraction. Loss of CAV protein expression is associated with impaired contraction in obstruction-induced bladder smooth muscle (BSM) hypertrophy. In this study, microarray analysis of bladder RNA revealed down-regulation of CAV1, CAV2, and CAV3 gene transcription in BSM from models of obstructive bladder disease in mice and humans. We identified and characterized regulatory regions responsible for CAV1, CAV2, and CAV3 gene expression in mice with obstruction-induced BSM hypertrophy, and in men with benign prostatic hyperplasia. DNA affinity chromatography and chromatin immunoprecipitation assays revealed a greater increase in binding of GATA-binding factor 6 (GATA-6) and NF-κB to their cognate binding motifs on CAV1, CAV2, and CAV3 promoters in obstructed BSM relative to that observed in control BSM. Knockout of NF-κB subunits, shRNA-mediated knockdown of GATA-6, or pharmacologic inhibition of GATA-6 and NF-κB in BSM increased CAV1, CAV2, and CAV3 transcription and promoter activity. Conversely, overexpression of GATA-6 decreased CAV2 and CAV3 transcription and promoter activity. Collectively, these data provide new insight into the mechanisms by which CAV gene expression is repressed in hypertrophied BSM in obstructive bladder disease.
Collapse
Affiliation(s)
| | - Cristiano M Gomes
- Division of Urology, University of Sao Paulo School of Medicine, Hospital das Clinicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Stephen A Zderic
- Department of Urology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elham Javed
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sankar Addya
- Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jagmohan Singh
- Division of Gastroenterology and Hepatology, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sreya Das
- Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ruth Birbe
- Department of Pathology and Laboratory Medicine, Cooper University Health Care, Camden, New Jersey
| | - Robert B Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Satish Rattan
- Division of Gastroenterology and Hepatology, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Deepak A Deshpande
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Raymond B Penn
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Samuel Chacko
- Division of Urology, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ettickan Boopathi
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania; Division of Urology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
104
|
Huang Q, Zhong W, Hu Z, Tang X. A review of the role of cav-1 in neuropathology and neural recovery after ischemic stroke. J Neuroinflammation 2018; 15:348. [PMID: 30572925 PMCID: PMC6302517 DOI: 10.1186/s12974-018-1387-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke starts a series of pathophysiological processes that cause brain injury. Caveolin-1 (cav-1) is an integrated protein and locates at the caveolar membrane. It has been demonstrated that cav-1 can protect blood–brain barrier (BBB) integrity by inhibiting matrix metalloproteases (MMPs) which degrade tight junction proteins. This article reviews recent developments in understanding the mechanisms underlying BBB dysfunction, neuroinflammation, and oxidative stress after ischemic stroke, and focuses on how cav-1 modulates a series of activities after ischemic stroke. In general, cav-1 reduces BBB permeability mainly by downregulating MMP9, reduces neuroinflammation through influencing cytokines and inflammatory cells, promotes nerve regeneration and angiogenesis via cav-1/VEGF pathway, reduces apoptosis, and reduces the damage mediated by oxidative stress. In addition, we also summarize some experimental results that are contrary to the above and explore possible reasons for these differences.
Collapse
Affiliation(s)
- Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China.
| |
Collapse
|
105
|
Krishna A, Sengupta D. Interplay between Membrane Curvature and Cholesterol: Role of Palmitoylated Caveolin-1. Biophys J 2018; 116:69-78. [PMID: 30579563 DOI: 10.1016/j.bpj.2018.11.3127] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/14/2018] [Accepted: 11/01/2018] [Indexed: 01/07/2023] Open
Abstract
Caveolin-1 (cav-1) is an important player in cell signaling and endocytosis that has been shown to colocalize with cholesterol-rich membrane domains. Experimental studies with varying cav-1 constructs have suggested that it can induce both cholesterol clustering and membrane curvature. Here, we probe the molecular origin of membrane curvature and cholesterol clustering by cav-1 by using coarse-grain molecular dynamics simulations. We have performed a series of simulations of a functionally important cav-1 construct, comprising the membrane-interacting domains and a C-terminal palmitoyl tail. Our results suggest that cav-1 is able to induce cholesterol clustering in the membrane leaflet to which it is bound as well as the opposing leaflet. A positive membrane curvature is observed upon cav-1 binding in cholesterol-containing bilayers. Interestingly, we observe an interplay between cholesterol clustering and membrane curvature such that cav-1 is able to induce higher membrane curvature in cholesterol-rich membranes. The role of the cav-1 palmitoyl tail is less clear and appears to increase the membrane contacts. Further, we address the importance of the secondary structure of cav-1 domains and show that it could play an important role in membrane curvature and cholesterol clustering. Our work is an important step toward a molecular picture of caveolae and vesicular endocytosis.
Collapse
Affiliation(s)
- Anjali Krishna
- CSIR-National Chemical Laboratory, Pune, Maharashtra, India
| | - Durba Sengupta
- CSIR-National Chemical Laboratory, Pune, Maharashtra, India.
| |
Collapse
|
106
|
Vaidyanathan R, Reilly L, Eckhardt LL. Caveolin-3 Microdomain: Arrhythmia Implications for Potassium Inward Rectifier and Cardiac Sodium Channel. Front Physiol 2018; 9:1548. [PMID: 30473666 PMCID: PMC6238080 DOI: 10.3389/fphys.2018.01548] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/16/2018] [Indexed: 11/13/2022] Open
Abstract
In human cardiac ventricular myocytes, caveolin-3 functions as a scaffolding and regulatory protein for signaling molecules and compartmentalizes ion channels. Our lab has recently explored this sub-cellular microdomain and found that potassium inward rectifier Kir2.x is found in association with caveolin-3. The three cardiac Kir2.x isoforms (Kir2.1, Kir2.2, and Kir2.3) are the molecular correlates of IK1 in the heart, of which Kir2.1 is the dominant isoform in the ventricle. Kir2.1 channels assemble with Kir2.2 and Kir2.3 forming hetero-tetramers that modulate IK1. IK1 sets the resting membrane potential and assists with terminal phase 3 ventricular repolarization. In our studies using native human ventricular tissue, Kir2.x co-localizes with caveolin-3 and significance of the association between Kir2.x and caveolin-3 is emphasized in relation to mutations in the gene which encodes caveolin-3, CAV3, associated with Long QT Syndrome 9 (LQT9). LQT9-associated CAV3 mutations cause decreased current density in Kir2.1 and Kir2.2 as homomeric and heteromeric channels, which affects repolarization and membrane potential stability. A portion of Kir2.1 cardiac localization parallels that of the cardiac sodium channel (Nav1.5). This may have implications for Long QT9 in which CAV3 mutations cause an increase in the late current of Nav1.5 (INa-L) via nNOS mediated nitrosylation of Nav1.5. In iPS-CMs, expression of LQT9 CAV3 mutations resulted in action potential duration (APD) prolongation and early-after depolarizations (EADs), supporting the arrhythmogenicity of LQT9. To evaluate the combined effect of the CAV3 mutants on INa-L and IK1, we studied both ventricular and Purkinje myocyte mathematical modeling. Interestingly, mathematical ventricular myocytes, similar to iPS-CMs, demonstrated EADs but no sustained arrhythmia. In contrast, Purkinje modeling demonstrated delayed-after depolarizations (DADs) driven mechanism for sustained arrhythmia, dependent on the combined loss of IK1 and gain of INa-L. This finding changes the overall assumed arrhythmia phenotype for LQT9. In future studies, we are exploring caveolar micro-domain disruption in heart failure and how this effects Kir2.x and Nav1.5. Here we review the caveolae cardiac microdomain of Kir2.x and Nav1.5 and explore some of the downstream effects of caveolin-3 and caveolae disruption in specific clinical scenarios.
Collapse
Affiliation(s)
- Ravi Vaidyanathan
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Louise Reilly
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Lee L Eckhardt
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
107
|
Bryant SM, Kong CHT, Watson JJ, Gadeberg HC, Roth DM, Patel HH, Cannell MB, James AF, Orchard CH. Caveolin-3 KO disrupts t-tubule structure and decreases t-tubular I Ca density in mouse ventricular myocytes. Am J Physiol Heart Circ Physiol 2018; 315:H1101-H1111. [PMID: 30028203 PMCID: PMC6415741 DOI: 10.1152/ajpheart.00209.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/25/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023]
Abstract
Caveolin-3 (Cav-3) is a protein that has been implicated in t-tubule formation and function in cardiac ventricular myocytes. In cardiac hypertrophy and failure, Cav-3 expression decreases, t-tubule structure is disrupted, and excitation-contraction coupling is impaired. However, the extent to which the decrease in Cav-3 expression underlies these changes is unclear. We therefore investigated the structure and function of myocytes isolated from the hearts of Cav-3 knockout (KO) mice. These mice showed cardiac dilatation and decreased ejection fraction in vivo compared with wild-type control mice. Isolated KO myocytes showed cellular hypertrophy, altered t-tubule structure, and decreased L-type Ca2+ channel current ( ICa) density. This decrease in density occurred predominantly in the t-tubules, with no change in total ICa, and was therefore a consequence of the increase in membrane area. Cav-3 KO had no effect on L-type Ca2+ channel expression, and C3SD peptide, which mimics the scaffolding domain of Cav-3, had no effect on ICa in KO myocytes. However, inhibition of PKA using H-89 decreased ICa at the surface and t-tubule membranes in both KO and wild-type myocytes. Cav-3 KO had no significant effect on Na+/Ca2+ exchanger current or Ca2+ release. These data suggest that Cav-3 KO causes cellular hypertrophy, thereby decreasing t-tubular ICa density. NEW & NOTEWORTHY Caveolin-3 (Cav-3) is a protein that inhibits hypertrophic pathways, has been implicated in the formation and function of cardiac t-tubules, and shows decreased expression in heart failure. This study demonstrates that Cav-3 knockout mice show cardiac dysfunction in vivo, while isolated ventricular myocytes show cellular hypertrophy, changes in t-tubule structure, and decreased t-tubular L-type Ca2+ current density, suggesting that decreased Cav-3 expression contributes to these changes in cardiac hypertrophy and failure.
Collapse
MESH Headings
- Action Potentials
- Animals
- Calcium Channels, L-Type/metabolism
- Calcium Signaling
- Caveolin 3/deficiency
- Caveolin 3/genetics
- Down-Regulation
- Genetic Predisposition to Disease
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phenotype
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
Collapse
Affiliation(s)
- Simon M Bryant
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol , Bristol , United Kingdom
| | - Cherrie H T Kong
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol , Bristol , United Kingdom
| | - Judy J Watson
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol , Bristol , United Kingdom
| | - Hanne C Gadeberg
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol , Bristol , United Kingdom
| | - David M Roth
- Veterans Affairs San Diego Healthcare System and Department of Anesthesiology, University of California-San Diego , La Jolla, California
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System and Department of Anesthesiology, University of California-San Diego , La Jolla, California
| | - Mark B Cannell
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol , Bristol , United Kingdom
| | - Andrew F James
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol , Bristol , United Kingdom
| | - Clive H Orchard
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol , Bristol , United Kingdom
| |
Collapse
|
108
|
Pérez-Verdaguer M, Capera J, Ortego-Domínguez M, Bielanska J, Comes N, Montoro RJ, Camps M, Felipe A. Caveolar targeting links Kv1.3 with the insulin-dependent adipocyte physiology. Cell Mol Life Sci 2018; 75:4059-4075. [PMID: 29947924 PMCID: PMC11105548 DOI: 10.1007/s00018-018-2851-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 12/23/2022]
Abstract
The voltage-dependent potassium channel Kv1.3 participates in peripheral insulin sensitivity. Genetic ablation of Kv1.3 triggers resistance to diet-induced weight gain, thereby pointing to this protein as a pharmacological target for obesity and associated type II diabetes. However, this role is under intense debate because Kv1.3 expression in adipose tissue raises controversy. We demonstrated that Kv1.3 is expressed in white adipose tissue from humans and rodents. Moreover, other channels, such as Kv1.1, Kv1.2, Kv1.4 and especially Kv1.5, from the same Shaker family are also present. Although elevated insulin levels and adipogenesis remodel the Kv phenotype, which could lead to multiple heteromeric complexes, Kv1.3 markedly participates in the insulin-dependent regulation of glucose uptake in mature adipocytes. Adipocyte differentiation increased the expression of Kv1.3, which is targeted to caveolae by molecular interactions with caveolin 1. Using a caveolin 1-deficient 3T3-L1 adipocyte cell line, we demonstrated that the localization of Kv1.3 in caveolar raft structures is important for proper insulin signaling. Insulin-dependent phosphorylation of the channel occurs at the onset of insulin-mediated signaling. However, when Kv1.3 was spatially outside of these lipid microdomains, impaired phosphorylation was exhibited. Our data shed light on the putative role of Kv1.3 in weight gain and insulin-dependent responses contributing to knowledge about adipocyte physiology.
