1
|
Huang G, Yang S, Long T, Gao Y, Lin G. Proteomic analysis of brain tissue from ducks with meningitis caused by Riemerella anatipestifer infection. Poult Sci 2024; 103:104059. [PMID: 39068696 PMCID: PMC11338091 DOI: 10.1016/j.psj.2024.104059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Riemerella anatipestifer is a Gram-negative, rod-shaped bacterium that is flagellated, non-budded, and encapsulated, measuring approximately 0.4 μm × 0.7 μm. After infecting ducklings with R. anatipestifer, the hosts exhibited pathological changes, such as bacterial meningitis, fibrinous pericarditis, and fibrinous peripheral hepatitis. The pathogenesis of meningitis caused by R. anatipestifer has not yet been elucidated. To investigate the key molecules or proteins involved in R. anatipestifer's penetration of the blood-brain barrier (BBB) and the subsequent development of duck meningitis, a duck meningitis model was established and characterized. Duckling brain tissues were collected and analyzed using 4D label-free proteomic technology. Differentially expressed proteins were analyzed using a series of bioinformatics methods and verified using RT-qPCR and Western-Blot. The results showed that the differentially expressed proteins were primarily related to intracellular transport, transport protein activity, and transmembrane transport protein activity, and were mainly enriched in pathways associated with reducing intercellular connections and adhesion and increasing cell migration and apoptosis. Thus, it is suggested that R. anatipestifer may penetrate the BBB via transcellular and paracellular pathways, causing neurological diseases such as meningitis. This study is the first to analyze R. anatipestifer-infected duckling brain tissue using proteomics, thus providing a direction for further research into the mechanisms of R. anatipestifer's penetration of the BBB.
Collapse
Affiliation(s)
- Guoliang Huang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Shengmei Yang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Ting Long
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Yuhan Gao
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Guozhen Lin
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China.
| |
Collapse
|
2
|
Kameni LE, Griffin M, Berry CE, Shariatzadeh S, Downer MA, Valencia C, Fazilat AZ, Nazerali R, Momeni A, Januszyk M, Longaker MT, Wan DC. Single-cell transcriptional analysis of irradiated skin reveals changes in fibroblast subpopulations and variability in caveolin expression. Radiat Oncol 2024; 19:82. [PMID: 38926892 PMCID: PMC11200992 DOI: 10.1186/s13014-024-02472-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Radiation-induced fibrosis (RIF) is an important late complication of radiation therapy, and the resulting damaging effects of RIF can significantly impact reconstructive outcomes. There is currently a paucity of effective treatment options available, likely due to the continuing knowledge gap surrounding the cellular mechanisms involved. In this study, detailed analyses of irradiated and non-irradiated human skin samples were performed incorporating histological and single-cell transcriptional analysis to identify novel features guiding development of skin fibrosis following radiation injury. METHODS Paired irradiated and contralateral non-irradiated skin samples were obtained from six female patients undergoing post-oncologic breast reconstruction. Skin samples underwent histological evaluation, immunohistochemistry, and biomechanical testing. Single-cell RNA sequencing was performed using the 10X single cell platform. Cells were separated into clusters using Seurat in R. The SingleR classifier was applied to ascribe cell type identities to each cluster. Differentially expressed genes characteristic to each cluster were then determined using non-parametric testing. RESULTS Comparing irradiated and non-irradiated skin, epidermal atrophy, dermal thickening, and evidence of thick, disorganized collagen deposition within the extracellular matrix of irradiated skin were readily appreciated on histology. These histologic features were associated with stiffness that was higher in irradiated skin. Single-cell RNA sequencing revealed six predominant cell types. Focusing on fibroblasts/stromal lineage cells, five distinct transcriptional clusters (Clusters 0-4) were identified. Interestingly, while all clusters were noted to express Cav1, Cluster 2 was the only one to also express Cav2. Immunohistochemistry demonstrated increased expression of Cav2 in irradiated skin, whereas Cav1 was more readily identified in non-irradiated skin, suggesting Cav1 and Cav2 may act antagonistically to modulate fibrotic cellular responses. CONCLUSION In response to radiation therapy, specific changes to fibroblast subpopulations and enhanced Cav2 expression may contribute to fibrosis. Altogether, this study introduces a novel pathway of caveolin involvement which may contribute to fibrotic development following radiation injury.
Collapse
Affiliation(s)
- Lionel E Kameni
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Charlotte E Berry
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Siavash Shariatzadeh
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mauricio A Downer
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Caleb Valencia
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander Z Fazilat
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rahim Nazerali
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Arash Momeni
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive, GK 102, Stanford, CA, 94305-5148, USA.
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, 257 Campus Drive, GK 102, Stanford, CA, 94305-5148, USA.
| |
Collapse
|
3
|
Juin SK, Ouseph R, Gondim DD, Jala VR, Sen U. Diabetic Nephropathy and Gaseous Modulators. Antioxidants (Basel) 2023; 12:antiox12051088. [PMID: 37237955 DOI: 10.3390/antiox12051088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic nephropathy (DN) remains the leading cause of vascular morbidity and mortality in diabetes patients. Despite the progress in understanding the diabetic disease process and advanced management of nephropathy, a number of patients still progress to end-stage renal disease (ESRD). The underlying mechanism still needs to be clarified. Gaseous signaling molecules, so-called gasotransmitters, such as nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), have been shown to play an essential role in the development, progression, and ramification of DN depending on their availability and physiological actions. Although the studies on gasotransmitter regulations of DN are still emerging, the evidence revealed an aberrant level of gasotransmitters in patients with diabetes. In studies, different gasotransmitter donors have been implicated in ameliorating diabetic renal dysfunction. In this perspective, we summarized an overview of the recent advances in the physiological relevance of the gaseous molecules and their multifaceted interaction with other potential factors, such as extracellular matrix (ECM), in the severity modulation of DN. Moreover, the perspective of the present review highlights the possible therapeutic interventions of gasotransmitters in ameliorating this dreaded disease.
Collapse
Affiliation(s)
- Subir Kumar Juin
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Microbiology & Immunology, Brown Cancer Center, Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rosemary Ouseph
- Division of Nephrology & Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dibson Dibe Gondim
- Department of Pathology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Venkatakrishna Rao Jala
- Department of Microbiology & Immunology, Brown Cancer Center, Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Kenworthy AK. The building blocks of caveolae revealed: caveolins finally take center stage. Biochem Soc Trans 2023; 51:855-869. [PMID: 37082988 PMCID: PMC10212548 DOI: 10.1042/bst20221298] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/22/2023]
Abstract
The ability of cells to divide, migrate, relay signals, sense mechanical stimuli, and respond to stress all rely on nanoscale invaginations of the plasma membrane known as caveolae. The caveolins, a family of monotopic membrane proteins, form the inner layer of the caveolar coat. Caveolins have long been implicated in the generation of membrane curvature, in addition to serving as scaffolds for signaling proteins. Until recently, however, the molecular architecture of caveolins was unknown, making it impossible to understand how they operate at a mechanistic level. Over the past year, two independent lines of evidence - experimental and computational - have now converged to provide the first-ever glimpse into the structure of the oligomeric caveolin complexes that function as the building blocks of caveolae. Here, we summarize how these discoveries are transforming our understanding of this long-enigmatic protein family and their role in caveolae assembly and function. We present new models inspired by the structure for how caveolins oligomerize, remodel membranes, interact with their binding partners, and reorganize when mutated. Finally, we discuss emerging insights into structural differences among caveolin family members that enable them to support the proper functions of diverse tissues and organisms.
Collapse
Affiliation(s)
- Anne K. Kenworthy
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, U.S.A
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| |
Collapse
|
5
|
Flourieusse A, Bourgeois P, Schenckbecher E, Palvair J, Legrand D, Labbé C, Bescond T, Avoscan L, Orlowski S, Rouleau A, Frelet-Barrand A. Formation of intracellular vesicles within the Gram+ Lactococcus lactis induced by the overexpression of Caveolin-1β. Microb Cell Fact 2022; 21:239. [PMCID: PMC9670397 DOI: 10.1186/s12934-022-01944-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/02/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract
Background
Caveolae are invaginated plasma membrane domains of 50–100 nm in diameter involved in many important physiological functions in eukaryotic cells. They are composed of different proteins, including the membrane-embedded caveolins and the peripheric cavins. Caveolin-1 has already been expressed in various expression systems (E. coli, insect cells, Toxoplasma gondii, cell-free system), generating intracellular caveolin-enriched vesicles in E. coli, insect cells and T. gondii. These systems helped to understand the protein insertion within the membrane and its oligomerization. There is still need for fundamental insights into the formation of specific domains on membrane, the deformation of a biological membrane driven by caveolin-1, the organization of a caveolar coat, and the requirement of specific lipids and proteins during the process. The aim of this study was to test whether the heterologously expressed caveolin-1β was able to induce the formation of intracellular vesicles within a Gram+ bacterium, Lactococcus lactis, since it displays a specific lipid composition different from E. coli and appears to emerge as a good alternative to E. coli for efficient overexpression of various membrane proteins.
Results
Recombinant bacteria transformed with the plasmid pNZ-HTC coding for the canine isoform of caveolin-1β were shown to produce caveolin-1β, in its functional oligomeric form, at a high expression level unexpected for an eukaryotic membrane protein. Electron microscopy revealed several intracellular vesicles from 30 to 60 nm, a size comparable to E. coli h-caveolae, beneath the plasma membrane of the overexpressing bacteria, showing that caveolin-1β is sufficient to induce membrane vesiculation. Immunolabelling studies showed antibodies on such neo-formed intracellular vesicles, but none on plasma membrane. Density gradient fractionation allowed the correlation between detection of oligomers on Western blot and appearance of vesicles measurable by DLS, showing the requirement of caveolin-1β oligomerization for vesicle formation.
Conclusions
Lactococcus lactis cells can heterologously overexpress caveolin-1β, generating caveolin-1β enriched intracellular neo-formed vesicles. These vesicles might be useful for potential co-expression of membrane proteins of pharmaceutical interest for their simplified functional characterization.
