1
|
Postrigan AE, Babushkina NP, Svintsova LI, Plotnikova IV, Skryabin NA. Clinical and Genetic Characteristics of Congenital Long QT Syndrome. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422100064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
2
|
Kaushal JB, Batra SK, Rachagani S. Hedgehog signaling and its molecular perspective with cholesterol: a comprehensive review. Cell Mol Life Sci 2022; 79:266. [PMID: 35486193 PMCID: PMC9990174 DOI: 10.1007/s00018-022-04233-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/18/2022] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Hedgehog (Hh) signaling is evolutionarily conserved and plays an instructional role in embryonic morphogenesis, organogenesis in various animals, and the central nervous system organization. Multiple feedback mechanisms dynamically regulate this pathway in a spatiotemporal and context-dependent manner to confer differential patterns in cell fate determination. Hh signaling is complex due to canonical and non-canonical mechanisms coordinating cell-cell communication. In addition, studies have demonstrated a regulatory framework of Hh signaling and shown that cholesterol is vital for Hh ligand biogenesis, signal generation, and transduction from the cell surface to intracellular space. Studies have shown the importance of a specific cholesterol pool, termed accessible cholesterol, which serves as a second messenger, conveying signals between smoothened (Smo) and patched 1 (Ptch1) across the plasma and ciliary membranes. Remarkably, recent high-resolution structural and molecular studies shed new light on the interplay between Hh signaling and cholesterol in membrane biology. These studies elucidated novel mechanistic insight into the release and dispersal of cholesterol-anchored Hh and the basis of Hh recognition by Ptch1. Additionally, the putative model of Smo activation by cholesterol binding and/or modification and Ptch1 antagonization of Smo has been explicated. However, the coupling mechanism of Hh signaling and cholesterol offered a new regulatory principle in cell biology: how effector molecules of the Hh signal network react to and remodel cholesterol accessibility in the membrane and selectively activate Hh signaling proteins thereof. Recognizing the biological importance of cholesterol in Hh signaling activation and transduction opens the door for translational research to develop novel therapeutic strategies. This review looks in-depth at canonical and non-canonical Hh signaling and the distinct proposed model of cholesterol-mediated regulation of Hh signaling components, facilitating a more sophisticated understanding of the Hh signal network and cholesterol biology.
Collapse
Affiliation(s)
- Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
3
|
Sych T, Levental KR, Sezgin E. Lipid–Protein Interactions in Plasma Membrane Organization and Function. Annu Rev Biophys 2022; 51:135-156. [DOI: 10.1146/annurev-biophys-090721-072718] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lipid–protein interactions in cells are involved in various biological processes, including metabolism, trafficking, signaling, host–pathogen interactions, and transmembrane transport. At the plasma membrane, lipid–protein interactions play major roles in membrane organization and function. Several membrane proteins have motifs for specific lipid binding, which modulate protein conformation and consequent function. In addition to such specific lipid–protein interactions, protein function can be regulated by the dynamic, collective behavior of lipids in membranes. Emerging analytical, biochemical, and computational technologies allow us to study the influence of specific lipid–protein interactions, as well as the collective behavior of membranes on protein function. In this article, we review the recent literature on lipid–protein interactions with a specific focus on the current state-of-the-art technologies that enable novel insights into these interactions. Expected final online publication date for the Annual Review of Biophysics, Volume 51 is May 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Taras Sych
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden;,
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden;,
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Montana V, Flint D, Waagepetersen HS, Schousboe A, Parpura V. Two Metabolic Fuels, Glucose and Lactate, Differentially Modulate Exocytotic Glutamate Release from Cultured Astrocytes. Neurochem Res 2021; 46:2551-2579. [PMID: 34057673 PMCID: PMC9015689 DOI: 10.1007/s11064-021-03340-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Astrocytes have a prominent role in metabolic homeostasis of the brain and can signal to adjacent neurons by releasing glutamate via a process of regulated exocytosis. Astrocytes synthesize glutamate de novo owing to the pyruvate entry to the citric/tricarboxylic acid cycle via pyruvate carboxylase, an astrocyte specific enzyme. Pyruvate can be sourced from two metabolic fuels, glucose and lactate. Thus, we investigated the role of these energy/carbon sources in exocytotic glutamate release from astrocytes. Purified astrocyte cultures were acutely incubated (1 h) in glucose and/or lactate-containing media. Astrocytes were mechanically stimulated, a procedure known to increase intracellular Ca2+ levels and cause exocytotic glutamate release, the dynamics of which were monitored using single cell fluorescence microscopy. Our data indicate that glucose, either taken-up from the extracellular space or mobilized from the intracellular glycogen storage, sustained glutamate release, while the availability of lactate significantly reduced the release of glutamate from astrocytes. Based on further pharmacological manipulation during imaging along with tandem mass spectrometry (proteomics) analysis, lactate alone, but not in the hybrid fuel, caused metabolic changes consistent with an increased synthesis of fatty acids. Proteomics analysis further unveiled complex changes in protein profiles, which were condition-dependent and generally included changes in levels of cytoskeletal proteins, proteins of secretory organelle/vesicle traffic and recycling at the plasma membrane in aglycemic, lactate or hybrid-fueled astrocytes. These findings support the notion that the availability of energy sources and metabolic milieu play a significant role in gliotransmission.