Collapse
Affiliation(s)
- Mireia Pérez-Verdaguer
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
| | - Jesusa Capera
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
| | - María Ortego-Domínguez
- Dpto. de Fisiología Médica y Biofísica, Universidad de Sevilla, Av. Dr. Fedriani, s/n., 41009, Seville, Spain
| | - Joanna Bielanska
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
- Max-Planck-Institute of Experimental Medicine, Molecular Biology of Neuronal Signals, AG Oncophysiology, Hermann-Rein-Str. 3, 37075, Göttingen, Germany
| | - Núria Comes
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
| | - Rafael J Montoro
- Dpto. de Fisiología Médica y Biofísica, Universidad de Sevilla, Av. Dr. Fedriani, s/n., 41009, Seville, Spain
| | - Marta Camps
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain
| | - Antonio Felipe
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
| |
Collapse
|
109
|
Abstract
Pulmonary vascular diseases are associated with several factors including infection, cigarette smoking, abuse of dietary suppressants and drugs, prolonged exposure to high altitude, and other causes which in part induce significant oxidative stress resulting in endothelial cell injury, apoptosis, hyperproliferation, and vaso-occlusive disease. Maintenance of normal endothelial cell function is a critical role of endothelial nitric oxide synthase (eNOS) activity and physiologic nitric oxide (NO) signaling in the vascular wall. eNOS expression and activity is regulated by the membrane-associated scaffolding protein caveolin-1 (Cav-1), the main protein constituent of caveolae. This chapter summarizes the literature and highlights unanswered questions related to how inflammation-associated oxidative stress affects Cav-1 expression and regulatory functions, and how dysregulated eNOS enzymatic activity promotes endothelial dysfunction. Focus is given to how the conversion of eNOS from a NO-producing enzyme to a transient oxidant-generating system is associated twith Cav-1 depletion, endothelial cell injury, and pulmonary vascular diseases. Importantly, the vascular defects observed in absence of Cav-1 that give rise to injured or hyperproliferative endothelial cells and promote remodeled vasculature can be rescued by "re-coupling," inhibiting, or genetically deleting eNOS, supporting the notion that strict control of Cav-1 expression and eNOS activity and signaling is critical for maintaining pulmonary vascular homeostasis.
Collapse
Affiliation(s)
- Suellen D S Oliveira
- Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Richard D Minshall
- Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
110
|
Lee JJA, Maruyama R, Duddy W, Sakurai H, Yokota T. Identification of Novel Antisense-Mediated Exon Skipping Targets in DYSF for Therapeutic Treatment of Dysferlinopathy. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:596-604. [PMID: 30439648 PMCID: PMC6234522 DOI: 10.1016/j.omtn.2018.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/05/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022]
Abstract
Dysferlinopathy is a progressive myopathy caused by mutations in the dysferlin (DYSF) gene. Dysferlin protein plays a major role in plasma-membrane resealing. Some patients with DYSF deletion mutations exhibit mild symptoms, suggesting some regions of DYSF can be removed without significantly impacting protein function. Antisense-mediated exon-skipping therapy uses synthetic molecules called antisense oligonucleotides to modulate splicing, allowing exons harboring or near genetic mutations to be removed and the open reading frame corrected. Previous studies have focused on DYSF exon 32 skipping as a potential therapeutic approach, based on the association of a mild phenotype with the in-frame deletion of exon 32. To date, no other DYSF exon-skipping targets have been identified, and the relationship between DYSF exon deletion pattern and protein function remains largely uncharacterized. In this study, we utilized a membrane-wounding assay to evaluate the ability of plasmid constructs carrying mutant DYSF, as well as antisense oligonucleotides, to rescue membrane resealing in patient cells. We report that multi-exon skipping of DYSF exons 26–27 and 28–29 rescues plasma-membrane resealing. Successful translation of these findings into the development of clinical antisense drugs would establish new therapeutic approaches that would be applicable to ∼5%–7% (exons 26–27 skipping) and ∼8% (exons 28–29 skipping) of dysferlinopathy patients worldwide.
Collapse
Affiliation(s)
- Joshua J A Lee
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Rika Maruyama
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - William Duddy
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, United Kingdom
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada; The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Science Research Chair, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
111
|
Abstract
The plasma membrane of eukaryotic cells is not a simple sheet of lipids and proteins but is differentiated into subdomains with crucial functions. Caveolae, small pits in the plasma membrane, are the most abundant surface subdomains of many mammalian cells. The cellular functions of caveolae have long remained obscure, but a new molecular understanding of caveola formation has led to insights into their workings. Caveolae are formed by the coordinated action of a number of lipid-interacting proteins to produce a microdomain with a specific structure and lipid composition. Caveolae can bud from the plasma membrane to form an endocytic vesicle or can flatten into the membrane to help cells withstand mechanical stress. The role of caveolae as mechanoprotective and signal transduction elements is reviewed in the context of disease conditions associated with caveola dysfunction.
Collapse
Affiliation(s)
- Robert G. Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, Queensland 4060, Australia
| |
Collapse
|
112
|
Bosma EK, van Noorden CJF, Schlingemann RO, Klaassen I. The role of plasmalemma vesicle-associated protein in pathological breakdown of blood-brain and blood-retinal barriers: potential novel therapeutic target for cerebral edema and diabetic macular edema. Fluids Barriers CNS 2018; 15:24. [PMID: 30231925 PMCID: PMC6146740 DOI: 10.1186/s12987-018-0109-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/10/2018] [Indexed: 12/14/2022] Open
Abstract
Breakdown of the blood–brain barrier (BBB) or inner blood–retinal barrier (BRB), induced by pathologically elevated levels of vascular endothelial growth factor (VEGF) or other mediators, can lead to vasogenic edema and significant clinical problems such as neuronal morbidity and mortality, or vision loss. Restoration of the barrier function with corticosteroids in the brain, or by blocking VEGF in the eye are currently the predominant treatment options for brain edema and diabetic macular edema, respectively. However, corticosteroids have side effects, and VEGF has important neuroprotective, vascular protective and wound healing functions, implying that long-term anti-VEGF therapy may also induce adverse effects. We postulate that targeting downstream effector proteins of VEGF and other mediators that are directly involved in the regulation of BBB and BRB integrity provide more attractive and safer treatment options for vasogenic cerebral edema and diabetic macular edema. The endothelial cell-specific protein plasmalemma vesicle-associated protein (PLVAP), a protein associated with trans-endothelial transport, emerges as candidate for this approach. PLVAP is expressed in a subset of endothelial cells throughout the body where it forms the diaphragms of caveolae, fenestrae and trans-endothelial channels. However, PLVAP expression in brain and eye barrier endothelia only occurs in pathological conditions associated with a compromised barrier function such as cancer, ischemic stroke and diabetic retinopathy. Here, we discuss the current understanding of PLVAP as a structural component of endothelial cells and regulator of vascular permeability in health and central nervous system disease. Besides providing a perspective on PLVAP identification, structure and function, and the regulatory processes involved, we also explore its potential as a novel therapeutic target for vasogenic cerebral edema and retinal macular edema.
Collapse
Affiliation(s)
- Esmeralda K Bosma
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Cornelis J F van Noorden
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.,Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.,Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands. .,Ocular Angiogenesis Group, Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, Room L3-154, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
113
|
Hodeify R, Nandakumar M, Own M, Courjaret RJ, Graumann J, Hubrack SZ, Machaca K. The CCT chaperonin is a novel regulator of Ca 2+ signaling through modulation of Orai1 trafficking. SCIENCE ADVANCES 2018; 4:eaau1935. [PMID: 30263962 PMCID: PMC6157965 DOI: 10.1126/sciadv.aau1935] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/14/2018] [Indexed: 05/23/2023]
Abstract
Store-operated Ca2+ entry (SOCE) encodes a range of cellular responses downstream of Ca2+ influx through the SOCE channel Orai1. Orai1 recycles at the plasma membrane (PM), with ~40% of the total Orai1 pool residing at the PM at steady state. The mechanisms regulating Orai1 recycling remain poorly understood. We map the domains in Orai1 that are required for its trafficking to and recycling at the PM. We further identify, using biochemical and proteomic approaches, the CCT [chaperonin-containing TCP-1 (T-complex protein 1)] chaperonin complex as a novel regulator of Orai1 recycling by primarily regulating Orai1 endocytosis. We show that Orai1 interacts with CCT through its intracellular loop and that inhibition of CCT-Orai1 interaction increases Orai1 PM residence. This increased residence is functionally significant as it results in prolonged Ca2+ signaling, early formation of STIM1-Orai1 puncta, and more rapid activation of NFAT (nuclear factor of activated T cells) downstream of SOCE. Therefore, the CCT chaperonin is a novel regulator of Orai1 trafficking and, as such, a modulator of Ca2+ signaling and effector activation kinetics.
Collapse
Affiliation(s)
- Rawad Hodeify
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Manjula Nandakumar
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Maryam Own
- Medical Program, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Raphael J. Courjaret
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Johannes Graumann
- Department of Biochemistry, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Satanay Z. Hubrack
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| |
Collapse
|
114
|
Kim YJ, Hirabayashi Y. Caveolin-1 prevents palmitate-induced NF-κB signaling by inhibiting GPRC5B-phosphorylation. Biochem Biophys Res Commun 2018; 503:2673-2677. [DOI: 10.1016/j.bbrc.2018.08.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 01/04/2023]
|
115
|
Tanaka S, Fujio Y, Nakayama H. Caveolae-Specific CaMKII Signaling in the Regulation of Voltage-Dependent Calcium Channel and Cardiac Hypertrophy. Front Physiol 2018; 9:1081. [PMID: 30131723 PMCID: PMC6090180 DOI: 10.3389/fphys.2018.01081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/19/2018] [Indexed: 02/04/2023] Open
Abstract
Cardiac hypertrophy is a major risk for the progression of heart failure; however, the underlying molecular mechanisms contributing to this process remain elusive. The caveolae microdomain plays pivotal roles in various cellular processes such as lipid homeostasis, signal transduction, and endocytosis, and also serves as a signaling platform. Although the caveolae microdomain has been postulated to have a major contribution to the development of cardiac pathologies, including cardiac hypertrophy, recent evidence has placed this role into question. Lack of direct evidence and appropriate methods for determining activation of caveolae-specific signaling has thus far limited the ability to obtain a definite answer to the question. In this review, we focus on the potential physiological and pathological roles of the multifunctional kinase Ca2+/calmodulin-dependent kinase II and voltage-dependent L-type calcium channel in the caveolae, toward gaining a better understanding of the contribution of caveolae-based signaling in cardiac hypertrophy.