Collapse
|
6
|
Gorospe B, Moura JJG, Gutierrez-Merino C, Samhan-Arias AK. Design and Experimental Evaluation of a Peptide Antagonist against Amyloid β(1-42) Interactions with Calmodulin and Calbindin-D28k. Int J Mol Sci 2022; 23:2289. [PMID: 35216403 PMCID: PMC9736327 DOI: 10.3390/ijms232315203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022] Open
Abstract
Amyloid β1-42 (Aβ(1-42)) oligomers have been linked to the pathogenesis of Alzheimer's disease (AD). Intracellular calcium (Ca2+) homeostasis dysregulation with subsequent alterations of neuronal excitability has been proposed to mediate Aβ neurotoxicity in AD. The Ca2+ binding proteins calmodulin (CaM) and calbindin-D28k, whose expression levels are lowered in human AD brains, have relevant roles in neuronal survival and activity. In previous works, we have shown that CaM has a high affinity for Aβ(1-42) oligomers and extensively binds internalized Aβ(1-42) in neurons. In this work, we have designed a hydrophobic peptide of 10 amino acid residues: VFAFAMAFML (amidated-C-terminus amino acid) mimicking the interacting domain of CaM with Aβ (1-42), using a combined strategy based on the experimental results obtained for Aβ(1-42) binding to CaM and in silico docking analysis. The increase in the fluorescence intensity of Aβ(1-42) HiLyteTM-Fluor555 has been used to monitor the kinetics of complex formation with CaM and with calbindin-D28k. The complexation between nanomolar concentrations of Aβ(1-42) and calbindin-D28k is also a novel finding reported in this work. We found that the synthetic peptide VFAFAMAFML (amidated-C-terminus amino acid) is a potent inhibitor of the formation of Aβ(1-42):CaM and of Aβ(1-42):calbindin-D28k complexes.
Collapse
Affiliation(s)
- Berta Gorospe
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Lisbon, Portugal
| | - José J. G. Moura
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Lisbon, Portugal
| | - Carlos Gutierrez-Merino
- Research Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Alejandro K. Samhan-Arias
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Lisbon, Portugal
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| |
Collapse
|
7
|
Low JY, Laiho M. Caveolae-Associated Molecules, Tumor Stroma, and Cancer Drug Resistance: Current Findings and Future Perspectives. Cancers (Basel) 2022; 14:cancers14030589. [PMID: 35158857 PMCID: PMC8833326 DOI: 10.3390/cancers14030589] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell membranes contain small invaginations called caveolae. They are a specialized lipid domain and orchestrate cellular signaling events, mechanoprotection, and lipid homeostasis. Formation of the caveolae depends on two classes of proteins, the caveolins and cavins, which form large complexes that allow their self-assembly into caveolae. Loss of either of these two proteins leads to distortion of the caveolae structure and disruption of many physiological processes that affect diseases of the muscle, metabolic states governing lipids, and the glucose balance as well as cancers. In cancers, the expression of caveolins and cavins is heterogenous, and they undergo alterations both in the tumors and the surrounding tumor microenvironment stromal cells. Remarkably, their expression and function has been associated with resistance to many cancer drugs. Here, we summarize the current knowledge of the resistance mechanisms and how this knowledge could be applied into the clinic in future. Abstract The discovery of small, “cave-like” invaginations at the plasma membrane, called caveola, has opened up a new and exciting research area in health and diseases revolving around this cellular ultrastructure. Caveolae are rich in cholesterol and orchestrate cellular signaling events. Within caveola, the caveola-associated proteins, caveolins and cavins, are critical components for the formation of these lipid rafts, their dynamics, and cellular pathophysiology. Their alterations underlie human diseases such as lipodystrophy, muscular dystrophy, cardiovascular disease, and diabetes. The expression of caveolins and cavins is modulated in tumors and in tumor stroma, and their alterations are connected with cancer progression and treatment resistance. To date, although substantial breakthroughs in cancer drug development have been made, drug resistance remains a problem leading to treatment failures and challenging translation and bench-to-bedside research. Here, we summarize the current progress in understanding cancer drug resistance in the context of caveola-associated molecules and tumor stroma and discuss how we can potentially design therapeutic avenues to target these molecules in order to overcome treatment resistance.
Collapse
Affiliation(s)
- Jin-Yih Low
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Correspondence: ; Tel.: +1-410-502-9748; Fax: +1-410-502-2821
| | - Marikki Laiho
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
8
|
Abstract
Caveolin-1 (CAV1) has long been implicated in cancer progression, and while widely accepted as an oncogenic protein, CAV1 also has tumor suppressor activity. CAV1 was first identified in an early study as the primary substrate of Src kinase, a potent oncoprotein, where its phosphorylation correlated with cellular transformation. Indeed, CAV1 phosphorylation on tyrosine-14 (Y14; pCAV1) has been associated with several cancer-associated processes such as focal adhesion dynamics, tumor cell migration and invasion, growth suppression, cancer cell metabolism, and mechanical and oxidative stress. Despite this, a clear understanding of the role of Y14-phosphorylated pCAV1 in cancer progression has not been thoroughly established. Here, we provide an overview of the role of Src-dependent phosphorylation of tumor cell CAV1 in cancer progression, focusing on pCAV1 in tumor cell migration, focal adhesion signaling and metabolism, and in the cancer cell response to stress pathways characteristic of the tumor microenvironment. We also discuss a model for Y14 phosphorylation regulation of CAV1 effector protein interactions via the caveolin scaffolding domain.
Collapse
|
9
|
Plasma from Volunteers Breathing Helium Reduces Hypoxia-Induced Cell Damage in Human Endothelial Cells-Mechanisms of Remote Protection Against Hypoxia by Helium. Cardiovasc Drugs Ther 2020; 33:297-306. [PMID: 31025141 PMCID: PMC6538579 DOI: 10.1007/s10557-019-06880-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Remote ischemic preconditioning protects peripheral organs against prolonged ischemia/reperfusion injury via circulating protective factors. Preconditioning with helium protected healthy volunteers against postischemic endothelial dysfunction. We investigated whether plasma from helium-treated volunteers can protect human umbilical vein endothelial cells (HUVECs) against hypoxia in vitro through release of circulating of factors. METHODS Healthy male volunteers inhaled heliox (79% helium, 21% oxygen) or air for 30 min. Plasma was collected at baseline, directly after inhalation, 6 h and 24 h after start of the experiment. HUVECs were incubated with either 5% or 10% of the plasma for 1 or 2 h and subjected to enzymatically induced hypoxia. Cell damage was measured by LDH content. Furthermore, caveolin 1 (Cav-1), hypoxia-inducible factor (HIF1α), extracellular signal-regulated kinase (ERK)1/2, signal transducer and activator of transcription (STAT3) and endothelial nitric oxide synthase (eNOS) were determined. RESULTS Prehypoxic exposure to 10% plasma obtained 6 h after helium inhalation decreased hypoxia-induced cell damage in HUVEC. Cav-1 knockdown in HUVEC abolished this effect. CONCLUSIONS Plasma of healthy volunteers breathing helium protects HUVEC against hypoxic cell damage, possibly involving circulating Cav-1.
Collapse
|
10
|
Xie H, Lu WC. Inhibition of transient receptor potential vanilloid 4 decreases the expressions of caveolin-1 and caveolin-2 after focal cerebral ischemia and reperfusion in rats. Neuropathology 2018; 38:337-346. [PMID: 29665111 DOI: 10.1111/neup.12469] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/20/2018] [Accepted: 03/18/2018] [Indexed: 01/26/2023]
Abstract
This study aimed to investigate the effects of transient receptor potential vanilloid 4 (TRPV4) inhibition on blood-brain barrier (BBB) integrity and the expressions of caveolae structural proteins caveolin-1 and caveolin-2 in rats with focal cerebral ischemia and reperfusion. BBB permeability was assessed by Evans blue extravasation. The mRNA and protein expressions of caveolin-1 and caveolin-2 were determined by RT-PCR, Western blot and immunohistochemistry assays. We found that BBB permeability significantly increased and reaches its peak at 72 h of reperfusion in cerebral ischemia-reperfusion rats and is able to be ameliorated by administration of HC-067047, an antagonist of TRPV4. Additionally, it shows a significant upregulation of caveolin-1 and caveolin-2 expression in cerebral microvessels of ischemic tissue. However, treatment with HC-067047 was shown to downregulate caveolin-1 and caveolin-2 expression during cerebral ischemia-reperfusion. This study demonstrates that inhibition of TRPV4 ameliorates BBB leakage induced by ischemia-reperfusion injury through the downregulation of caveolin-1 and caveolin-2.
Collapse
Affiliation(s)
- Hui Xie
- Department of Histology and Embryology, College of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Wei-Cheng Lu
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
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: 2.1] [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
|
12
|
Schilling JM, Head BP, Patel HH. Caveolins as Regulators of Stress Adaptation. Mol Pharmacol 2018; 93:277-285. [PMID: 29358220 PMCID: PMC5820539 DOI: 10.1124/mol.117.111237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/19/2018] [Indexed: 12/21/2022] Open
Abstract
Caveolins have been recognized over the past few decades as key regulators of cell physiology. They are ubiquitously expressed and regulate a number of processes that ultimately impact efficiency of cellular processes. Though not critical to life, they are central to stress adaptation in a number of organs. The following review will focus specifically on the role of caveolin in stress adaptation in the heart, brain, and eye, three organs that are susceptible to acute and chronic stress and that show as well declining function with age. In addition, we consider some novel molecular mechanisms that may account for this stress adaptation and also offer potential to drive the future of caveolin research.