Collapse
Affiliation(s)
- Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
116
|
Wickert M, Hildick KL, Baillie GL, Jelinek R, Aparisi Rey A, Monory K, Schneider M, Ross RA, Henley JM, Lutz B. The F238L Point Mutation in the Cannabinoid Type 1 Receptor Enhances Basal Endocytosis via Lipid Rafts. Front Mol Neurosci 2018; 11:230. [PMID: 30026687 PMCID: PMC6041392 DOI: 10.3389/fnmol.2018.00230] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/12/2018] [Indexed: 11/22/2022] Open
Abstract
Defining functional domains and amino acid residues in G protein coupled receptors (GPCRs) represent an important way to improve rational drug design for this major class of drug targets. The cannabinoid type 1 (CB1) receptor is one of the most abundant GPCRs in the central nervous system and is involved in many physiological and pathophysiological processes. Interestingly, cannabinoid type 1 receptor with a phenylalanine 238 to leucine mutation (CB1F238L) has been already linked to a number of both in vitro and in vivo alterations. While CB1F238L causes significantly reduced presynaptic neurotransmitter release at the cellular level, behaviorally this mutation induces increased risk taking, social play behavior and reward sensitivity in rats. However, the molecular mechanisms underlying these changes are not fully understood. In this study, we tested whether the F238L mutation affects trafficking and axonal/presynaptic polarization of the CB1 receptor in vitro. Steady state or ligand modulated surface expression and lipid raft association was analyzed in human embryonic kidney 293 (HEK293) cells stably expressing either wild-type cannabinoid type 1 receptor (CB1wt) or CB1F238L receptor. Axonal/presynaptic polarization of the CB1F238L receptor was assessed in transfected primary hippocampal neurons. We show that in vitro the CB1F238L receptor displays increased association with lipid rafts, which coincides with increased lipid raft mediated constitutive endocytosis, leading to a reduction in steady state surface expression of the CB1F238L receptor. Furthermore, the CB1F238L receptor showed increased axonal polarization in primary hippocampal neurons. These data demonstrate that endocytosis of the CB1 receptor is an important mediator of axonal/presynaptic polarization and that phenylalanine 238 plays a key role in CB1 receptor trafficking and axonal polarization.
Collapse
Affiliation(s)
- Melanie Wickert
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Keri L Hildick
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Gemma L Baillie
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ruth Jelinek
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Alejandro Aparisi Rey
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Krisztina Monory
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Miriam Schneider
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Psychology, University of Heidelberg, Heidelberg, Germany
| | - Ruth A Ross
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jeremy M Henley
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.,German Resilience Center (DRZ), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
117
|
Abstract
Viroporins are short polypeptides encoded by viruses. These small membrane proteins assemble into oligomers that can permeabilize cellular lipid bilayers, disrupting the physiology of the host to the advantage of the virus. Consequently, efforts during the last few decades have been focused towards the discovery of viroporin channel inhibitors, but in general these have not been successful to produce licensed drugs. Viroporins are also involved in viral pathogenesis by engaging in critical interactions with viral proteins, or disrupting normal host cellular pathways through coordinated interactions with host proteins. These protein-protein interactions (PPIs) may become alternative attractive drug targets for the development of antivirals. In this sense, while thus far most antiviral molecules have targeted viral proteins, focus is moving towards targeting host proteins that are essential for virus replication. In principle, this largely would overcome the problem of resistance, with the possibility of using repositioned existing drugs. The precise role of these PPIs, their strain- and host- specificities, and the structural determination of the complexes involved, are areas that will keep the fields of virology and structural biology occupied for years to come. In the present review, we provide an update of the efforts in the characterization of the main PPIs for most viroporins, as well as the role of viroporins in these PPIs interactions.
Collapse
Affiliation(s)
| | - David Bhella
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| |
Collapse
|
118
|
Caveolin-1 Scaffolding Domain Peptides Alleviate Liver Fibrosis by Inhibiting TGF-β1/Smad Signaling in Mice. Int J Mol Sci 2018; 19:ijms19061729. [PMID: 29891777 PMCID: PMC6032240 DOI: 10.3390/ijms19061729] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/08/2018] [Accepted: 06/08/2018] [Indexed: 12/31/2022] Open
Abstract
Liver fibrosis is the common pathological process characterized by activation of hepatic stellate cells (HSCs) and overproduction of extracellular matrix (ECM). Caveolin-1 (Cav1), the principal component of caveolae, is regarded as an important inhibitor of multiple signaling molecules including transforming growth factor β1(TGF-β1) signaling. To evaluate the role of Cav1 in liver fibrosis, Cav1 deficient (Cav1−/−) and wild type (WT) mice were subjected to liver fibrosis induced by carbon tetrachloride (CCl4). Results indicated no significant difference between Cav1−/− and WT mice in inflammation or collagen content before CCl4 treatment. After CCl4 administration, Cav1−/− mice showed enhanced TGF-β1 signaling, as reflected by a significantly greater amount of phosphorylation of Smad2 and collagen deposition in livers over WT animals. Qualitative and quantitative analysis indicated that inflammatory injury to the liver was markedly aggravated, accompanied by increased degeneration and necrosis of hepatocytes, higher alanine aminotransferase (ALT)/aspartate aminotransferase (AST), TGF-α and IL-1β levels in Cav1−/− animals. The mRNA and protein levels of α-smooth muscle actin (α-SMA), Collagen α1(I), and Collagen α1(III) were further enhanced in Cav1−/− animals. We also observed a significant decrease in collagen content in Cav1−/− and WT animals administrated with Cav1 scaffolding domain peptides (CSD). In vitro study indicated that phosphorylation of Smad2 was inhibited after CSD treatment, accompanied by decreased protein levels of α-SMA, Collagen α1(I), and Collagen α1(III) in HSCs. We conclude that Cav1 is an important inhibitor of TGF-β1/Smad signaling in HSCs activation and collagen production, which might make it a promising target for therapy of liver fibrosis.
Collapse
|
119
|
Abstract
Researchers reconstitute caveolae assembly in cell-free extracts to investigate how these membrane microdomains interact with signaling proteins.
Collapse
|
120
|
Jung W, Sierecki E, Bastiani M, O'Carroll A, Alexandrov K, Rae J, Johnston W, Hunter DJB, Ferguson C, Gambin Y, Ariotti N, Parton RG. Cell-free formation and interactome analysis of caveolae. J Cell Biol 2018; 217:2141-2165. [PMID: 29716956 PMCID: PMC5987714 DOI: 10.1083/jcb.201707004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/24/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023] Open
Abstract
Caveolae are linked to signaling protein regulation through interactions with caveolins. We describe a cell-free system for the biogenesis of caveolae and show phosphorylated-caveolins preferentially bind signaling proteins. Our validation in vivo shows that phosphorylated CAV1 recruits TRAF2 to the endosome to form a signaling platform. Caveolae have been linked to the regulation of signaling pathways in eukaryotic cells through direct interactions with caveolins. Here, we describe a cell-free system based on Leishmania tarentolae (Lt) extracts for the biogenesis of caveolae and show its use for single-molecule interaction studies. Insertion of expressed caveolin-1 (CAV1) into Lt membranes was analogous to that of caveolin in native membranes. Electron tomography showed that caveolins generate domains of precise size and curvature. Cell-free caveolae were used in quantitative assays to test the interaction of membrane-inserted caveolin with signaling proteins and to determine the stoichiometry of interactions. Binding of membrane-inserted CAV1 to several proposed binding partners, including endothelial nitric-oxide synthase, was negligible, but a small number of proteins, including TRAF2, interacted with CAV1 in a phosphorylation-(CAV1Y14)–stimulated manner. In cells subjected to oxidative stress, phosphorylated CAV1 recruited TRAF2 to the early endosome forming a novel signaling platform. These findings lead to a novel model for cellular stress signaling by CAV1.
Collapse
Affiliation(s)
- WooRam Jung
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Emma Sierecki
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Michele Bastiani
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Ailis O'Carroll
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Kirill Alexandrov
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - James Rae
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Wayne Johnston
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Dominic J B Hunter
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Charles Ferguson
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Yann Gambin
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Nicholas Ariotti
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Robert G Parton
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia .,The University of Queensland, The Centre for Microscopy and Microanalysis, Brisbane, Queensland, Australia
| |
Collapse
|
121
|
Vladimirov VI, Zernii EY, Baksheeva VE, Wimberg H, Kazakov AS, Tikhomirova NK, Nemashkalova EL, Mitkevich VA, Zamyatnin AA, Lipkin VM, Philippov PP, Permyakov SE, Senin II, Koch KW, Zinchenko DV. Photoreceptor calcium sensor proteins in detergent-resistant membrane rafts are regulated via binding to caveolin-1. Cell Calcium 2018; 73:55-69. [PMID: 29684785 DOI: 10.1016/j.ceca.2018.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/07/2018] [Accepted: 04/10/2018] [Indexed: 01/25/2023]
Abstract
Rod cell membranes contain cholesterol-rich detergent-resistant membrane (DRM) rafts, which accumulate visual cascade proteins as well as proteins involved in regulation of phototransduction such as rhodopsin kinase and guanylate cyclases. Caveolin-1 is the major integral component of DRMs, possessing scaffolding and regulatory activities towards various signaling proteins. In this study, photoreceptor Ca2+-binding proteins recoverin, NCS1, GCAP1, and GCAP2, belonging to neuronal calcium sensor (NCS) family, were recognized as novel caveolin-1 interacting partners. All four NCS proteins co-fractionate with caveolin-1 in DRMs, isolated from illuminated bovine rod outer segments. According to pull-down assay, surface plasmon resonance spectroscopy and isothermal titration calorimetry data, they are capable of high-affinity binding to either N-terminal fragment of caveolin-1 (1-101), or its short scaffolding domain (81-101) via a novel structural site. In recoverin this site is localized in C-terminal domain in proximity to the third EF-hand motif and composed of aromatic amino acids conserved among NCS proteins. Remarkably, the binding of NCS proteins to caveolin-1 occurs only in the absence of calcium, which is in agreement with higher accessibility of the caveolin-1 binding site in their Ca2+-free forms. Consistently, the presence of caveolin-1 produces no effect on regulatory activity of Ca2+-saturated recoverin or NCS1 towards rhodopsin kinase, but upregulates GCAP2, which potentiates guanylate cyclase activity being in Ca2+-free conformation. In addition, the interaction with caveolin-1 decreases cooperativity and augments affinity of Ca2 + binding to recoverin apparently by facilitating exposure of its myristoyl group. We suggest that at low calcium NCS proteins are compartmentalized in photoreceptor rafts via binding to caveolin-1, which may enhance their activity or ensure their faster responses on Ca2+-signals thereby maintaining efficient phototransduction recovery and light adaptation.