Collapse
Affiliation(s)
- Jan M Schilling
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| | - Brian P Head
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| | - Hemal H Patel
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| |
Collapse
|
13
|
Smit KF, Konkel M, Kerindongo R, Landau MA, Zuurbier CJ, Hollmann MW, Preckel B, Nieuwland R, Albrecht M, Weber NC. Helium alters the cytoskeleton and decreases permeability in endothelial cells cultured in vitro through a pathway involving Caveolin-1. Sci Rep 2018; 8:4768. [PMID: 29555979 PMCID: PMC5859123 DOI: 10.1038/s41598-018-23030-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/05/2018] [Indexed: 01/31/2023] Open
Abstract
Caveolins are involved in anaesthetic-induced cardioprotection. Actin filaments are located in close connection to Caveolins in the plasma membrane. We hypothesised that helium might affect the cytoskeleton and induce secretion of Caveolin. HCAEC, HUVEC and Cav-1 siRNA transfected HUVEC were exposed for 20 minutes to either helium (5% CO2, 25% O2, 70% He) or control gas (5% CO2, 25% O2, 70% N2). Cells and supernatants were collected for infrared Western blot analysis, immunofluorescence staining, nanoparticle tracking analysis and permeability measurements. Helium treatment increased the cortical localisation of F-actin fibers in HUVEC. After 6 hours, helium decreased cellular Caveolin-1 (Cav-1) levels and increased Cav-1 levels in the supernatant. Cell permeability was decreased 6 and 12 hours after helium treatment, and increased levels of Vascular Endothelial - Cadherin (VE-Cadherin) and Connexin 43 (Cx43) were observed. Transfection with Cav-1 siRNA abolished the effects of helium treatment on VE-Cadherin, Cx43 levels and permeability. Supernatant obtained after helium treatment reduced cellular permeability in remote HUVEC, indicating that increased levels of Cav-1 are responsible for the observed alterations. These findings suggest that Cav-1 is secreted after helium exposure in vitro, altering the cytoskeleton and increasing VE-Cadherin and Cx43 expression resulting in decreased permeability in HUVEC.
Collapse
Affiliation(s)
- Kirsten F Smit
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Moritz Konkel
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Anaesthesiology, UKSH, Campus Kiel, Kiel, Germany
| | - Raphaela Kerindongo
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Maximilian A Landau
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Anaesthesiology, UKSH, Campus Kiel, Kiel, Germany
| | - Coert J Zuurbier
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, and Vesicle Observation Centre, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Martin Albrecht
- Department of Anaesthesiology, UKSH, Campus Kiel, Kiel, Germany
| | - Nina C Weber
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A), Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Abu-Taha IH, Heijman J, Feng Y, Vettel C, Dobrev D, Wieland T. Regulation of heterotrimeric G-protein signaling by NDPK/NME proteins and caveolins: an update. J Transl Med 2018; 98:190-197. [PMID: 29035382 DOI: 10.1038/labinvest.2017.103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 07/17/2017] [Accepted: 07/31/2017] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G proteins are pivotal mediators of cellular signal transduction in eukaryotic cells and abnormal G-protein signaling plays an important role in numerous diseases. During the last two decades it has become evident that the activation status of heterotrimeric G proteins is both highly localized and strongly regulated by a number of factors, including a receptor-independent activation pathway of heterotrimeric G proteins that does not involve the classical GDP/GTP exchange and relies on nucleoside diphosphate kinases (NDPKs). NDPKs are NTP/NDP transphosphorylases encoded by the nme/nm23 genes that are involved in a variety of cellular events such as proliferation, migration, and apoptosis. They therefore contribute, for example, to tumor metastasis, angiogenesis, retinopathy, and heart failure. Interestingly, NDPKs are translocated and/or upregulated in human heart failure. Here we describe recent advances in the current understanding of NDPK functions and how they have an impact on local regulation of G-protein signaling.
Collapse
Affiliation(s)
- Issam H Abu-Taha
- Institute of Pharmacology, West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, The Netherlands
| | - Yuxi Feng
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Christiane Vettel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany
| |
Collapse
|
15
|
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: 4.9] [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
|
16
|
Busija AR, Patel HH, Insel PA. Caveolins and cavins in the trafficking, maturation, and degradation of caveolae: implications for cell physiology. Am J Physiol Cell Physiol 2017; 312:C459-C477. [PMID: 28122734 DOI: 10.1152/ajpcell.00355.2016] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/09/2023]
Abstract
Caveolins (Cavs) are ~20 kDa scaffolding proteins that assemble as oligomeric complexes in lipid raft domains to form caveolae, flask-shaped plasma membrane (PM) invaginations. Caveolae ("little caves") require lipid-lipid, protein-lipid, and protein-protein interactions that can modulate the localization, conformational stability, ligand affinity, effector specificity, and other functions of proteins that are partners of Cavs. Cavs are assembled into small oligomers in the endoplasmic reticulum (ER), transported to the Golgi for assembly with cholesterol and other oligomers, and then exported to the PM as an intact coat complex. At the PM, cavins, ~50 kDa adapter proteins, oligomerize into an outer coat complex that remodels the membrane into caveolae. The structure of caveolae protects their contents (i.e., lipids and proteins) from degradation. Cellular changes, including signal transduction effects, can destabilize caveolae and produce cavin dissociation, restructuring of Cav oligomers, ubiquitination, internalization, and degradation. In this review, we provide a perspective of the life cycle (biogenesis, degradation), composition, and physiologic roles of Cavs and caveolae and identify unanswered questions regarding the roles of Cavs and cavins in caveolae and in regulating cell physiology.1.
Collapse
Affiliation(s)
- Anna R Busija
- Department of Anesthesiology, University of California, San Diego, La Jolla, California.,Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Hemal H Patel
- Department of Anesthesiology, University of California, San Diego, La Jolla, California
| | - Paul A Insel
- Department of Medicine, University of California, San Diego, La Jolla, California; and .,Department of Pharmacology, University of California, San Diego, La Jolla, California
| |
Collapse
|
17
|
Fernando CA, Liu Y, Sowa G, Segal SS. Attenuated rapid onset vasodilation with greater force production in skeletal muscle of caveolin-2-/- mice. Am J Physiol Heart Circ Physiol 2016; 311:H415-25. [PMID: 27317631 DOI: 10.1152/ajpheart.00082.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/15/2016] [Indexed: 11/22/2022]
Abstract
Caveolin-2 (Cav2) is a major protein component of caveolae in membranes of vascular smooth muscle and endothelium, yet its absence alters the ultrastructure of skeletal muscle fibers. To gain insight into Cav2 function in skeletal muscle, we tested the hypothesis that genetic deletion of Cav2 would alter microvascular reactivity and depress contractile function of skeletal muscle in vivo. In the left gluteus maximus muscle (GM) of anesthetized Cav2(-/-) and wild-type (WT) male mice (age, 6 mo), microvascular responses to physiological agonists and to GM contractions were studied at 34°C. For feed arteries (FA), first- (1A), second- (2A) and third-order (3A) arterioles, respective mean diameters at rest (45, 35, 25, 12 μm) and during maximal dilation (65, 55, 45, 30 μm) were similar between groups. Cumulative dilations to ACh (10(-9) to 10(-5) M) and constrictions to norepinephrine (10(-9) to 10(-5) M) were also similar between groups, as were steady-state dilations during rhythmic twitch contractions (2 and 4 Hz; 30 s). For single tetanic contractions (100 Hz; 100, 250, and 500 ms), rapid onset vasodilation (ROV) increased with contraction duration throughout networks in GM of both groups but was reduced by nearly half in Cav2(-/-) mice compared with WT mice (P < 0.05). Nevertheless, maximal force during tetanic contraction was ∼40% greater in GM of Cav2(-/-) vs. WT mice (152 ± 14 vs. 110 ± 3 mN per square millimeter, respectively; P < 0.05). Thus, while structural and functional properties of resistance networks are well maintained in the GM of Cav2(-/-) mice, diminished ROV with greater force production reveals novel physiological roles for Cav2 in skeletal muscle.
Collapse
Affiliation(s)
- Charmain A Fernando
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
| | - Yajun Liu
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
| | - Grzegorz Sowa
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
18
|
Patel A, Sabbineni H, Clarke A, Somanath PR. Novel roles of Src in cancer cell epithelial-to-mesenchymal transition, vascular permeability, microinvasion and metastasis. Life Sci 2016; 157:52-61. [PMID: 27245276 DOI: 10.1016/j.lfs.2016.05.036] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 12/21/2022]
Abstract
The Src-family kinases (SFKs), an intracellularly located group of non-receptor tyrosine kinases are involved in oncogenesis. The importance of SFKs has been implicated in the promotion of tumor cell motility, proliferation, inhibition of apoptosis, invasion and metastasis. Recent evidences indicate that specific effects of SFKs on epithelial-to-mesenchymal transition (EMT) as well as on endothelial and stromal cells in the tumor microenvironment can have profound effects on tumor microinvasion and metastasis. Although, having been studied extensively, these novel features of SFKs may contribute to greater understanding of benefits from Src inhibition in various types of cancers. Here we review the novel role of SFKs, particularly c-Src in mediating EMT, modulation of tumor endothelial-barrier, transendothelial migration (microinvasion) and metastasis of cancer cells, and discuss the utility of Src inhibitors in vascular normalization and cancer therapy.
Collapse
Affiliation(s)
- Ami Patel
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States
| | - Harika Sabbineni
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Andrea Clarke
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States; Department of Medicine, Vascular Biology Center and Cancer Center, Augusta University, Augusta, GA, United States.
| |
Collapse
|
19
|
Desai A, Xu J, Aysola K, Akinbobuyi O, White M, Reddy VE, Okoli J, Clark C, Partridge EE, Childs E, Beech DJ, Rice MV, Reddy E, Rao VN. Molecular Mechanism Linking BRCA1 Dysfunction to High Grade Serous Epithelial Ovarian Cancers with Peritoneal Permeability and Ascites. ACTA ACUST UNITED AC 2015; 1. [PMID: 26665166 DOI: 10.15744/2454-3284.1.103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer constitutes the second most common gynecological cancer with a five-year survival rate of 40%. Among the various histotypes associated with hereditary ovarian cancer, high-grade serous epithelial ovarian carcinoma (HGSEOC) is the most predominant and women with inherited mutations in BRCA1 have a lifetime risk of 40-60%. HGSEOC is a challenge for clinical oncologists, due to late presentation of patient, diagnosis and high rate of relapse. Ovarian tumors have a wide range of clinical presentations including development of ascites as a result of deregulated endothelial function thereby causing increased vascular permeability of peritoneal vessels. The molecular mechanisms remain elusive. Studies have shown that fallopian tube cancers develop in women with BRCA1 gene mutations more often than previously suspected. Recent studies suggest that many primary peritoneal cancers and some high-grade serous epithelial ovarian carcinomas actually start in the fallopian tubes. In this article we have addressed the molecular pathway of a recently identified potential biomarker Ubc9 whose deregulated expression due to BRCA1 dysfunction can result in HGSEOC with peritoneal permeability and formation of ascites. We also discuss the role of downstream targets Caveolin-1 and Vascular Endothelial Growth Factor (VEGF) in the pathogenesis of ascites in ovarian carcinomas. Finally we hypothesize a signaling axis between Ubc9 over expression, loss of Caveolin-1 and induction of VEGF in BRCA1 mutant HGSEOC cells. We suggest that Ubc9-mediated stimulation of VEGF as a novel mechanism underlying ovarian cancer aggressiveness and ascites formation. Agents that target Ubc9 and VEGF signaling may represent a novel therapeutic strategy to impede peritoneal growth and spread of HGSEOC.
Collapse
Affiliation(s)
- A Desai
- Cancer Biology Program, Department of OB/GYN, School of Medicine, Georgia Cancer Center for Excellence, Grady Health System, Atlanta, USA
| | - J Xu
- Department of Internal Medicine, School of Medicine, Georgia Cancer Center for Excellence, Grady Health System, Atlanta, USA
| | - K Aysola
- Department of Surgery, Morehouse, School of Medicine, Georgia Cancer Center for Excellence, Grady Health System, Atlanta, USA
| | - O Akinbobuyi
- Department of Internal Medicine, University of Buffalo, Erie County Medical Center, Buffalo NY
| | - M White
- Philadelphia College of Osteopathic Medicine, Suwanee GA
| | - V E Reddy
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hernández-Mosqueira C, Velez-delValle C, Kuri-Harcuch W. Tissue alkaline phosphatase is involved in lipid metabolism and gene expression and secretion of adipokines in adipocytes. Biochim Biophys Acta Gen Subj 2015; 1850:2485-96. [PMID: 26391843 DOI: 10.1016/j.bbagen.2015.09.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/20/2015] [Accepted: 09/11/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND Alkaline phosphatases are dimeric hydrolytic enzymes that dephosphorylate nucleotides and proteins. AP-TNAP is found primarily in skeletal tissues were it plays a major role in the mineralization of the extracellular matrix and bone formation. METHODS In this study we found through conventional and real time PCR assays that Alpl, the gene encoding for AP-TNAP is expressed in adipose tissue and in 3 T3-F442A adipocytes. We evaluated, using RNAi its role in adipocyte metabolism, and its cytoplasmic location by immunohistochemistry. RESULTS Alpl is highly expressed late in adipogenesis during adipose terminal differentiation. Knocking down Alpl increased the expression of the genes encoding for glycerophosphate dehydrogenase, and for the adipokines adiponectin, and FABP4 (aP2) but decreased that of leptin, and it also increased secretion of FABP4; these 3 proteins are important in adipocyte systemic signaling and insulin sensitivity. Inhibition of alkaline phosphatase activity in adipocytes by levamisole reduced lipolysis and the expression of various lipogenic genes. We found the enzyme intracytoplasmically, forming aggregates in close surroundings of the lipid droplets during lipolysis. CONCLUSIONS We suggest that AP-TNAP activity is involved in lipid and energy metabolism of fat cells, and it might regulate glucose metabolism and insulin sensitivity via adipokine synthesis and secretion. GENERAL SIGNIFICANCE The activity of AP-TNAP might have a critical role in the energy balance of the adipocyte, probably participating in obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Claudia Hernández-Mosqueira
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute, Apdo. Postal 14-740, México City, 07000, México
| | - Cristina Velez-delValle
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute, Apdo. Postal 14-740, México City, 07000, México
| | - Walid Kuri-Harcuch
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute, Apdo. Postal 14-740, México City, 07000, México.
| |
Collapse
|
21
|
Chettimada S, Yang J, Moon HG, Jin Y. Caveolae, caveolin-1 and cavin-1: Emerging roles in pulmonary hypertension. World J Respirol 2015; 5:126-134. [PMID: 28529892 PMCID: PMC5438095 DOI: 10.5320/wjr.v5.i2.126] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 02/25/2015] [Accepted: 06/16/2015] [Indexed: 02/06/2023] Open
Abstract
Caveolae are flask-shaped invaginations of cell membrane that play a significant structural and functional role. Caveolae harbor a variety of signaling molecules and serve to receive, concentrate and transmit extracellular signals across the membrane. Caveolins are the main structural proteins residing in the caveolae. Caveolins and another category of newly identified caveolae regulatory proteins, named cavins, are not only responsible for caveolae formation, but also interact with signaling complexes in the caveolae and regulate transmission of signals across the membrane. In the lung, two of the three caveolin isoforms, i.e., cav-1 and -2, are expressed ubiquitously. Cavin protein family is composed of four proteins, named cavin-1 (or PTRF for polymerase I and transcript release factor), cavin-2 (or SDPR for serum deprivation protein response), cavin-3 (or SRBC for sdr-related gene product that binds to-c-kinase) and cavin-4 (or MURC for muscle restricted coiled-coiled protein or cavin-4). All the caveolin and cavin proteins are essential regulators for caveolae dynamics. Recently, emerging evidence suggest that caveolae and its associated proteins play crucial roles in development and progression of pulmonary hypertension. The focus of this review is to outline and discuss the contrast in alteration of cav-1 (cav-1),-2 and cavin-1 (PTRF) expression and downstream signaling mechanisms between human and experimental models of pulmonary hypertension.
Collapse
|
22
|
Low JY, Nicholson HD. Epigenetic modifications of caveolae associated proteins in health and disease. BMC Genet 2015; 16:71. [PMID: 26112043 PMCID: PMC4482180 DOI: 10.1186/s12863-015-0231-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Caveolae are small, “omega-shaped” invaginations at the plasma membrane of the cell which are involved in a variety of processes including cholesterol transport, potocytosis and cell signalling. Within caveolae there are caveolae-associated proteins, and changes in expression of these molecules have been described to play a role in the pathophysiology of various diseases including cancer and cardiovascular disease. Evidence is beginning to accumulate that epigenetic processes may regulate the expression of these caveolae related genes, and hence contribute to disease progression. Here, we summarize the current knowledge of the role of epigenetic modification in regulating the expression of these caveolae related genes and how this relates to changes in cellular physiology and in health and disease.
Collapse
Affiliation(s)
- Jin-Yih Low
- Department of Anatomy, Otago School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, 9054, New Zealand.
| | - Helen D Nicholson
- Department of Anatomy, Otago School of Medical Sciences, University of Otago, P.O. Box 913, Dunedin, 9054, New Zealand.
| |
Collapse
|
23
|
Vu LT, Keschrumrus V, Zhang X, Zhong JF, Su Q, Kabeer MH, Loudon WG, Li SC. Tissue elasticity regulated tumor gene expression: implication for diagnostic biomarkers of primitive neuroectodermal tumor. PLoS One 2015; 10:e0120336. [PMID: 25774514 PMCID: PMC4361745 DOI: 10.1371/journal.pone.0120336] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 02/05/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The tumor microenvironment consists of both physical and chemical factors. Tissue elasticity is one physical factor contributing to the microenvironment of tumor cells. To test the importance of tissue elasticity in cell culture, primitive neuroectodermal tumor (PNET) stem cells were cultured on soft polyacrylamide (PAA) hydrogel plates that mimics the elasticity of brain tissue compared with PNET on standard polystyrene (PS) plates. We report the molecular profiles of PNET grown on either PAA or PS. METHODOLOGY/PRINCIPAL FINDINGS A whole-genome microarray profile of transcriptional expression between the two culture conditions was performed as a way to probe effects of substrate on cell behavior in culture. The results showed more genes downregulated on PAA compared to PS. This led us to propose microRNA (miRNA) silencing as a potential mechanism for downregulation. Bioinformatic analysis predicted a greater number of miRNA binding sites from the 3' UTR of downregulated genes and identified as specific miRNA binding sites that were enriched when cells were grown on PAA-this supports the hypothesis that tissue elasticity plays a role in influencing miRNA expression. Thus, Dicer was examined to determine if miRNA processing was affected by tissue elasticity. Dicer genes were downregulated on PAA and had multiple predicted miRNA binding sites in its 3' UTR that matched the miRNA binding sites found enriched on PAA. Many differentially regulated genes were found to be present on PS but downregulated on PAA were mapped onto intron sequences. This suggests expression of alternative polyadenylation sites within intron regions that provide alternative 3' UTRs and alternative miRNA binding sites. This results in tissue specific transcriptional downregulation of mRNA in humans by miRNA. We propose a mechanism, driven by the physical characteristics of the microenvironment by which downregulation of genes occur. We found that tissue elasticity-mediated cytokines (TGFβ2 and TNFα) signaling affect expression of ECM proteins. CONCLUSIONS Our results suggest that tissue elasticity plays important roles in miRNA expression, which, in turn, regulate tumor growth or tumorigenicity.
Collapse
Affiliation(s)
- Long T. Vu
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave., Orange, CA, 92868, United States of America
- Department of Biological Science, California State University, Fullerton, CA, 92834, United States of America
| | - Vic Keschrumrus
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave., Orange, CA, 92868, United States of America
| | - Xi Zhang
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States of America
| | - Jiang F. Zhong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States of America
| | - Qingning Su
- Bioengineering Research Center, School of Medicine, Shenzhen University, Shenzhen, 518057, Guangdong, China
| | - Mustafa H. Kabeer
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave., Orange, CA, 92868, United States of America
- Department of Pediatric Surgery, CHOC Children's Hospital, 1201 West La Veta Ave., Orange, CA, 92868, United States of America
- Department of Surgery, University of California Irvine School of Medicine, 333 City Blvd. West, Suite 700, Orange, CA 92868, United States of America
| | - William G. Loudon
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave., Orange, CA, 92868, United States of America
- Department of Neurological Surgery, Saint Joseph Hospital, Orange, CA, 92868, United States of America
- Department of Neurological Surgery, University of California Irvine School of Medicine, Orange, CA, 92862, United States of America
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Hospital Research Institute, University of California Irvine, 1201 West La Veta Ave., Orange, CA, 92868, United States of America
- Department of Neurology, University of California Irvine School of Medicine, Orange, CA, 92697–4292, United States of America
- Department of Biological Science, California State University, Fullerton, CA, 92834, United States of America
| |
Collapse
|
24
|
Xu L, Guo R, Xie Y, Ma M, Ye R, Liu X. Caveolae: molecular insights and therapeutic targets for stroke. Expert Opin Ther Targets 2015; 19:633-50. [PMID: 25639269 DOI: 10.1517/14728222.2015.1009446] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Caveolae are specialized plasma membrane micro-invaginations of most mammalian cell types. The organization and function of caveolae are carried out by their coat proteins, caveolins and adaptor proteins, cavins. Caveolae/caveolins physically interact with membrane-associated signaling molecules and function in cholesterol incorporation, signaling transduction and macromolecular transport/permeability. AREAS COVERED Recent investigations have implicated a check-and-balance role of caveolae in the pathophysiology of cerebral ischemia. Caveolin knockout mice displayed exacerbated ischemic injury, whereas caveolin peptide exerted remarkable protection against ischemia/reperfusion injury. This review attempts to provide a comprehensive synopsis of how caveolae/caveolins modulate blood-brain barrier permeability, pro-survival signaling, angiogenesis and neuroinflammation, and how this may contribute to a better understanding of the participation of caveolae in ischemic cascade. The role of caveolin in the preconditioning-induced tolerance against ischemia is also discussed. EXPERT OPINION Caveolae represent a novel target for cerebral ischemia. It remains open how to manipulate caveolin expression in a practical way to recapitulate the beneficial therapeutic outcomes. Caveolin peptides and associated antagomirs may be efficacious and deserve further investigations for their potential benefits for stroke.