Collapse
Affiliation(s)
- Vasiliy I Vladimirov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia
| | - Evgeni Yu Zernii
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia; Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| | - Viktoriia E Baksheeva
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Hanna Wimberg
- Department of Neurosciences, Biochemistry Group, University of Oldenburg, Oldenburg, 26111 Germany
| | - Alexey S Kazakov
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia
| | - Natalya K Tikhomirova
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Ekaterina L Nemashkalova
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Andrey A Zamyatnin
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia; Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Valery M Lipkin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia
| | - Pavel P Philippov
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Sergei E Permyakov
- Protein Research Group, Institute for Biological Instrumentation of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia
| | - Ivan I Senin
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992 Russia
| | - Karl-W Koch
- Department of Neurosciences, Biochemistry Group, University of Oldenburg, Oldenburg, 26111 Germany
| | - Dmitry V Zinchenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia
| |
Collapse
|
122
|
Bryant SM, Kong CHT, Watson JJ, Gadeberg HC, James AF, Cannell MB, Orchard CH. Caveolin 3-dependent loss of t-tubular I Ca during hypertrophy and heart failure in mice. Exp Physiol 2018; 103:652-665. [PMID: 29473235 PMCID: PMC6099270 DOI: 10.1113/ep086731] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/15/2018] [Indexed: 12/29/2022]
Abstract
NEW FINDINGS What is the central question of this study? Heart failure is associated with redistribution of L-type Ca2+ current (ICa ) away from the t-tubule membrane to the surface membrane of cardiac ventricular myocytes. However, the underlying mechanism and its dependence on severity of pathology (hypertrophy versus failure) are unclear. What is the main finding and its importance? Increasing severity of response to transverse aortic constriction, from hypertrophy to failure, was accompanied by graded loss of t-tubular ICa and loss of regulation of ICa by caveolin 3. Thus, the pathological loss of t-tubular ICa , which contributes to impaired excitation-contraction coupling and thereby cardiac function in vivo, appears to be attributable to loss of caveolin 3-dependent stimulation of t-tubular ICa . ABSTRACT Previous work has shown redistribution of L-type Ca2+ current (ICa ) from the t-tubules to the surface membrane of rat ventricular myocytes after myocardial infarction. However, whether this occurs in all species and in response to other insults, the relationship of this redistribution to the severity of the pathology, and the underlying mechanism, are unknown. We have therefore investigated the response of mouse hearts and myocytes to pressure overload induced by transverse aortic constriction (TAC). Male C57BL/6 mice underwent TAC or equivalent sham operation 8 weeks before use. ICa and Ca2+ transients were measured in isolated myocytes, and expression of caveolin 3 (Cav3), junctophilin 2 (Jph2) and bridging integrator 1 (Bin1) was determined. C3SD peptide was used to disrupt Cav3 binding to its protein partners. Some animals showed cardiac hypertrophy in response to TAC with little evidence of heart failure, whereas others showed greater hypertrophy and pulmonary congestion. These graded changes were accompanied by graded cellular hypertrophy, t-tubule disruption, decreased expression of Jph2 and Cav3, and decreased t-tubular ICa density, with no change at the cell surface, and graded impairment of Ca2+ release at t-tubules. C3SD decreased ICa density in control but not in TAC myocytes. These data suggest that the graded changes in cardiac function and size that occur in response to TAC are paralleled by graded changes in cell structure and function, which will contribute to the impaired function observed in vivo. They also suggest that loss of t-tubular ICa is attributable to loss of Cav3-dependent stimulation of ICa .
Collapse
Affiliation(s)
- Simon M Bryant
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Cherrie H T Kong
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Judy J Watson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Hanne C Gadeberg
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Andrew F James
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Mark B Cannell
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Clive H Orchard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
123
|
Filippini A, Sica G, D'Alessio A. The caveolar membrane system in endothelium: From cell signaling to vascular pathology. J Cell Biochem 2018; 119:5060-5071. [PMID: 29637636 DOI: 10.1002/jcb.26793] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/16/2018] [Indexed: 12/12/2022]
Abstract
Caveolae are 50- to 100-nm cholesterol and glycosphingolipid-rich flask-shaped invaginations commonly observed in many terminally differentiated cells. These organelles have been described in many cell types and are particularly abundant in endothelial cells, where they have been involved in the regulation of certain signaling pathways. Specific scaffolding proteins termed caveolins, along with the more recently discovered members of the cavin family, represent the major protein components during caveolae biogenesis. In addition, multiple studies aimed to investigate the expression and the regulation of these proteins significantly contributed to elucidate the role of caveolae and caveolins in endothelial cell physiology and disease. The aim of this review is to survey recent evidence of the involvement of the caveolar network in endothelial cell biology and endothelial cell dysfunction.
Collapse
Affiliation(s)
- Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gigliola Sica
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessio D'Alessio
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
124
|
Sonnino S, Chiricozzi E, Grassi S, Mauri L, Prioni S, Prinetti A. Gangliosides in Membrane Organization. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:83-120. [PMID: 29747825 DOI: 10.1016/bs.pmbts.2017.12.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Since the structure of GM1 was elucidated 55years ago, researchers have been attracted by the sialylated glycans of gangliosides. Gangliosides head groups, protruding toward the extracellular space, significantly contribute to the cell glycocalyx; and in certain cells, such as neurons, are major determinants of the features of the cell surface. Expression of glycosyltransferases involved in the de novo biosynthesis of gangliosides is tightly regulated along cell differentiation and activation, and is regarded as the main metabolic mechanism responsible for the acquisition of cell-specific ganglioside patterns. The resulting sialooligosaccharides are characterized by a high degree of geometrical complexity and by highly dynamic properties, which seem to be functional for complex interactions with other molecules sitting on the same cellular membrane (cis-interactions) or soluble molecules present in the extracellular environment, or molecules associated with the surface of other cells (trans-interactions). There is no doubt that the multifaceted biological functions of gangliosides are largely dependent on oligosaccharide-mediated molecular interactions. However, gangliosides are amphipathic membrane lipids, and their chemicophysical, aggregational, and, consequently, biological properties are dictated by the properties of the monomers as a whole, which are not merely dependent on the structures of their polar head groups. In this chapter, we would like to focus on the peculiar chemicophysical features of gangliosides (in particular, those of the nervous system), that represent an important driving force determining the organization and properties of cellular membranes, and to emphasize the causal connections between altered ganglioside-dependent membrane organization and relevant pathological conditions.
Collapse
|
125
|
Chen Z, D S Oliveira S, Zimnicka AM, Jiang Y, Sharma T, Chen S, Lazarov O, Bonini MG, Haus JM, Minshall RD. Reciprocal regulation of eNOS and caveolin-1 functions in endothelial cells. Mol Biol Cell 2018; 29:1190-1202. [PMID: 29563255 PMCID: PMC5935069 DOI: 10.1091/mbc.e17-01-0049] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We hypothesized that the maintenance of vascular homeostasis is critically dependent on the expression and reciprocal regulation of caveolin-1 (Cav-1) and endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs). Skeletal muscle biopsies from subjects with type 2 diabetes showed 50% less Cav-1 and eNOS than those from lean healthy controls. The Cav-1:eNOS expression ratio was 200:1 in primary culture human ECs. Cav-1 small interfering RNA (siRNA) reduced eNOS protein and gene expression in association with a twofold increase in eNOS phosphorylation and nitrate production per molecule of eNOS, which was reversed in cells overexpressing Adv-Cav-1-GFP. Upon addition of the Ca2+ ionophore A23187 to activate eNOS, we observed eNOS Ser1177 phosphorylation, its translocation to β-catenin-positive cell–cell junctions, and increased colocalization of eNOS and Cav-1 within 5 min. We also observed Cav-1 S-nitrosylation and destabilization of Cav-1 oligomers in cells treated with A23187 as well as insulin or albumin, and this could be blocked by L-NAME, PP2, or eNOS siRNA. Finally, caveola-mediated endocytosis of albumin or insulin was reduced by Cav-1 or eNOS siRNA, and the effect of Cav-1 siRNA was rescued by Adv-Cav-1-GFP. Thus, Cav-1 stabilizes eNOS expression and regulates its activity, whereas eNOS-derived NO promotes caveola-mediated endocytosis.
Collapse
Affiliation(s)
- Zhenlong Chen
- Departments of aAnesthesiology, University of Illinois at Chicago, Chicago, IL 60612
| | - Suellen D S Oliveira
- Departments of aAnesthesiology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Ying Jiang
- Departments of aAnesthesiology, University of Illinois at Chicago, Chicago, IL 60612
| | - Tiffany Sharma
- Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Stone Chen
- Whitney M. Young Magnet High School, Chicago, IL 60607
| | - Orly Lazarov
- Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Jacob M Haus
- Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Richard D Minshall
- Departments of aAnesthesiology, University of Illinois at Chicago, Chicago, IL 60612.,Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| |
Collapse
|
126
|
Marin R, Diaz M. Estrogen Interactions With Lipid Rafts Related to Neuroprotection. Impact of Brain Ageing and Menopause. Front Neurosci 2018; 12:128. [PMID: 29559883 PMCID: PMC5845729 DOI: 10.3389/fnins.2018.00128] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/16/2018] [Indexed: 12/22/2022] Open
Abstract
Estrogens (E2) exert a plethora of neuroprotective actions against aged-associated brain diseases, including Alzheimer's disease (AD). Part of these actions takes place through binding to estrogen receptors (ER) embedded in signalosomes, where numerous signaling proteins are clustered. Signalosomes are preferentially located in lipid rafts which are dynamic membrane microstructures characterized by a peculiar lipid composition enriched in gangliosides, saturated fatty acids, cholesterol, and sphingolipids. Rapid E2 interactions with ER-related signalosomes appear to trigger intracellular signaling ultimately leading to the activation of molecular mechanisms against AD. We have previously observed that the reduction of E2 blood levels occurring during menopause induced disruption of ER-signalosomes at frontal cortical brain areas. These molecular changes may reduce neuronal protection activities, as similar ER signalosome derangements were observed in AD brains. The molecular impairments may be associated with changes in the lipid composition of lipid rafts observed in neurons during menopause and AD. These evidences indicate that the changes in lipid raft structure during aging may be at the basis of alterations in the activity of ER and other neuroprotective proteins integrated in these membrane microstructures. Moreover, E2 is a homeostatic modulator of lipid rafts. Recent work has pointed to this relevant aspect of E2 activity to preserve brain integrity, through mechanisms affecting lipid uptake and local biosynthesis in the brain. Some evidences have demonstrated that estrogens and the docosahexaenoic acid (DHA) exert synergistic effects to stabilize brain lipid matrix. DHA is essential to enhance molecular fluidity at the plasma membrane, promoting functional macromolecular interactions in signaling platforms. In support of this, DHA detriment in neuronal lipid rafts has been associated with the most common age-associated neuropathologies, namely AD and Parkinson disease. Altogether, these findings indicate that E2 may participate in brain preservation through a dual membrane-related mechanism. On the one hand, E2 interacting with ER related signalosomes may protect against neurotoxic insults. On the other hand, E2 may exert lipostatic actions to preserve lipid balance in neuronal membrane microdomains. The different aspects of the emerging multifunctional role of estrogens in membrane-related signalosomes will be discussed in this review.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Medicine, Faculty of Health Sciences, University of La Laguna, Tenerife, Spain.,Fisiología y Biofísica de la Membrana Celular en Patologías Neurodegenerativas y Tumorales, Consejo Superior de Investigaciones Cientificas, Unidad Asociada de Investigación, Universidad de La Laguna Tenerife, Tenerife, Spain
| | - Mario Diaz
- Fisiología y Biofísica de la Membrana Celular en Patologías Neurodegenerativas y Tumorales, Consejo Superior de Investigaciones Cientificas, Unidad Asociada de Investigación, Universidad de La Laguna Tenerife, Tenerife, Spain.,Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Edaphology and Geology, University of La Laguna, Tenerife, Spain
| |
Collapse
|
127
|
Correll RN, Makarewich CA, Zhang H, Zhang C, Sargent MA, York AJ, Berretta RM, Chen X, Houser SR, Molkentin JD. Caveolae-localized L-type Ca2+ channels do not contribute to function or hypertrophic signalling in the mouse heart. Cardiovasc Res 2018; 113:749-759. [PMID: 28402392 DOI: 10.1093/cvr/cvx046] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 03/07/2017] [Indexed: 12/17/2022] Open
Abstract
Aims L-type Ca2+ channels (LTCCs) in adult cardiomyocytes are localized to t-tubules where they initiate excitation-contraction coupling. Our recent work has shown that a subpopulation of LTCCs found at the surface sarcolemma in caveolae of adult feline cardiomyocytes can also generate a Ca2+ microdomain that activates nuclear factor of activated T-cells signaling and cardiac hypertrophy, although the relevance of this paradigm to hypertrophy regulation in vivo has not been examined. Methods and results Here we generated heart-specific transgenic mice with a putative caveolae-targeted LTCC activator protein that was ineffective in initiating or enhancing cardiac hypertrophy in vivo. We also generated transgenic mice with cardiac-specific overexpression of a putative caveolae-targeted inhibitor of LTCCs, and while this protein inhibited caveolae-localized LTCCs without effects on global Ca2+ handling, it similarly had no effect on cardiac hypertrophy in vivo. Cardiac hypertrophy was elicited by pressure overload for 2 or 12 weeks or with neurohumoral agonist infusion. Caveolae-specific LTCC activator or inhibitor transgenic mice showed no greater change in nuclear factor of activated T-cells activity after 2 weeks of pressure overload stimulation compared with control mice. Conclusion Our results indicate that LTCCs in the caveolae microdomain do not affect cardiac function and are not necessary for the regulation of hypertrophic signaling in the adult mouse heart.