Collapse
Affiliation(s)
- Lili Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University , Nanjing 210002 , China
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Sodium current in the heart flows principally through the pore protein NaV1.5, which is part of a complex of interacting proteins that serve both to target and localize the complex in the membrane, and to modulate function by such post-translational modifications as phosphorylation and nitrosylation. Multiple mutations in seven different NaV1.5 interacting proteins have been associated with dysfunctional sodium current and inherited cardiac diseases, including long QT syndrome, Brugada syndrome, atrial fibrillation, and cardiomyopathy, as well as sudden infant death syndrome (SIDS). Mutations in as yet unidentified interacting proteins may account for cardiac disease for which a genetic basis has not yet been established. Characterizing the mechanisms by which these mutations cause disease may give insight into etiologies and treatments of more common acquired cardiac disease, such as ischemia and heart failure.
Collapse
Affiliation(s)
- John W Kyle
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin, Madison, Wisconsin, USA 53792
| | - Jonathan C Makielski
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin, Madison, Wisconsin, USA 53792
| |
Collapse
|
26
|
Wang X, Liu T, Bai Y, Liao H, Qiu S, Chang Z, Liu Y, Yan X, Guo H. Polymerase I and transcript release factor acts as an essential modulator of glioblastoma chemoresistance. PLoS One 2014; 9:e93439. [PMID: 24747515 PMCID: PMC3991573 DOI: 10.1371/journal.pone.0093439] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 03/04/2014] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES This study is to investigate if polymerase I and transcript release factor (PTRF) acts as a modulator in glioblastoma (GBM) chemoresistance. METHODS Multidrug resistant (MDR) GBM cell line U251AR was established by exposing the U251 cell line to imatinib. The 2D-DIGE and MALDI-TOF/TOF-MS were performed on U251 and U251AR cell lines to screen MDR-related proteins. The expression of PTRF was determined by Western blot and quantitative RT-PCR analyses. RESULTS When compared with the parental U251 cells, expression of 21 proteins was significantly altered in U251AR cells. Among the 21 differentially expressed proteins, the expression of PTRF was up-regulated by 2.14 folds in U251AR cells when compared with that in the parental U251 cells. Knockdown of PTRF in GBM cell lines significantly increased chemosensitivity of cells to various chemical drugs and decreased the expression levels of caveolin1, a major structural component of caveolae. Expression levels of PTRF and caveolin1 were significantly up-regulated in the relapsed GBM patients. The mRNA level of PTRF and caveolin1 showed a positive correlation in the same GBM specimens. CONCLUSIONS Our results indicate that PTRF acts as a modulator in GBM chemoresistance.
Collapse
Affiliation(s)
- Xin Wang
- The National Key Clinic Specialty, the Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, P. R. China
| | - Tianzhu Liu
- The National Key Clinic Specialty, the Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Yifeng Bai
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, P. R. China
| | - Hongzhan Liao
- The National Key Clinic Specialty, the Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Shengcong Qiu
- The National Key Clinic Specialty, the Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Zhenhua Chang
- Department of Laboratory Medicine, Tongchuan People's Hospital, Tongchuan, P. R. China
| | - Yanting Liu
- The National Key Clinic Specialty, the Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Xiaohui Yan
- Clinical Research Centre, Nanfang Hospital of Southern Medical University, Guangzhou, P. R. China
| | - Hongbo Guo
- The National Key Clinic Specialty, the Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
27
|
Swärd K, Albinsson S, Rippe C. Arterial dysfunction but maintained systemic blood pressure in cavin-1-deficient mice. PLoS One 2014; 9:e92428. [PMID: 24658465 PMCID: PMC3962402 DOI: 10.1371/journal.pone.0092428] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/21/2014] [Indexed: 12/26/2022] Open
Abstract
Caveolae are omega-shaped plasma membrane micro-domains that are abundant in cells of the vascular system. Formation of caveolae depends on caveolin-1 and cavin-1 and lack of either protein leads to loss of caveolae. Mice with caveolin-1 deficiency have dysfunctional blood vessels, but whether absence of cavin-1 similarly leads to vascular dysfunction is not known. Here we addressed this hypothesis using small mesenteric arteries from cavin-1-deficient mice. Cavin-1-reporter staining was intense in mesenteric arteries, brain arterioles and elsewhere in the vascular system, with positive staining of both endothelial and smooth muscle cells. Arterial expression of cavin-1, -2 and -3 was reduced in knockout (KO) arteries as was expression of caveolin-1, -2 and -3. Caveolae were absent in the endothelial and smooth muscle layers of small mesenteric arteries as determined by electron microscopy. Arginase, a negative regulator of nitric oxide production, was elevated in cavin-1 deficient arteries as was contraction in response to the α1-adrenergic agonist cirazoline. Detailed assessment of vascular dimensions revealed increased media thickness and reduced distensibility, arguing that enhanced contraction was due to increased muscle mass. Contrasting with increased α1-adrenergic contraction, myogenic tone was essentially absent and this appeared to be due in part to increased nitric oxide production. Vasomotion was less frequent in the knock-out vessels. In keeping with the opposing influences on arterial resistance of increased agonist-induced contractility and reduced myogenic tone, arterial blood pressure was unchanged in vivo. We conclude that deficiency of cavin-1 affects the function of small arteries, but that opposing influences on arterial resistance balance each other such that systemic blood pressure in unstressed mice is well maintained.
Collapse
Affiliation(s)
- Karl Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Catarina Rippe
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Cheng J, Valdivia CR, Vaidyanathan R, Balijepalli RC, Ackerman MJ, Makielski JC. Caveolin-3 suppresses late sodium current by inhibiting nNOS-dependent S-nitrosylation of SCN5A. J Mol Cell Cardiol 2013; 61:102-10. [PMID: 23541953 PMCID: PMC3720711 DOI: 10.1016/j.yjmcc.2013.03.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 03/11/2013] [Accepted: 03/12/2013] [Indexed: 10/27/2022]
Abstract
AIMS Mutations in CAV3-encoding caveolin-3 (Cav3) have been implicated in type 9 long QT syndrome (LQT9) and sudden infant death syndrome (SIDS). When co-expressed with SCN5A-encoded cardiac sodium channels these mutations increased late sodium current (INa) but the mechanism was unclear. The present study was designed to address the mechanism by which the LQT9-causing mutant Cav3-F97C affects the function of caveolar SCN5A. METHODS AND RESULTS HEK-293 cells expressing SCN5A and LQT9 mutation Cav3-F97C resulted in a 2-fold increase in late INa compared to Cav3-WT. This increase was reversed by the neural nitric oxide synthase (nNOS) inhibitor L-NMMA. Based on these findings, we hypothesized that an nNOS complex mediated the effect of Cav3 on SCN5A. A SCN5A macromolecular complex was established in HEK-293 cells by transiently expressing SCN5A, α1-syntrophin (SNTA1), nNOS, and Cav3. Compared with Cav3-WT, Cav3-F97C produced significantly larger peak INa amplitudes, and showed 3.3-fold increase in the late INa associated with increased S-nitrosylation of SCN5A. L-NMMA reversed both the Cav3-F97C induced increase in late and peak INa and decreased S-nitrosylation of SCN5A. Overexpression of Cav3-F97C in adult rat cardiomyocytes caused a significant increase in late INa compared to Cav3-WT, and prolonged the action potential duration (APD90) in a nNOS-dependent manner. CONCLUSIONS Cav3 is identified as an important negative regulator for cardiac late INa via nNOS dependent direct S-nitrosylation of SCN5A. This provides a molecular mechanism for how Cav3 mutations increase late INa to cause LQT9. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".
Collapse
Affiliation(s)
- Jianding Cheng
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI 53792, USA
- Department of Forensic Pathology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Carmen R. Valdivia
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI 53792, USA
| | - Ravi Vaidyanathan
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI 53792, USA
| | - Ravi C. Balijepalli
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI 53792, USA
| | - Michael J. Ackerman
- Divisions of Cardiovascular Diseases and Pediatric Cardiology, Departments of Medicine, Pediatrics, and Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jonathan C. Makielski
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI 53792, USA
| |
Collapse
|
29
|
Lige B, Romano JD, Sampels V, Sonda S, Joiner KA, Coppens I. Introduction of caveolae structural proteins into the protozoan Toxoplasma results in the formation of heterologous caveolae but not caveolar endocytosis. PLoS One 2012; 7:e51773. [PMID: 23272165 PMCID: PMC3522706 DOI: 10.1371/journal.pone.0051773] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/08/2012] [Indexed: 11/30/2022] Open
Abstract
Present on the plasma membrane of most metazoans, caveolae are specialized microdomains implicated in several endocytic and trafficking mechanisms. Caveolins and the more recently discovered cavins are the major protein components of caveolae. Previous studies reported that caveolar invaginations can be induced de novo on the surface of caveolae-negative mammalian cells upon heterologous expression of caveolin-1. However, it remains undocumented whether other components in the transfected cells participate in caveolae formation. To address this issue, we have exploited the protozoan Toxoplasma as a heterologous expression system to provide insights into the minimal requirements for caveogenesis and caveolar endocytosis. Upon expression of caveolin-1, Toxoplasma accumulates prototypical exocytic caveolae 'precursors' in the cytoplasm. Toxoplasma expressing caveolin-1 alone, or in conjunction with cavin-1, neither develops surface-located caveolae nor internalizes caveolar ligands. These data suggest that the formation of functional caveolae at the plasma membrane in Toxoplasma and, by inference in all non-mammalian cells, requires effectors other than caveolin-1 and cavin-1. Interestingly, Toxoplasma co-expressing caveolin-1 and cavin-1 displays an impressive spiraled network of membranes containing the two proteins, in the cytoplasm. This suggests a synergistic activity of caveolin-1 and cavin-1 in the morphogenesis and remodeling of membranes, as illustrated for Toxoplasma.
Collapse
Affiliation(s)
- Bao Lige
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| | - Vera Sampels
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| | - Sabrina Sonda
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| | - Keith A. Joiner
- Arizona Health Science Center, University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| |
Collapse
|
30
|
Increased caveolae density and caveolin-1 expression accompany impaired NO-mediated vasorelaxation in diet-induced obesity. Histochem Cell Biol 2012; 139:309-21. [DOI: 10.1007/s00418-012-1032-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2012] [Indexed: 01/24/2023]
|
31
|
Tourkina E, Hoffman S. Caveolin-1 signaling in lung fibrosis. Open Rheumatol J 2012; 6:116-22. [PMID: 22802909 PMCID: PMC3396359 DOI: 10.2174/1874312901206010116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 03/27/2012] [Accepted: 04/04/2012] [Indexed: 01/05/2023] Open
Abstract
Caveolin-1 is a master regulator of several signaling cascades because it is able to bind to and thereby inhibit members of a variety of kinase families. While associated with caveolae and involved in their generation, caveolin-1 is also present at other sites. A variety of studies have suggested that caveolin-1 may be a useful therapeutic target in fibrotic diseases of the lung and other tissues because in these diseases a low level of caveolin-1 expression is associated with a high level of collagen expression and fibrosis. Reduced caveolin-1 expression is observed not only in the fibroblasts that secrete collagen, but also in epithelial cells and monocytes. This is intriguing because both epithelial cells and monocytes have been suggested to be precursors of fibroblasts. Likely downstream effects of loss of caveolin-1 in fibrosis include activation of TGF-β signaling and upregulation of CXCR4 in monocytes resulting in their enhanced migration into damaged tissue where its ligand CXCL12 is produced. Finally, it may be possible to target caveolin-1 in fibrotic diseases without the use of gene therapy. A caveolin-1 peptide (caveolin-1 scaffolding domain) has been identified that retains the function of the full-length molecule to inhibit kinases and that can be modified by addition of the Antennapedia internalization sequence to allow it to enter cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Elena Tourkina
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 912, MSC 637, Charleston, SC 29425, USA
| | | |
Collapse
|
32
|
Jablonski EM, Hughes FM. The potential role of caveolin-1 in inhibition of aquaporins during the AVD. Biol Cell 2012; 98:33-42. [PMID: 16354160 DOI: 10.1042/bc20040131] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND INFORMATION During apoptosis, the first morphological change is a distinct cell shrinkage known as the AVD (apoptotic volume decrease). This event is driven by a loss of intracellular K(+), which creates an osmotic gradient, drawing water out of the cell through AQPs (aquaporins). Loss of water in balance with K(+) would create a shrunken cell with an equivalent intracellular concentration of K(+) ([K(+)](i) = 140 mM). However, we have previously shown that the [K(+)](i) of the shrunken apoptotic cell is 35 mM, and this level is absolutely essential for the activation of apoptotic enzymes. We have recently found that AQPs are inactivated following the AVD, so that continued loss of K(+) will reduce the intracellular concentration to this critical level. Using thymocytes, we have investigated the expression profile and regulation of the AQP family members. RESULTS In the present study, we have found that AQP1, AQP8 and AQP9 are present in non-apoptotic thymocytes and localized primarily to the plasma membrane. Expression and localization did not change when these cells were induced to undergo apoptosis by growth factor withdrawal for 24 h. To explore other possible mechanisms by which these water channels are inactivated, we investigated their association with CAV-1 (caveolin-1), binding to which is known to inactivate a variety of proteins. We found that CAV-1 is present in thymocytes and that this protein co-localizes with a portion of AQP1 in normal (non-apoptotic) thymocytes. However, thymocytes induced to undergo apoptosis greatly increase their AQP1/CAV-1 association. CONCLUSIONS Taken together, these results indicate that AQPs are localized to the plasma membrane of shrunken apoptotic thymocytes where increased binding to CAV-1 potentially inactivates them. AQP inactivation, coupled with continued K(+) efflux, then allows the [K(+)](i) to decrease to levels conducive for the activation of downstream apoptotic enzymes and the completion of the apoptotic cascade.
Collapse
|
33
|
Dávalos A, Fernández-Hernando C, Sowa G, Derakhshan B, Lin MI, Lee JY, Zhao H, Luo R, Colangelo C, Sessa WC. Quantitative proteomics of caveolin-1-regulated proteins: characterization of polymerase i and transcript release factor/CAVIN-1 IN endothelial cells. Mol Cell Proteomics 2010; 9:2109-24. [PMID: 20585024 PMCID: PMC2953909 DOI: 10.1074/mcp.m110.001289] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Indexed: 12/15/2022] Open
Abstract
Caveolae are organelles abundant in the plasma membrane of many specialized cells including endothelial cells (ECs), epithelial cells, and adipocytes, and in these cells, caveolin-1 (Cav-1) is the major coat protein essential for the formation of caveolae. To identify proteins that require Cav-1 for stable incorporation into membrane raft domains, a quantitative proteomics analysis using isobaric tagging for relative and absolute quantification was performed on rafts isolated from wild-type and Cav-1-deficient mice. In three independent experiments, 117 proteins were consistently identified in membrane rafts with the largest differences in the levels of Cav-2 and in the caveola regulatory proteins Cavin-1 and Cavin-2. Because the lung is highly enriched in ECs, we validated and characterized the role of the newly described protein Cavin-1 in several cardiovascular tissues and in ECs. Cavin-1 was highly expressed in ECs lining blood vessels and in cultured ECs. Knockdown of Cavin-1 reduced the levels of Cav-1 and -2 and weakly influenced the formation of high molecular weight oligomers containing Cav-1 and -2. Cavin-1 silencing enhanced basal nitric oxide release from ECs but blocked proangiogenic phenotypes such as EC proliferation, migration, and morphogenesis in vitro. Thus, these data support an important role of Cavin-1 as a regulator of caveola function in ECs.
Collapse
Affiliation(s)
- Alberto Dávalos
- From the ‡Department of Pharmacology and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Carlos Fernández-Hernando
- From the ‡Department of Pharmacology and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Grzegorz Sowa
- ¶Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri 65212
| | - Behrad Derakhshan
- From the ‡Department of Pharmacology and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Michelle I. Lin
- From the ‡Department of Pharmacology and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Ji Y. Lee
- ‖W. M. Keck Foundation Biotechnology Resource Laboratory, Biostatistics Resources and
| | - Hongyu Zhao
- ‖W. M. Keck Foundation Biotechnology Resource Laboratory, Biostatistics Resources and
| | - Ruiyan Luo
- ‖W. M. Keck Foundation Biotechnology Resource Laboratory, Biostatistics Resources and
| | - Christopher Colangelo
- **W. M. Keck Foundation Biotechnology Resource Laboratory, Mass Spectrometry Resources, Yale University, New Haven, Connecticut 06510
| | - William C. Sessa
- From the ‡Department of Pharmacology and Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
34
|
Lin AEJ, Guttman JA. Hijacking the endocytic machinery by microbial pathogens. PROTOPLASMA 2010; 244:75-90. [PMID: 20574860 DOI: 10.1007/s00709-010-0164-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 05/19/2010] [Indexed: 05/24/2023]
Abstract
Understanding the mechanisms that microbes exploit to invade host cells and cause disease is crucial if we are to eliminate their threat. Although pathogens use a variety of microbial factors to trigger entry into non-phagocytic cells, their targeting of the host cell process of endocytosis has emerged as a common theme. To accomplish this, microbes often rewire the normal course of particle internalization, frequently usurping theoretical maximal sizes to permit entry and reconfiguring molecular components that were once thought to be required for vesicle formation. Here, we discuss recent advances in our understanding of how toxins, viruses, bacteria, and fungi manipulate the host cell endocytic machinery to generate diseases. Additionally, we will reveal the advantages of using these organisms to expand our general knowledge of endocytic mechanisms in eukaryotic cells.
Collapse
Affiliation(s)
- Ann En-Ju Lin
- Department of Biological Sciences, Shrum Science Centre, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | | |
Collapse
|
35
|
Pojoga LH, Romero JR, Yao TM, Loutraris P, Ricchiuti V, Coutinho P, Guo C, Lapointe N, Stone JR, Adler GK, Williams GH. Caveolin-1 ablation reduces the adverse cardiovascular effects of N-omega-nitro-L-arginine methyl ester and angiotensin II. Endocrinology 2010; 151:1236-46. [PMID: 20097717 PMCID: PMC2840694 DOI: 10.1210/en.2009-0514] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Caveolae are the major cellular membrane structure through which extracellular mediators transmit information to intracellular signaling pathways. In vascular tissue (but not ventricular myocardium), caveolin-1 (cav-1) is the main component of caveolae; cav-1 modulates enzymes and receptors, such as the endothelial nitric oxide synthase and the angiotensin II (AngII) type 1 receptor. Evidence suggests that AngII and aldosterone (ALDO) are important mediators of ventricular injury. We have described a model of biventricular damage in rodents that relies on treatment with N-omega-nitro-l-arginine methyl ester (L-NAME (nitric oxide synthase inhibitor)) and AngII. This damage initiated at the vascular level and was observed only in the presence of ALDO and an activated mineralocorticoid receptor (MR). We hypothesize that cav-1 modulates the adverse cardiac effects mediated by ALDO in this animal model. To test this hypothesis, we assessed the ventricular damage and measures of inflammation, in wild-type (WT) and cav-1 knockout (KO) mice randomized to either placebo or L-NAME/AngII treatment. Despite displaying cardiac hypertrophy at baseline and higher blood pressure responses to L-NAME/AngII, cav-1 KO mice displayed, as compared with WT, decreased treatment-induced biventricular damage as well as decreased transcript levels of the proinflammatory marker plasminogen activator inhibitor-1. Additionally, L-NAME/AngII induced an increase in cardiac MR levels in WT but not cav-1-ablated mice. Moreover and despite similar circulating ALDO levels in both genotypes, the myocardial damage (as determined histologically and by plasminogen activator inhibitor-1 mRNA levels) was less sensitive to ALDO levels in cav-1 KO vs. WT mice, consistent with decreased MR signaling in the cav-1 KO. Thus, we conclude that the L-NAME/AngII-induced biventricular damage is mediated by a mechanism partially dependent on cav-1 and signaling via MR/ALDO.