Collapse
Affiliation(s)
- Robert N Correll
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Catherine A Makarewich
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Hongyu Zhang
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Chen Zhang
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Michelle A Sargent
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Allen J York
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Remus M Berretta
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Xiongwen Chen
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Steven R Houser
- Department of Physiology, Cardiovascular Research Center, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA.,Department of Pediatrics, Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH 45229-3039, USA
| |
Collapse
|
128
|
Howie J, Wypijewski KJ, Plain F, Tulloch LB, Fraser NJ, Fuller W. Greasing the wheels or a spanner in the works? Regulation of the cardiac sodium pump by palmitoylation. Crit Rev Biochem Mol Biol 2018; 53:175-191. [PMID: 29424237 DOI: 10.1080/10409238.2018.1432560] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The ubiquitous sodium/potassium ATPase (Na pump) is the most abundant primary active transporter at the cell surface of multiple cell types, including ventricular myocytes in the heart. The activity of the Na pump establishes transmembrane ion gradients that control numerous events at the cell surface, positioning it as a key regulator of the contractile and metabolic state of the myocardium. Defects in Na pump activity and regulation elevate intracellular Na in cardiac muscle, playing a causal role in the development of cardiac hypertrophy, diastolic dysfunction, arrhythmias and heart failure. Palmitoylation is the reversible conjugation of the fatty acid palmitate to specific protein cysteine residues; all subunits of the cardiac Na pump are palmitoylated. Palmitoylation of the pump's accessory subunit phospholemman (PLM) by the cell surface palmitoyl acyl transferase DHHC5 leads to pump inhibition, possibly by altering the relationship between the pump catalytic α subunit and specifically bound membrane lipids. In this review, we discuss the functional impact of PLM palmitoylation on the cardiac Na pump and the molecular basis of recognition of PLM by its palmitoylating enzyme DHHC5, as well as effects of palmitoylation on Na pump cell surface abundance in the cardiac muscle. We also highlight the numerous unanswered questions regarding the cellular control of this fundamentally important regulatory process.
Collapse
Affiliation(s)
- Jacqueline Howie
- a Institute of Cardiovascular and Medical Sciences , University of Glasgow , Glasgow , UK
| | | | - Fiona Plain
- b Molecular and Clinical Medicine , University of Dundee , Dundee , UK
| | - Lindsay B Tulloch
- b Molecular and Clinical Medicine , University of Dundee , Dundee , UK
| | - Niall J Fraser
- b Molecular and Clinical Medicine , University of Dundee , Dundee , UK
| | - William Fuller
- a Institute of Cardiovascular and Medical Sciences , University of Glasgow , Glasgow , UK
| |
Collapse
|
129
|
Overmiller AM, McGuinn KP, Roberts BJ, Cooper F, Brennan-Crispi DM, Deguchi T, Peltonen S, Wahl JK, Mahoney MG. c-Src/Cav1-dependent activation of the EGFR by Dsg2. Oncotarget 2018; 7:37536-37555. [PMID: 26918609 PMCID: PMC5122330 DOI: 10.18632/oncotarget.7675] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/25/2016] [Indexed: 12/31/2022] Open
Abstract
The desmosomal cadherin, desmoglein 2 (Dsg2), is deregulated in a variety of human cancers including those of the skin. When ectopically expressed in the epidermis of transgenic mice, Dsg2 activates multiple mitogenic signaling pathways and increases susceptibility to tumorigenesis. However, the molecular mechanism responsible for Dsg2-mediated cellular signaling is poorly understood. Here we show overexpression as well as co-localization of Dsg2 and EGFR in cutaneous SCCs in vivo. Using HaCaT keratinocytes, knockdown of Dsg2 decreases EGFR expression and abrogates the activation of EGFR, c-Src and Stat3, but not Erk1/2 or Akt, in response to EGF ligand stimulation. To determine whether Dsg2 mediates signaling through lipid microdomains, sucrose density fractionation illustrated that Dsg2 is recruited to and displaces Cav1, EGFR and c-Src from light density lipid raft fractions. STED imaging confirmed that the presence of Dsg2 disperses Cav1 from the cell-cell borders. Perturbation of lipid rafts with the cholesterol-chelating agent MβCD also shifts Cav1, c-Src and EGFR out of the rafts and activates signaling pathways. Functionally, overexpression of Dsg2 in human SCC A431 cells enhances EGFR activation and increases cell proliferation and migration through a c-Src and EGFR dependent manner. In summary, our data suggest that Dsg2 stimulates cell growth and migration by positively regulating EGFR level and signaling through a c-Src and Cav1-dependent mechanism using lipid rafts as signal modulatory platforms.
Collapse
Affiliation(s)
- Andrew M Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kathleen P McGuinn
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Brett J Roberts
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Felicia Cooper
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Donna M Brennan-Crispi
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Takahiro Deguchi
- Laboratory of Biophysics, Department of Cell Biology and Anatomy, University of Turku, Turku, Finland
| | - Sirkku Peltonen
- Department of Dermatology, University of Turku and Turku Hospital, Turku, Finland
| | - James K Wahl
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Mỹ G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
130
|
Norman R, Fuller W, Calaghan S. Caveolae and the cardiac myocyte. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
131
|
Bozic J, Markotic A, Cikes-Culic V, Novak A, Borovac JA, Vucemilovic H, Trgo G, Ticinovic Kurir T. Ganglioside GM3 content in skeletal muscles is increased in type 2 but decreased in type 1 diabetes rat models: Implications of glycosphingolipid metabolism in pathophysiology of diabetes. J Diabetes 2018; 10:130-139. [PMID: 28544772 DOI: 10.1111/1753-0407.12569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/10/2017] [Accepted: 04/13/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ganglioside GM3 is found in the plasma membrane, where its accumulation attenuates insulin receptor signaling. Considering the role of skeletal muscles in insulin-stimulated glucose uptake, the aim of the present study was to determine the expression of GM3 and its precursors in skeletal muscles of rat models of type 1 and type 2 diabetes mellitus (T1DM and T2DM, respectively). METHODS Diabetes was induced in male Sprague-Dawley rats by streptozotocin injection (55 mg/kg, i.p., for T1DM induction; 35 mg/kg, i.p., for T2DM induction), followed by feeding of rats with either a normal pellet diet (T1DM) or a high-fat diet (T2DM). Rats were killed 2 weeks after diabetes induction and samples of skeletal muscle were collected. Frozen quadriceps muscle sections were stained with a primary antibody against GM3 (Neu5Ac) and visualized using a secondary antibody coupled with Texas Red. The muscle content of ganglioside GM3 and its precursors was analyzed by high-performance thin-layer chromatography (HPTLC) followed by GM3 immunostaining. RESULTS Muscle GM3 content was significantly higher in T2DM compared with control rats (P < 0.001). Furthermore, levels of the GM3 precursors ceramide, glucosylceramide, and lactosylceramide were significantly higher in T2DM compared with control rats (P < 0.05), whereas ceramide content was significantly lower in T1DM rats (P < 0.05). The intensity of the GM3 band on HPTLC was significantly higher in T2DM rats (P < 0.001) and significantly lower in T1DM rats (P < 0.05) compared with control. CONCLUSIONS The expression patterns of GM3 ganglioside and its precursors in diabetic rats suggest that the role of glycosphingolipid metabolism may differ between T2DM and T1DM.
Collapse
Affiliation(s)
- Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, Split, Croatia
| | - Anita Markotic
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, Split, Croatia
| | - Vedrana Cikes-Culic
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, Split, Croatia
| | - Anela Novak
- Department of Internal Medicine, University Hospital Split, Split, Croatia
| | - Josip A Borovac
- Department of Pathophysiology, University of Split School of Medicine, Split, Croatia
| | - Hrvoje Vucemilovic
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Split, Split, Croatia
| | - Gorana Trgo
- Department of Internal Medicine, University Hospital Split, Split, Croatia
| | - Tina Ticinovic Kurir
- Department of Pathophysiology, University of Split School of Medicine, Split, Croatia
- Department of Internal Medicine, University Hospital Split, Split, Croatia
| |
Collapse
|
132
|
Impairment of Fas-ligand-caveolin-1 interaction inhibits Fas-ligand translocation to rafts and Fas-ligand-induced cell death. Cell Death Dis 2018; 9:73. [PMID: 29358576 PMCID: PMC5833370 DOI: 10.1038/s41419-017-0109-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/10/2017] [Accepted: 10/24/2017] [Indexed: 11/17/2022]
Abstract
Fas-ligand/CD178 belongs to the TNF family proteins and can induce apoptosis through death receptor Fas/CD95. The important requirement for Fas-ligand-dependent cell death induction is its localization to rafts, cholesterol- and sphingolipid-enriched micro-domains of membrane, involved in regulation of different signaling complexes. Here, we demonstrate that Fas-ligand physically associates with caveolin-1, the main protein component of rafts. Experiments with cells overexpressing Fas-ligand revealed a FasL N-terminal pre-prolin-rich region, which is essential for the association with caveolin-1. We found that the N-terminal domain of Fas-ligand bears two caveolin-binding sites. The first caveolin-binding site binds the N-terminal domain of caveolin-1, whereas the second one appears to interact with the C-terminal domain of caveolin-1. The deletion of both caveolin-binding sites in Fas-ligand impairs its distribution between cellular membranes, and attenuates a Fas-ligand-induced cytotoxicity. These results demonstrate that the interaction of Fas-ligand and caveolin-1 represents a molecular basis for Fas-ligand translocation to rafts, and the subsequent induction of Fas-ligand-dependent cell death. A possibility of a similar association between other TNF family members and caveolin-1 is discussed.
Collapse
|
133
|
|
134
|
Volonte D, Vyas AR, Chen C, Dacic S, Stabile LP, Kurland BF, Abberbock SR, Burns TF, Herman JG, Di YP, Galbiati F. Caveolin-1 promotes the tumor suppressor properties of oncogene-induced cellular senescence. J Biol Chem 2017; 293:1794-1809. [PMID: 29247004 DOI: 10.1074/jbc.m117.815902] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/04/2017] [Indexed: 11/06/2022] Open
Abstract
Oncogene-induced senescence (OIS) is considered a powerful tumor suppressor mechanism. Caveolin-1 acts as a scaffolding protein to functionally regulate signaling molecules. We demonstrate that a lack of caveolin-1 expression inhibits oncogenic K-Ras (K-RasG12V)-induced premature senescence in mouse embryonic fibroblasts and normal human bronchial epithelial cells. Oncogenic K-Ras induces senescence by limiting the detoxification function of MTH1. We found that K-RasG12V promotes the interaction of caveolin-1 with MTH1, which results in inhibition of MTH1 activity. Lung cancer cells expressing oncogenic K-Ras have bypassed the senescence barrier. Interestingly, overexpression of caveolin-1 restores cellular senescence in both A549 and H460 lung cancer cells and inhibits their transformed phenotype. In support of these findings, our in vivo data demonstrate that overexpression of oncogenic K-Ras (K-RasG12D) induces cellular senescence in the lung of wildtype but not caveolin-1-null mice. A lack of K-RasG12D-induced premature senescence in caveolin-1-null mice results in the formation of more abundant lung tumors. Consistent with these data, caveolin-1-null mice overexpressing K-RasG12D display accelerated mortality. Finally, our animal data were supported by human sample analysis in which we show that caveolin-1 expression is dramatically down-regulated in lung adenocarcinomas from lung cancer patients, both at the mRNA and protein levels, and that low caveolin-1 expression is associated with poor survival. Together, our data suggest that lung cancer cells escape oncogene-induced premature senescence through down-regulation of caveolin-1 expression to progress from premalignant lesions to cancer.
Collapse
Affiliation(s)
| | - Avani R Vyas
- From the Department of Pharmacology and Chemical Biology
| | - Chen Chen
- the Department of Environmental and Occupational Health, and
| | - Sanja Dacic
- the Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Laura P Stabile
- From the Department of Pharmacology and Chemical Biology.,the Lung Cancer Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Brenda F Kurland
- the Lung Cancer Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania 15232.,the Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania 15261, and
| | - Shira R Abberbock
- the Lung Cancer Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Timothy F Burns
- the Lung Cancer Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - James G Herman
- the Lung Cancer Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Yuanpu Peter Di
- the Department of Environmental and Occupational Health, and
| | | |
Collapse
|
135
|
de Almeida CJG. Caveolin-1 and Caveolin-2 Can Be Antagonistic Partners in Inflammation and Beyond. Front Immunol 2017; 8:1530. [PMID: 29250058 PMCID: PMC5715436 DOI: 10.3389/fimmu.2017.01530] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/27/2017] [Indexed: 12/26/2022] Open
Abstract
Caveolins, encoded by the CAV gene family, are the main protein components of caveolae. In most tissues, caveolin-1 (Cav-1) and caveolin-2 (Cav-2) are co-expressed, and Cav-2 targeting to caveolae depends on the formation of heterooligomers with Cav-1. Notwithstanding, Cav-2 has unpredictable activities, opposing Cav-1 in the regulation of some cellular processes. While the major roles of Cav-1 as a modulator of cell signaling in inflammatory processes and in immune responses have been extensively discussed elsewhere, the aim of this review is to focus on data revealing the distinct activity of Cav-1 and Cav-2, which suggest that these proteins act antagonistically to fine-tune a variety of cellular processes relevant to inflammation.
Collapse
|
136
|
Volonte D, Liu Z, Shiva S, Galbiati F. Caveolin-1 controls mitochondrial function through regulation of m-AAA mitochondrial protease. Aging (Albany NY) 2017; 8:2355-2369. [PMID: 27705926 PMCID: PMC5115893 DOI: 10.18632/aging.101051] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/19/2016] [Indexed: 11/25/2022]
Abstract
Mitochondrial proteases ensure mitochondrial integrity and function after oxidative stress by providing mitochondrial protein quality control. However, the molecular mechanisms that regulate this basic biological function in eukaryotic cells remain largely unknown. Caveolin-1 is a scaffolding protein involved in signal transduction. We find that AFG3L2, a m-AAA type of mitochondrial protease, is a novel caveolin-1-interacting protein in vitro. We show that oxidative stress promotes the translocation of both caveolin-1 and AFG3L2 to mitochondria, enhances the interaction of caveolin-1 with AFG3L2 in mitochondria and stimulates mitochondrial protease activity in wild-type fibroblasts. Localization of AFG3L2 to mitochondria after oxidative stress is inhibited in fibroblasts lacking caveolin-1, which results in impaired mitochondrial protein quality control, an oxidative phosphorylation to aerobic glycolysis switch and reduced ATP production. Mechanistically, we demonstrate that a lack of caveolin-1 does not alter either mitochondrial number or morphology but leads to the cytoplasmic and proteasome-dependent degradation of complexes I, III, IV and V upon oxidant stimulation. Restoration of mitochondrial respiratory chain complexes in caveolin-1 null fibroblasts reverts the enhanced glycolysis observed in these cells. Expression of a mutant form of AFG3L2, which has reduced affinity for caveolin-1, fails to localize to mitochondria and promotes degradation of complex IV after oxidative stress. Thus, caveolin-1 maintains mitochondrial integrity and function when cells are challenged with free radicals by promoting the mitochondrial localization of m-AAA protease and its quality control functions.
Collapse
Affiliation(s)
- Daniela Volonte
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Zhongmin Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Vascular Medicine Institute and Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ferruccio Galbiati
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
137
|
Bryant SM, Kong CHT, Cannell MB, Orchard CH, James AF. Loss of caveolin-3-dependent regulation of I Ca in rat ventricular myocytes in heart failure. Am J Physiol Heart Circ Physiol 2017; 314:H521-H529. [PMID: 29101175 PMCID: PMC5899261 DOI: 10.1152/ajpheart.00458.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
β2-Adrenoceptors and L-type Ca2+ current (ICa) redistribute from the t-tubules to the surface membrane of ventricular myocytes from failing hearts. The present study investigated the role of changes in caveolin-3 and PKA signaling, both of which have previously been implicated in this redistribution. ICa was recorded using the whole cell patch-clamp technique from ventricular myocytes isolated from the hearts of rats that had undergone either coronary artery ligation (CAL) or equivalent sham operation 18 wk earlier. ICa distribution between the surface and t-tubule membranes was determined using formamide-induced detubulation (DT). In sham myocytes, β2-adrenoceptor stimulation increased ICa in intact but not DT myocytes; however, forskolin (to increase cAMP directly) and H-89 (to inhibit PKA) increased and decreased, respectively, ICa at both the surface and t-tubule membranes. C3SD peptide (which decreases binding to caveolin-3) inhibited ICa in intact but not DT myocytes but had no effect in the presence of H-89. In contrast, in CAL myocytes, β2-adrenoceptor stimulation increased ICa in both intact and DT myocytes, but C3SD had no effect on ICa; forskolin and H-89 had similar effects as in sham myocytes. These data show the redistribution of β2-adrenoceptor activity and ICa in CAL myocytes and suggest constitutive stimulation of ICa by PKA in sham myocytes via concurrent caveolin-3-dependent (at the t-tubules) and caveolin-3-independent mechanisms, with the former being lost in CAL myocytes. NEW & NOTEWORTHY In ventricular myocytes from normal hearts, regulation of the L-type Ca2+ current by β2-adrenoceptors and the constitutive regulation by caveolin-3 is localized to the t-tubules. In heart failure, the regulation of L-type Ca2+ current by β2-adrenoceptors is redistributed to the surface membrane, and the constitutive regulation by caveolin-3 is lost.
Collapse
Affiliation(s)
- Simon M Bryant
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Cherrie H T Kong
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Mark B Cannell
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Clive H Orchard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Andrew F James
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| |
Collapse
|
138
|
Li X, Chen W, Li P, Wei J, Cheng Y, Liu P, Yan Q, Xu X, Cui Y, Gu Z, Simoncini T, Fu X. Follicular Stimulating Hormone Accelerates Atherogenesis by Increasing Endothelial VCAM-1 Expression. Theranostics 2017; 7:4671-4688. [PMID: 29187895 PMCID: PMC5706091 DOI: 10.7150/thno.21216] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 09/08/2017] [Indexed: 01/02/2023] Open
Abstract
Rationale: Postmenopausal atherosclerosis (AS) has for decades been attributed to estrogen deficiency. Although the follicular stimulating hormone (FSH) levels rise sharply in parallel, the direct effect of FSH on AS has never been investigated. In this study, we explored the possible role of FSH in the development of AS. Methods: This was a prospective cohort study of 48 healthy premenopausal and 15 postmenopausal women. ApoE knockout mice were used as atherosclerosis model and human umbilical vascular endothelial cells (HUVECs) were cultured as cell model. Serum hormones and vascular cell adhesion molecule-1 (VCAM-1) levels were measured. Real-time PCR, histology for atherosclerotic lesions, immunofluorescence, luciferase assay, transfection experiments, flow chamber adhesion assay and western blot were performed. Results: In ApoE knockout mice, administration of FSH increased the atherosclerotic lesions and serum VCAM-1 concentration. Importantly, in blood samples of postmenopausal women, we detected significantly higher levels of FSH and VCAM-1 compared with those from premenopausal women, and there was a positive correlation between these two molecules. In cultured HUVECs, FSH receptor (FSHR) mRNA and protein expression were detected and FSH enhanced VCAM-1 expression. This effect was mediated by the activation of nuclear factor κB (NF-κB), which was sequentially enhanced by the activation of PI3K/Akt/mTOR cascade. FSH first enhanced GαS activity resulting in elevated cAMP level and PKA activity, which relayed the signals from FSHR to the PI3K/Akt/mTOR cascade. Furthermore, FSHR was detected in endothelial caveolae fraction and interacted with caveolin-1 and GαS. The disruption of caveolae or the silencing of caveolin-1 blocked FSH effects on signaling activation and VCAM-1 expression, suggesting the existence of a functional signaling module in membrane caveolae. Finally, FSH increased human monocyte adhesion to HUVECs which was reversed by the VCAM-1 neutralizing antibody. Conclusion: FSHR was located in the membrane caveolae of HUVECs and FSH promoted VCAM-1 expression via FSHR/GαS /cAMP/PKA and PI3K/Akt/mTOR/NF-κB pathway. This may contribute to the deleterious role of FSH in the development of AS in postmenopausal women.
Collapse
|
139
|
TIE2 Associates with Caveolae and Regulates Caveolin-1 To Promote Their Nuclear Translocation. Mol Cell Biol 2017; 37:MCB.00142-17. [PMID: 28760776 DOI: 10.1128/mcb.00142-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/21/2017] [Indexed: 01/20/2023] Open
Abstract
DNA repair pathways are aberrant in cancer, enabling tumor cells to survive standard therapies-chemotherapy and radiotherapy. Our group previously reported that, upon irradiation, the membrane-bound tyrosine kinase receptor TIE2 translocates into the nucleus and phosphorylates histone H4 at Tyr51, recruiting ABL1 to the DNA repair complexes that participate in the nonhomologous end-joining pathway. However, no specific molecular mechanisms of TIE2 endocytosis have been reported. Here, we show that irradiation or ligand-induced TIE2 trafficking is dependent on caveolin-1, the main component of caveolae. Subcellular fractionation and confocal microscopy demonstrated TIE2/caveolin-1 complexes in the nucleus, and using inhibitor or small interfering RNAs (siRNAs) against caveolin-1 or Tie2 inhibited their trafficking. TIE2 was found in caveolae and directly phosphorylated caveolin-1 at Tyr14 in vitro and in vivo This modification regulated the generation of TIE2/caveolin-1 complexes and was essential for TIE2/caveolin-1 nuclear translocation. Our data further demonstrate that the combination of TIE2 and caveolin-1 inhibitors resulted in significant radiosensitization of malignant glioma cells, which will guide the development of combinatorial treatment with radiotherapy for patients with glioblastoma.