Collapse
Affiliation(s)
- Luminita H Pojoga
- Brigham and Women's Hospital/Harvard Medical School, Department of Endocrinology, Diabetes, and Hypertension, 221 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lajoie P, Nabi IR. Lipid Rafts, Caveolae, and Their Endocytosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 282:135-63. [DOI: 10.1016/s1937-6448(10)82003-9] [Citation(s) in RCA: 266] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
37
|
Kozera L, White E, Calaghan S. Caveolae act as membrane reserves which limit mechanosensitive I(Cl,swell) channel activation during swelling in the rat ventricular myocyte. PLoS One 2009; 4:e8312. [PMID: 20011535 PMCID: PMC2788708 DOI: 10.1371/journal.pone.0008312] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 11/14/2009] [Indexed: 12/22/2022] Open
Abstract
Background Many ion channels are preferentially located in caveolae where compartmentalisation/scaffolding with signal transduction components regulates their activity. Channels that are mechanosensitive may be additionally dependent on caveolar control of the mechanical state of the membrane. Here we test which mechanism underlies caveolar-regulation of the mechanosensitive ICl,swell channel in the adult cardiac myocyte. Methodology/Principal Findings Rat ventricular myocytes were exposed to solution of 0.02 tonicity (T; until lysis), 0.64T for 10–15 min (swelling), and/or methyl-β-cyclodextrin (MBCD; to disrupt caveolae). MBCD and 0.64T swelling reduced the number of caveolae visualised by electron microscopy by 75 and 50% respectively. MBCD stimulated translocation of caveolin 3 from caveolae-enriched buoyant membrane fractions, but both caveolin 1 and 3 remained in buoyant fractions after swelling. ICl,swell inhibition in control cells decreased time to half-maximal volume (t0.5,vol; 0.64T), consistent with a role for ICl,swell in volume regulation. MBCD-treated cells showed reduced time to lysis (0.02T) and t0.5,vol (0.64T) compared with controls. The negative inotropic response to swelling (an index of ICl,swell activation) was enhanced by MBCD. Conclusions/Significance These data show that disrupting caveolae removes essential membrane reserves, which speeds swelling in hyposmotic conditions, and thereby promotes activation of ICl,swell. They illustrate a general principle whereby caveolae as a membrane reserve limit increases in membrane tension during stretch/swelling thereby restricting mechanosensitive channel activation.
Collapse
Affiliation(s)
- Lukasz Kozera
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, United Kingdom
| | - Ed White
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, United Kingdom
| | - Sarah Calaghan
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Hezel M, de Groat WC, Galbiati F. Caveolin-3 promotes nicotinic acetylcholine receptor clustering and regulates neuromuscular junction activity. Mol Biol Cell 2009; 21:302-10. [PMID: 19940021 PMCID: PMC2808226 DOI: 10.1091/mbc.e09-05-0381] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This study identifies caveolin-3 as a key component of the signaling machinery that regulates clustering of the nicotinic acetylcholine receptor and controls neuromuscular junction function. The molecular mechanisms that regulate the organization and activity of the neuromuscular junction remain to be fully identified. Caveolae are invaginations of the plasma membrane. Caveolin-3 is the structural protein component of caveolae in muscle cells. We show that caveolin-3 is expressed at the neuromuscular junction, that it associates with the nicotinic acetylcholine receptor (nAChR), and that a lack of caveolin-3 inhibits clustering of the nAChR in myotubes. At the molecular level, we demonstrate that caveolin-3 is a novel muscle-specific kinase (MuSK) binding protein and that altered nAChR clustering in caveolin-3–lacking myotubes results from inhibition of agrin-induced phosphorylation/activation of MuSK and activation of Rac-1. Functional studies in caveolin-3 null mice show abnormal neuromuscular junction activity that is consistent with altered nAChR localization at the sarcolemma. Together, these data identify caveolin-3 as a critical component of the signaling machinery that drives nicotinic acetylcholine receptor clustering and controls neuromuscular junction function.
Collapse
Affiliation(s)
- Michael Hezel
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
39
|
Zaas DW, Swan ZD, Brown BJ, Li G, Randell SH, Degan S, Sunday ME, Wright JR, Abraham SN. Counteracting signaling activities in lipid rafts associated with the invasion of lung epithelial cells by Pseudomonas aeruginosa. J Biol Chem 2009; 284:9955-64. [PMID: 19211560 DOI: 10.1074/jbc.m808629200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa has the capacity to invade lung epithelial cells by co-opting the intrinsic endocytic properties of lipid rafts, which are rich in cholesterol, sphingolipids, and proteins, such as caveolin-1 and -2. We compared intratracheal Pseudomonas infection in wild type and caveolin-deficient mice to investigate the role of caveolin proteins in the pathogenesis of Pseudomonas pneumonia. Unlike wild type mice, which succumb to pneumonia, caveolin-deficient mice are resistant to Pseudomonas. We observed that Pseudomonas invasion of lung epithelial cells is dependent on caveolin-2 but not caveolin-1. Phosphorylation of caveolin-2 by Src family kinases is an essential event for Pseudomonas invasion. Our studies also reveal the existence of a distinct signaling mechanism in lung epithelial cells mediated by COOH-terminal Src kinase (Csk) that negatively regulates Pseudomonas invasion. Csk migrates to lipid raft domains, where it decreases phosphorylation of caveolin-2 by inactivating c-Src. Whereas Pseudomonas co-opts the endocytic properties of caveolin-2 for invasion, there also exists in these cells an intrinsic Csk-dependent cellular defense mechanism aimed at impairing this activity. The success of Pseudomonas in co-opting lipid raft-mediated endocytosis to invade lung epithelial cells may depend on the relative strengths of these counteracting signaling activities.
Collapse
Affiliation(s)
- David W Zaas
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Balijepalli RC, Kamp TJ. Caveolae, ion channels and cardiac arrhythmias. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2009; 98:149-60. [PMID: 19351512 DOI: 10.1016/j.pbiomolbio.2009.01.012] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Caveolae are specialized membrane microdomains enriched in cholesterol and sphingolipids which are present in multiple cell types including cardiomyocytes. Along with the essential scaffolding protein caveolin-3, a number of different ion channels and transporters have been localized to caveolae in cardiac myocytes including L-type Ca2+ channels (Ca(v)1.2), Na+ channels (Na(v)1.5), pacemaker channels (HCN4), Na+/Ca2+ exchanger (NCX1) and others. Closely associated with these channels are specific macromolecular signaling complexes that provide highly localized regulation of the channels. Mutations in the caveolin-3 gene (CAV3) have been linked with the congenital long QT syndrome (LQT9), and mutations in caveolar-localized ion channels may contribute to other inherited arrhythmias. Changes in the caveolar microdomain in acquired heart disease may also lead to dysregulation and dysfunction of ion channels, altering the risk of arrhythmias in conditions such as heart failure. This review highlights the existing evidence identifying and characterizing ion channels localized to caveolae in cardiomyocytes and their role in arrhythmogenesis.
Collapse
Affiliation(s)
- Ravi C Balijepalli
- Department of Medicine, Cellular and Molecular Arrhythmia Research Program, University of Wisconsin, Madison, WI 53792, USA
| | | |
Collapse
|
41
|
Chapter 4 The Biology of Caveolae. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 273:117-62. [DOI: 10.1016/s1937-6448(08)01804-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
42
|
Src family kinases as mediators of endothelial permeability: effects on inflammation and metastasis. Cell Tissue Res 2008; 335:249-59. [PMID: 18815812 DOI: 10.1007/s00441-008-0682-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 08/19/2008] [Indexed: 01/09/2023]
Abstract
Src family kinases (SFKs) are signaling enzymes that have long been recognized to regulate critical cellular processes such as proliferation, survival, migration, and metastasis. Recently, considerable work has elucidated mechanisms by which SFKs regulate normal and pathologic processes in vascular biology, including endothelial cell proliferation and permeability. Further, when inappropriately activated, SFKs promote pathologic inflammatory processes and tumor metastasis, in part through their effects on the regulation of endothelial monolayer permeability. In this review, we discuss the roles of aberrantly activated SFKs in mediating endothelial permeability in the context of inflammatory states and tumor cell metastasis. We further summarize recent efforts to translate Src-specific inhibitors into therapy for systemic inflammatory conditions and numerous solid organ cancers.
Collapse
|
43
|
Sverdlov M, Shajahan AN, Minshall RD. Tyrosine phosphorylation-dependence of caveolae-mediated endocytosis. J Cell Mol Med 2008; 11:1239-50. [PMID: 18205698 PMCID: PMC4401290 DOI: 10.1111/j.1582-4934.2007.00127.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Caveolae are flask-shaped plasma membrane invaginations that mediate endocytosis and transcytosis of plasma macromolecules, such as albumin, insulin and low-density lipoprotein (LDL), as well as certain viruses, bacteria and bacterial toxins. Caveolae-mediated transcytosis of macromolecules is critical for maintaining vascular homeostasis by regulating the oncotic pressure gradient and tissue delivery of drugs, vitamins, lipids and ions. Entrapment of cargo within caveolae induces activation of signalling cascades leading to caveolae fission and internalization. Activation of Src tyrosine kinase is an early and essential step that triggers detachment of loaded caveolae from the plasma membrane. In this review, we examine how Src-mediated phosphorylation regulates caveolae-mediated transport by orchestrating the localization and activity of essential proteins of the endocytic machinery to regulate caveolae formation and fission.