Collapse
|
140
|
Peng Y, Prater AR, Deutscher SL. Targeting aggressive prostate cancer-associated CD44v6 using phage display selected peptides. Oncotarget 2017; 8:86747-86768. [PMID: 29156833 PMCID: PMC5689723 DOI: 10.18632/oncotarget.21421] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022] Open
Abstract
There is a crucial need to identify new biomarkers associated with aggressive prostate cancer (PCa) including those associated with cancer stem cells (CSCs). CD44v6, generated by alternative splicing of CD44, has been proposed as a CSC biomarker due to its correlation with aggressive PCa disease. We hypothesized that phage display selected peptides that target CD44v6 may serve as theranostic agents for aggressive PCa. Here, a 15 amino acid peptide ("PFT") was identified by affinity selection against a peptide derived from the v6 region of CD44v6. Synthesized PFT exhibited specific binding to CD44v6 with an equilibrium dissociation constant (Kd) of 743.4 nM. PFT also bound CD44v6 highly expressed on human PCa cell lines. Further, an aggressive form of PCa cells (v6A3) was isolated and tagged by a novel CSC reporter vector. The v6A3 cells had a CSC-like phenotype including enriched CD44v6 expression, enhanced clonogenicity, resistance to chemotherapeutics, and generation of heterogeneous offspring. PFT exhibited preferential binding to v6A3 cells compared to parental cells. Immunohistofluorescence studies with human PCa tissue microarrays (TMA) indicated that PFT was highly accurate in detecting CD44v6-positive aggressive PCa cells, and staining positivity was significantly higher in late stage, metastatic and higher-grade samples. Taken together, this study provides for the first time phage display selected peptides that target CD44v6 overexpressed on PCa cells. Peptide PFT may be explored as an aid in the diagnosis and therapy of advanced PCa disease.
Collapse
Affiliation(s)
- Ying Peng
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Biochemistry, University of Missouri-Columbia, Columbia, MO, USA
| | - Austin R Prater
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Biochemistry, University of Missouri-Columbia, Columbia, MO, USA
| | - Susan L Deutscher
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Biochemistry, University of Missouri-Columbia, Columbia, MO, USA
| |
Collapse
|
141
|
Rajamanickam GD, Kastelic JP, Thundathil JC. Na/K-ATPase regulates bovine sperm capacitation through raft- and non-raft-mediated signaling mechanisms. Mol Reprod Dev 2017; 84:1168-1182. [PMID: 28833817 DOI: 10.1002/mrd.22879] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 08/09/2017] [Indexed: 11/06/2022]
Abstract
Highly dynamic lipid microdomains (rafts) in the sperm plasma membrane contain several signaling proteins that regulate sperm capacitation. Na/K-ATPase isoforms (testis-specific isoform ATP1A4 and ubiquitous isoform ATP1A1) are abundant in bovine sperm plasma membrane. We previously reported that incubation of bovine sperm with ouabain, a specific Na/K-ATPase ligand, induced tyrosine phosphorylation of several sperm proteins during capacitation. The objective of this study was to investigate the roles of lipid rafts and non-rafts in Na/K-ATPase enzyme activity and signaling during bovine sperm capacitation. Content of ATP1A4 and, to a lesser extent, ATP1A1 was increased in raft and non-raft fractions of capacitated sperm, although non-raft enzyme activities of both isoforms were higher than the corresponding activities in rafts from capacitated sperm. Yet, ATP1A4 was the predominant isoform responsible for total Na/K-ATPase activity in both rafts and non-rafts. A comparative increase in phosphorylation of signaling molecules was observed in both raft (CAV1) and non-raft (EGFR and ERK1/2) membrane fractions during capacitation. Although SRC was phosphorylated in both membrane fractions, the non-raft fraction possessed more of this activated form. We also inferred, by immunoprecipitation, that ATP1A4 interacted with CAV1 and EGFR in the raft fraction, whereas interactions of ATP1A4 with SRC, EGFR, and ERK1/2 occurred in the non-raft fraction of ouabain-capacitated sperm; conversely, ATP1A1 interacted only with CAV1 in both fractions of uncapacitated and capacitated sperm. In conclusion, both raft and non-raft cohorts of Na/K-ATPase isoforms contributed to phosphorylation of signaling molecules during bovine sperm capacitation.
Collapse
Affiliation(s)
- Gayathri D Rajamanickam
- Faculty of Veterinary Medicine, Department of Production Animal Health, University of Calgary, Calgary, Alberta, Canada
| | - John P Kastelic
- Faculty of Veterinary Medicine, Department of Production Animal Health, University of Calgary, Calgary, Alberta, Canada
| | - Jacob C Thundathil
- Faculty of Veterinary Medicine, Department of Production Animal Health, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
142
|
Meng F, Saxena S, Liu Y, Joshi B, Wong TH, Shankar J, Foster LJ, Bernatchez P, Nabi IR. The phospho-caveolin-1 scaffolding domain dampens force fluctuations in focal adhesions and promotes cancer cell migration. Mol Biol Cell 2017; 28:2190-2201. [PMID: 28592633 PMCID: PMC5531735 DOI: 10.1091/mbc.e17-05-0278] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/02/2017] [Indexed: 12/15/2022] Open
Abstract
Caveolin-1 (Cav1), a major Src kinase substrate phosphorylated on tyrosine-14 (Y14), contains the highly conserved membrane-proximal caveolin scaffolding domain (CSD; amino acids 82-101). Here we show, using CSD mutants (F92A/V94A) and membrane-permeable CSD-competing peptides, that Src kinase-dependent pY14Cav1 regulation of focal adhesion protein stabilization, focal adhesion tension, and cancer cell migration is CSD dependent. Quantitative proteomic analysis of Cav1-GST (amino acids 1-101) pull downs showed sixfold-increased binding of vinculin and, to a lesser extent, α-actinin, talin, and filamin, to phosphomimetic Cav1Y14D relative to nonphosphorylatable Cav1Y14F. Consistently, pY14Cav1 enhanced CSD-dependent vinculin tension in focal adhesions, dampening force fluctuation and synchronously stabilizing cellular focal adhesions in a high-tension mode, paralleling effects of actin stabilization. This identifies pY14Cav1 as a molecular regulator of focal adhesion tension and suggests that functional interaction between Cav1 Y14 phosphorylation and the CSD promotes focal adhesion traction and, thereby, cancer cell motility.
Collapse
Affiliation(s)
- Fanrui Meng
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sandeep Saxena
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Youtao Liu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Bharat Joshi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Timothy H Wong
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jay Shankar
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology and Michael Smith Labs, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Pascal Bernatchez
- James Hogg Research Centre, Institute for Heart + Lung Health, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ivan R Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
143
|
Survive or thrive: tradeoff strategy for cellular senescence. Exp Mol Med 2017; 49:e342. [PMID: 28572574 PMCID: PMC5519021 DOI: 10.1038/emm.2017.94] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/06/2017] [Accepted: 02/16/2017] [Indexed: 12/12/2022] Open
Abstract
Aging-dependent cellular behaviors toward extrinsic stress are characterized by the confined localization of certain molecules to either nuclear or perinuclear regions. Although most growth factors can activate downstream signaling in aging cells, they do not in fact have any impact on the cells because the signals cannot reach their genetic targets in the nucleus. For the same reason, varying apoptotic stress factors cannot stimulate the apoptotic pathway in senescent cells. Thus, the operation of a functional nuclear barrier in an aging-dependent manner has been investigated. To elucidate the mechanism for this process, the housekeeping transcription factor Sp1 was identified as a general regulator of nucleocytoplasmic trafficking (NCT) genes, including various nucleoporins, importins, exportins and Ran GTPase cycle-related genes. Interestingly, the posttranslational modification of Sp1 is readily influenced by extrinsic stress, including oxidative and metabolic stress. The decrease in SP1 O-GlcNAcylation under oxidative stress or during replicative senescence makes it susceptible to proteosomal degradation, resulting in defective NCT functions and leading to nuclear barrier formation. The operation of the nuclear barrier in aging provides a fundamental mechanism for cellular protection against stress and promotes survival at the expense of growth via stress-sensitive transcriptional control.
Collapse
|
144
|
Caveolin-1: An Oxidative Stress-Related Target for Cancer Prevention. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7454031. [PMID: 28546853 PMCID: PMC5436035 DOI: 10.1155/2017/7454031] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/23/2017] [Accepted: 03/07/2017] [Indexed: 01/19/2023]
Abstract
Aberrant oxidative metabolism is one of the hallmarks of cancer. Reactive species overproduction could promote carcinogenesis via inducing genetic mutations and activating oncogenic pathways, and thus, antioxidant therapy was considered as an important strategy for cancer prevention and treatment. Caveolin-1 (Cav-1), a constituent protein of caveolae, has been shown to mediate tumorigenesis and progression through oxidative stress modulation recently. Reactive species could modulate the expression, degradation, posttranslational modifications, and membrane trafficking of Cav-1, while Cav-1-targeted treatments could scavenge the reactive species. More importantly, emerging evidences have indicated that multiple antioxidants could exert antitumor activities in cancer cells and protective activities in normal cells by modulating the Cav-1 pathway. Altogether, these findings indicate that Cav-1 may be a promising oxidative stress-related target for cancer antioxidant prevention. Elucidating the underlying interaction mechanisms between oxidative stress and Cav-1 is helpful for enhancing the preventive effects of antioxidants on cancer, for improving clinical outcomes of antioxidant-related therapeutics in cancer patients, and for developing Cav-1 targeted drugs. Herein, we summarize the available evidence of the roles of Cav-1 and oxidative stress in tumorigenesis and development and shed novel light on designing strategies for cancer prevention or treatment by utilizing the interaction mode between Cav-1 and oxidative stress.
Collapse
|
145
|
Pleasant-Jenkins D, Reese C, Chinnakkannu P, Kasiganesan H, Tourkina E, Hoffman S, Kuppuswamy D. Reversal of maladaptive fibrosis and compromised ventricular function in the pressure overloaded heart by a caveolin-1 surrogate peptide. J Transl Med 2017; 97:370-382. [PMID: 28112757 PMCID: PMC5909408 DOI: 10.1038/labinvest.2016.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic ventricular pressure overload (PO) results in congestive heart failure (CHF) in which myocardial fibrosis develops in concert with ventricular dysfunction. Caveolin-1 is important in fibrosis in various tissues due to its decreased expression in fibroblasts and monocytes. The profibrotic effects of low caveolin-1 can be blocked with the caveolin-1 scaffolding domain peptide (CSD, a caveolin-1 surrogate) using both mouse models and human cells. We have studied the beneficial effects of CSD on mice in which PO was induced by trans-aortic constriction (TAC). Beneficial effects observed in TAC mice receiving CSD injections daily included: improved ventricular function (increased ejection fraction, stroke volume, and cardiac output; reduced wall thickness); decreased collagen I, collagen chaperone HSP47, fibronectin, and CTGF levels; decreased activation of non-receptor tyrosine kinases Pyk2 and Src; and decreased activation of eNOS. To determine the source of cells that contribute to fibrosis in CHF, flow cytometric studies were performed that suggested that myofibroblasts in the heart are in large part bone marrow-derived. Two CD45+ cell populations were observed. One (Zone 1) contained CD45+/HSP47-/macrophage marker+ cells (macrophages). The second (Zone 2) contained CD45moderate/HSP47+/macrophage marker- cells often defined as fibrocytes. TAC increased the number of cells in Zones 1 and 2 and the level of HSP47 in Zone 2. These studies are a first step in elucidating the mechanism of action of CSD in heart fibrosis and promoting the development of CSD as a novel treatment to reduce fibrosis and improve ventricular function in CHF patients.