Collapse
Affiliation(s)
- Maria Sverdlov
- Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois, College of Medicine at Chicago, Chicago, IL 60612, USA
| | | | | |
Collapse
|
44
|
de Diesbach P, Medts T, Carpentier S, D'Auria L, Van Der Smissen P, Platek A, Mettlen M, Caplanusi A, van den Hove MF, Tyteca D, Courtoy PJ. Differential subcellular membrane recruitment of Src may specify its downstream signalling. Exp Cell Res 2008; 314:1465-79. [DOI: 10.1016/j.yexcr.2008.01.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 12/21/2007] [Accepted: 01/14/2008] [Indexed: 12/22/2022]
|
45
|
Abstract
Studies on the structure and function of caveolae have revealed how this versatile subcellular organelle can influence numerous signalling pathways. This brief review will discuss a few of the key features of caveolae as it relates to signalling and disease processes.
Collapse
Affiliation(s)
- Candice M Thomas
- Department of Pediatrics and the Kentucky Pediatric Research Institute, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
46
|
Boulware MI, Kordasiewicz H, Mermelstein PG. Caveolin proteins are essential for distinct effects of membrane estrogen receptors in neurons. J Neurosci 2007; 27:9941-50. [PMID: 17855608 PMCID: PMC6672640 DOI: 10.1523/jneurosci.1647-07.2007] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
It has become widely accepted that along with its ability to directly regulate gene expression, estradiol also influences cell signaling and brain function via rapid membrane-initiated events. Many of these novel signaling processes are dependent on estrogen receptors (ERs) localized to the neuronal membrane. However, the mechanism(s) by which ERs are able to trigger cell signaling when targeted to the neuronal membrane surface has yet to be determined. In hippocampal neurons, we find that caveolin proteins are essential for the regulation of CREB (cAMP response element-binding protein) phosphorylation after estradiol activation of metabotropic glutamate receptor (mGluR) signaling. Furthermore, caveolin-1 (CAV1) and CAV3 differentially regulate the ability of estradiol to activate two discrete signaling pathways. ER alpha activation of mGluR1a is dependent on CAV1, whereas CAV3 is necessary for ER alpha and ER beta activation of mGluR2/3. These results are consistent with previous reports in non-neuronal cells, implicating the importance of caveolin proteins in rapid estrogen signaling. In addition, the functional isolation of distinct estrogen-sensitive signaling pathways by different caveolin proteins suggests novel mechanisms through which the membrane-initiated effects of estradiol are orchestrated.
Collapse
Affiliation(s)
- Marissa I. Boulware
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Holly Kordasiewicz
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
47
|
Mir KD, Parr RD, Schroeder F, Ball JM. Rotavirus NSP4 interacts with both the amino- and carboxyl-termini of caveolin-1. Virus Res 2007; 126:106-15. [PMID: 17379346 PMCID: PMC1978065 DOI: 10.1016/j.virusres.2007.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Revised: 01/29/2007] [Accepted: 02/05/2007] [Indexed: 11/25/2022]
Abstract
Rotavirus NSP4 plays multiple roles in viral pathogenesis, morphogenesis and replication. We previously reported a direct interaction between full-length NSP4 and the enterotoxic peptide composed of NSP4 residues 114-135 with full-length caveolin-1, the structural protein of caveolae. Caveolin-1 forms a hairpin loop in the cytoplasmic leaflet of plasma membrane caveolae. This unique orientation results in both termini of caveolin-1 exposed to the cytoplasm. The goal of this study was to map the caveolin-1 residues that interact with NSP4 to obtain a more complete picture of this binding event. Utilizing reverse yeast two-hybrid analyses and direct peptide binding assays, the NSP4 binding site was localized to caveolin-1 residues 2-22 and 161-178, at the amino- and carboxyl-termini, respectively. However, NSP4 binding to one of the termini was sufficient for the interaction.
Collapse
Affiliation(s)
- Kiran D. Mir
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, Department of Pathobiology, College Station, TX 77843
| | - Rebecca D. Parr
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, Department of Pathobiology, College Station, TX 77843
| | - Friedhelm Schroeder
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, Department of Physiology and Pharmacology, College Station, TX 77843
| | - Judith M. Ball
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, Department of Pathobiology, College Station, TX 77843
- *corresponding author Phone: (979) 845-9710, Fax: (979) 845-9231, , Texas A&M University, TVMC, TAMU 4467, College Station, TX 77843
| |
Collapse
|
48
|
Fang PK, Solomon KR, Zhuang L, Qi M, McKee M, Freeman MR, Yelick PC. Caveolin-1alpha and -1beta perform nonredundant roles in early vertebrate development. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 169:2209-22. [PMID: 17148682 PMCID: PMC1762473 DOI: 10.2353/ajpath.2006.060562] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Caveolins are integral membrane proteins that localize predominantly to lipid rafts, where they oligomerize to form flask-shaped organelles termed caveolae and play important roles in membrane trafficking, signal transduction, and other cellular processes. To investigate potential roles for caveolin-1 (cav-1) in development, cav-1alpha and -1beta cDNAs were functionally characterized in the zebrafish. Cav-1alpha and -1beta mRNAs exhibited overlapping but distinct expression patterns throughout embryogenesis. Targeted depletion of either Cav-1 isoform, using antisense morpholino oligomers, resulted in a substantial loss of caveolae and dramatic neural, eye, and somite defects by 12 hours after fertilization, the time at which mRNA levels of both isoforms substantially increased in wild-type animals. Morphant phenotypes were rescued by injection of homotypic (cav-1alpha/cav-1alpha) but not heterotypic (cav-1alpha/cav-1beta) zebrafish and human cav-1 cRNAs, revealing nonredundant and evolutionarily conserved functions for the individual Cav-1 isoforms. Mutation of a known Cav-1 phosphorylation site unique to Cav-1alpha (Y14-->F) resulted in a failure to rescue the cav-1alpha morphant phenotype, verifying an essential role for Cav-1alpha specifically and implicating this residue in early developmental functions. Cav-1alpha and -1beta morphants also exhibited disruption in the actin cytoskeleton. These results support important and previously unanticipated roles for the Caveolin-1 isoforms in vertebrate organogenesis.
Collapse
Affiliation(s)
- Ping-Ke Fang
- Urological Diseases Research Center, Department of Orthopaedic Surgery, Children's Hospital Boston, Harvard Medical School, Enders Research Laboratories, Suite 1161, 300 Longwood Ave., Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Eldstrom J, Van Wagoner DR, Moore ED, Fedida D. Localization of Kv1.5 channels in rat and canine myocyte sarcolemma. FEBS Lett 2006; 580:6039-46. [PMID: 17054951 DOI: 10.1016/j.febslet.2006.09.069] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 09/27/2006] [Indexed: 10/24/2022]
Abstract
Voltage-gated potassium (Kv) channel subtypes localize to the plasma membrane of a number of cell types, and the sarcolemma in myocytes. Because many signaling molecules concentrate in subdomains of the plasma membrane, the localization of Kv channels to these sites may have important implications for channel function and regulation. In this study, the association of the voltage-gated potassium channel Kv1.5 with a specific subtype of lipid rafts, caveolae, in rat and canine cardiac myocytes has been investigated. Interactions between caveolin-3 and beta-dystroglycan or eNOS, as well as between Kv1.5 and alpha-actinin were readily detected in co-immunoprecipitation experiments, whereas no association between Kv1.5 and caveolin-3 was evident. Wide-field microscopy and deconvolution techniques revealed that the percent co-localization of Kv1.5 with caveolin-3 was extremely low in atrial myocytes from rat and canine hearts (8+/-1% and 12.2+/-2%, respectively), and limited in ventricular myocytes (11+/-4% and 20+/-3% in rat and canine, respectively). Immunoelectron microscopic imaging of rat atrial and ventricular tissues showed that Kv1.5 and caveolin-3 labeling generally did not overlap. In HEK293 cells stably expressing the channel, Kv1.5 did not target to the low buoyant density raft fraction along with flotillin but instead fractionated along with the non-raft associated transferrin receptor. Taken together, these results suggest that Kv1.5 is not present in caveolae of rat and canine heart.
Collapse
Affiliation(s)
- Jodene Eldstrom
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3.
| | | | | | | |
Collapse
|
50
|
Halayko AJ, Stelmack GL. The association of caveolae, actin, and the dystrophin-glycoprotein complex: a role in smooth muscle phenotype and function? Can J Physiol Pharmacol 2006; 83:877-91. [PMID: 16333360 DOI: 10.1139/y05-107] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Smooth muscle cells exhibit phenotypic and mechanical plasticity. During maturation, signalling pathways controlling actin dynamics modulate contractile apparatus-associated gene transcription and contractile apparatus remodelling resulting from length change. Differentiated myocytes accumulate abundant caveolae that evolve from the structural association of lipid rafts with caveolin-1, a protein with domains that confer unique functional properties. Caveolae and caveolin-1 modulate and participate in receptor-mediated signalling, and thus contribute to functional diversity of phenotypically similar myocytes. In mature smooth muscle, caveolae are partitioned into discrete linear domains aligned with structural proteins that tether actin to the extracellular matrix. Caveolin-1 binds with beta-dystroglycan, a subunit of the dystrophin glycoprotein complex (DGC), and with filamin, an actin binding protein that organizes cortical actin, to which integrins and focal adhesion complexes are anchored. The DGC is linked to the actin cytoskeleton by a dystrophin subunit and is a receptor for extracellular laminin. Thus, caveolae and caveolin-associated signalling proteins and receptors are linked via structural proteins to a dynamic filamentous actin network. Despite development of transgenic models to investigate caveolins and membrane-associated actin-linking proteins in skeletal and cardiac muscle function, only superficial understanding of this association in smooth muscle phenotype and function has emerged.
Collapse
Affiliation(s)
- Andrew J Halayko
- Department of Physiology, University of Manitoba, Winnipeg, Canada.
| | | |
Collapse
|