Collapse
Affiliation(s)
- Dorea Pleasant-Jenkins
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Charles Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Harinath Kasiganesan
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| |
Collapse
|
146
|
Fu P, Chen F, Pan Q, Zhao X, Zhao C, Cho WCS, Chen H. The different functions and clinical significances of caveolin-1 in human adenocarcinoma and squamous cell carcinoma. Onco Targets Ther 2017; 10:819-835. [PMID: 28243118 PMCID: PMC5317307 DOI: 10.2147/ott.s123912] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Caveolin-1 (Cav-1), a major structural protein of caveolae, is an integral membrane protein which plays an important role in the progression of carcinoma. However, whether Cav-1 acts as a tumor promoter or a tumor suppressor still remains controversial. For example, the tumor-promoting function of Cav-1 has been found in renal cancer, prostate cancer, tongue squamous cell carcinoma (SCC), lung SCC and bladder SCC. In contrast, Cav-1 also plays an inhibitory role in esophagus adenocarcinoma, lung adenocarcinoma and cutaneous SCC. The role of Cav-1 is still controversial in thyroid cancer, hepatocellular carcinoma, gastric adenocarcinoma, colon adenocarcinoma, breast cancer, pancreas cancer, oral SCC, laryngeal SCC, head and neck SCC, esophageal SCC and cervical SCC. Besides, it has been reported that the loss of stromal Cav-1 might predict poor prognosis in breast cancer, gastric cancer, pancreas cancer, prostate cancer, oral SCC and esophageal SCC. However, the accumulation of stromal Cav-1 has been found to be promoted by the progression of tongue SCC. Taken together, Cav-1 seems playing a different role in different cancer subtypes even of the same organ, as well as acting differently in the same cancer subtype of different organs. Thus, we hereby explore the functions of Cav-1 in human adenocarcinoma and SCC from the perspective of clinical significances and pathogenesis. We envision that novel targets may come with the further investigation of Cav-1 in carcinogenesis.
Collapse
Affiliation(s)
- Pin Fu
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | - Fuchun Chen
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang
| | - Qi Pan
- Department of Thoracosurgery, Traditional Chinese Medical Hospital of Wenling, Wenling, Zhejiang
| | - Xianda Zhao
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | - Chen Zhao
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan
| | | | - Honglei Chen
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan; Department of Pathology, Maternal and Child Health Hospital of Hubei, Wuhan, People's Republic of China
| |
Collapse
|
147
|
Mierke J, Christoph M, Pfluecke C, Jellinghaus S, Wunderlich C, Strasser RH, Ibrahim K, Poitz DM. Atheroprotective role of Caveolin-1 and eNOS in an innovative transplantation model is mainly mediated by local effects. Biochim Biophys Acta Mol Basis Dis 2017; 1863:529-536. [DOI: 10.1016/j.bbadis.2016.11.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/03/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022]
|
148
|
Tonegawa K, Otsuka W, Kumagai S, Matsunami S, Hayamizu N, Tanaka S, Moriwaki K, Obana M, Maeda M, Asahi M, Kiyonari H, Fujio Y, Nakayama H. Caveolae-specific activation loop between CaMKII and L-type Ca 2+ channel aggravates cardiac hypertrophy in α 1-adrenergic stimulation. Am J Physiol Heart Circ Physiol 2016; 312:H501-H514. [PMID: 28039202 DOI: 10.1152/ajpheart.00601.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/01/2016] [Accepted: 11/15/2016] [Indexed: 11/22/2022]
Abstract
Activation of CaMKII induces a myriad of biological processes and plays dominant roles in cardiac hypertrophy. Caveolar microdomain contains many calcium/calmodulin-dependent kinase II (CaMKII) targets, including L-type Ca2+ channel (LTCC) complex, and serves as a signaling platform. The location of CaMKII activation is thought to be critical; however, the roles of CaMKII in caveolae are still elusive due to lack of methodology for the assessment of caveolae-specific activation. Our aim was to develop a novel tool for the specific analysis of CaMKII activation in caveolae and to determine the functional role of caveolar CaMKII in cardiac hypertrophy. To assess the caveolae-specific activation of CaMKII, we generated a fusion protein composed of phospholamban and caveolin-3 (cPLN-Cav3) and GFP fusion protein with caveolin-binding domain fused to CaMKII inhibitory peptide (CBD-GFP-AIP), which inhibits CaMKII activation specifically in caveolae. Caveolae-specific activation of CaMKII was detected using phosphospecific antibody for PLN (Thr17). Furthermore, adenoviral overexpression of LTCC β2a-subunit (β2a) in NRCMs showed its constitutive phosphorylation by CaMKII, which induces hypertrophy, and that both phosphorylation and hypertrophy are abolished by CBD-GFP-AIP expression, indicating that β2a phosphorylation occurs specifically in caveolae. Finally, β2a phosphorylation was observed after phenylephrine stimulation in β2a-overexpressing mice, and attenuation of cardiac hypertrophy after chronic phenylephrine stimulation was observed in nonphosphorylated mutant of β2a-overexpressing mice. We developed novel tools for the evaluation and inhibition of caveolae-specific activation of CaMKII. We demonstrated that phosphorylated β2a dominantly localizes to caveolae and induces cardiac hypertrophy after α1-adrenergic stimulation in mice.NEW & NOTEWORTHY While signaling in caveolae is thought to be important in cardiac hypertrophy, direct evidence is missing due to lack of tools to assess caveolae-specific signaling. This is the first study to demonstrate caveolae-specific activation of CaMKII signaling in cardiac hypertrophy induced by α1-adrenergic stimulation using an originally developed tool.
Collapse
Affiliation(s)
- Kota Tonegawa
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Wataru Otsuka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Shohei Kumagai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Sachi Matsunami
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Nao Hayamizu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Kazumasa Moriwaki
- Faculty of Medicine, Department of Pharmacology, Osaka Medical College, Takatsuki, Osaka, Japan; and
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Makiko Maeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Michio Asahi
- Faculty of Medicine, Department of Pharmacology, Osaka Medical College, Takatsuki, Osaka, Japan; and
| | - Hiroshi Kiyonari
- Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmacological Sciences, Osaka University, Suita, Osaka, Japan;
| |
Collapse
|
149
|
Brauers E, Roos A, Kollipara L, Zahedi RP, Beckmann A, Mohanadas N, Bauer H, Häusler M, Thoma S, Kress W, Senderek J, Weis J. The Caveolin-3 G56S sequence variant of unknown significance: Muscle biopsy findings and functional cell biological analysis. Proteomics Clin Appl 2016; 11. [PMID: 27739254 PMCID: PMC5248598 DOI: 10.1002/prca.201600007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 12/31/2022]
Abstract
Purpose In the era of next‐generation sequencing, we are increasingly confronted with sequence variants of unknown significance. This phenomenon is also known for variations in Caveolin‐3 and can complicate the molecular diagnosis of the disease. Here, we aimed to study the ambiguous character of the G56S Caveolin‐3 variant. Experimental design A comprehensive approach combining genetic and morphological studies of muscle derived from carriers of the G56S Caveolin‐3 variant were carried out and linked to biochemical assays (including phosphoblot studies and proteome profiling) and morphological investigations of cultured myoblasts. Results Muscles showed moderate chronic myopathic changes in all carriers of the variant. Myogenic RCMH cells expressing the G56S Caveolin‐3 protein presented irregular Caveolin‐3 deposits within the Golgi in addition to a regular localization of the protein to the plasma membrane. This result was associated with abnormal findings on the ultra‐structural level. Phosphoblot studies revealed that G56S affects EGFR‐signaling. Proteomic profiling demonstrated alterations in levels of physiologically relevant proteins which are indicative for antagonization of G56S Caveolin‐3 expression. Remarkably, some proteomic alterations were enhanced by osmotic/mechanical stress. Conclusions and clinical relevance Our studies suggest that G56S might influence the manifestation of myopathic changes upon the presence of additional cellular stress burden. Results of our studies moreover improve the current understanding of (genetic) causes of myopathic disorders classified as caveolinopathies.
Collapse
Affiliation(s)
- Eva Brauers
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andreas Roos
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS e.V, Dortmund, Germany
| | | | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften - ISAS e.V, Dortmund, Germany
| | - Alf Beckmann
- Medizinisches Versorgungszentrum Dr. Eberhard und Partner, Dortmund, Germany
| | - Nilane Mohanadas
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Hartmut Bauer
- Department of Neurology, Marien-Hospital, Euskirchen, Germany
| | - Martin Häusler
- Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Wolfram Kress
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Jan Senderek
- Friedrich-Baur-Institut, Neurologische Klinik und Poliklinik, Ludwig-Maximilians-University, München, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
150
|
Chakrabarti S, Chang A, Liu NJ, Gintzler AR. Chronic opioid treatment augments caveolin-1 scaffolding: relevance to stimulatory μ-opioid receptor adenylyl cyclase signaling. J Neurochem 2016; 139:737-747. [PMID: 27726130 DOI: 10.1111/jnc.13852] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/21/2016] [Accepted: 09/15/2016] [Indexed: 01/25/2023]
Abstract
Caveolin-1 is the predominant structural protein of caveolae, a subset of (lipid) membrane rafts that compartmentalize cell signaling. Caveolin-1 binds most to G protein-coupled receptors and their signaling partners, thereby enhancing interactions among signaling cascade components and the relative activation of specific G protein-coupled pathways. This study reveals that chronic opioid exposure of μ-opioid receptor (MOR) expressing Chinese hamster ovary cells (MOR-CHO) and chronic in vivo morphine exposure of rat spinal cord augmented recruitment of multiple components of MOR-adenylyl cyclase (AC) stimulatory signaling by caveolin-1. Strikingly, in MOR-CHO and spinal cord, blocking the caveolin-1 scaffolding domain substantially attenuated the chronic morphine-induced increased interaction of caveolin-1 with MOR, Gsα, protein phosphatase 2A (PP2A), and AC. Chronic morphine treatment also increased interactions among the above signaling proteins, thus enabling sufentanil to stimulate (rather than inhibit) cAMP production within lipid membrane microdomains. The latter finding underscores the functionality of augmented interactions among MOR, Gs α, PP2A, and AC. In the aggregate, our data strongly suggest that augmented caveolin-1 scaffolding undergirds the ability of chronic opioids to recruit an ancillary signaling pathway by acting as an organizing template for MOR-Gs α-AC signaling and delimiting the membrane compartment(s) in which it occurs. Since caveolin-1 binds to a wide spectrum of signaling molecules, altered caveolin-1 scaffolding following chronic opioid treatment is likely to pertain to most, if not all, MOR signaling partners. The chronic morphine-induced trigger that augments caveolin-1 scaffolding could represent a seminal perturbation that initiates the wide spectrum of adaptations thought to contribute to opioid tolerance and dependence.
Collapse
Affiliation(s)
- Sumita Chakrabarti
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Andrew Chang
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Nai-Jiang Liu
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Alan R Gintzler
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| |
Collapse
|