Collapse
Affiliation(s)
- Vedrana Montana
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Daniel Flint
- Luxumbra Strategic Research, LLC, Arlington, VA, USA
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
5
|
Liu Y, Wang M, Wang D, Fay WP, Korthuis RJ, Sowa G. Elevated postischemic tissue injury and leukocyte-endothelial adhesive interactions in mice with global deficiency in caveolin-2: role of PAI-1. Am J Physiol Heart Circ Physiol 2021; 320:H1185-H1198. [PMID: 33416452 PMCID: PMC8362680 DOI: 10.1152/ajpheart.00682.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 01/13/2023]
Abstract
Ischemia/reperfusion (I/R)-induced rapid inflammation involving activation of leukocyte-endothelial adhesive interactions and leukocyte infiltration into tissues is a major contributor to postischemic tissue injury. However, the molecular mediators involved in this pathological process are not fully known. We have previously reported that caveolin-2 (Cav-2), a protein component of plasma membrane caveolae, regulated leukocyte infiltration in mouse lung carcinoma tumors. The goal of the current study was to examine if Cav-2 plays a role in I/R injury and associated acute leukocyte-mediated inflammation. Using a mouse small intestinal I/R model, we demonstrated that I/R downregulates Cav-2 protein levels in the small bowel. Further study using Cav-2-deficient mice revealed aggravated postischemic tissue injury determined by scoring of villi length in H&E-stained tissue sections, which correlated with increased numbers of MPO-positive tissue-infiltrating leukocytes determined by IHC staining. Intravital microscopic analysis of upstream events relative to leukocyte transmigration and tissue infiltration revealed that leukocyte-endothelial cell adhesive interactions in postcapillary venules, namely leukocyte rolling and adhesion were also enhanced in Cav-2-deficient mice. Mechanistically, Cav-2 deficiency increased plasminogen activator inhibitor-1 (PAI-1) protein levels in the intestinal tissue and a pharmacological inhibition of PAI-1 had overall greater inhibitory effect on both aggravated I/R tissue injury and enhanced leukocyte-endothelial interactions in postcapillary venules in Cav-2-deficient mice. In conclusion, our data suggest that Cav-2 protein alleviates tissue injury in response to I/R by dampening PAI-1 protein levels and thereby reducing leukocyte-endothelial adhesive interactions.NEW & NOTEWORTHY The role of caveolin-2 in regulating ischemia/reperfusion (I/R) tissue injury and the mechanisms underlying its effects are unknown. This study uses caveolin-2-deficient mouse and small intestinal I/R injury models to examine the role of caveolin-2 in the leukocyte-dependent reperfusion injury. We demonstrate for the first time that caveolin-2 plays a protective role from the I/R-induced leukocyte-dependent reperfusion injury by reducing PAI-1 protein levels in intestinal tissue and leukocyte-endothelial adhesive interactions in postcapillary venules.
Collapse
Affiliation(s)
- Yajun Liu
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Meifang Wang
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Derek Wang
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - William P Fay
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Medicine, University of Missouri, Columbia, Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Grzegorz Sowa
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
6
|
Lolo FN, Jiménez-Jiménez V, Sánchez-Álvarez M, Del Pozo MÁ. Tumor-stroma biomechanical crosstalk: a perspective on the role of caveolin-1 in tumor progression. Cancer Metastasis Rev 2021; 39:485-503. [PMID: 32514892 DOI: 10.1007/s10555-020-09900-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor stiffening is a hallmark of malignancy that actively drives tumor progression and aggressiveness. Recent research has shed light onto several molecular underpinnings of this biomechanical process, which has a reciprocal crosstalk between tumor cells, stromal fibroblasts, and extracellular matrix remodeling at its core. This dynamic communication shapes the tumor microenvironment; significantly determines disease features including therapeutic resistance, relapse, or metastasis; and potentially holds the key for novel antitumor strategies. Caveolae and their components emerge as integrators of different aspects of cell function, mechanotransduction, and ECM-cell interaction. Here, we review our current knowledge on the several pivotal roles of the essential caveolar component caveolin-1 in this multidirectional biomechanical crosstalk and highlight standing questions in the field.
Collapse
Affiliation(s)
- Fidel Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel Del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
7
|
Zhang C, Wu Q, Huang H, Chen X, Huang T, Li W, Zhang J, Liu Y. Caveolin-1 upregulates Fut8 expression by activating the Wnt/β-catenin pathway to enhance HCC cell proliferative and invasive ability. Cell Biol Int 2020; 44:2202-2212. [PMID: 32710651 DOI: 10.1002/cbin.11426] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/12/2020] [Accepted: 07/23/2020] [Indexed: 01/09/2023]
Abstract
Caveolin-1 (Cav-1), a critical structural protein of caveolae, plays an oncogenic role by participating in abnormal protein glycosylation in hepatocellular carcinoma (HCC). However, the mechanism by which Cav-1 regulates glycosylation and glycosyltransferase expression has yet to be fully defined. Here, we show that Cav-1 promotes the expression of α-1,6-fucosyltransferase (Fut8), which catalyzes the transfer of GDP-fucose to the core structure of the N-sugar chain. In this study, we show that the mouse HCC cell line, Hepa1-6, which has low Fut8 transcriptional and protein levels, also lacks Cav-1 expression, whereas the mouse HCC cell line, Hca-F, has strong Fut8 expression and high transcriptional and protein levels of Cav-1. Subsequently, Cav-1 overexpression in Hepa1-6 was found to activate Wnt/β-catenin signaling, which leads to downstream binding of the T cell factor/lymphoid enhancer factor to the Fut8 promoter region for activation of its transcription. In contrast, knockdown of Cav-1 expression in Hca-F caused the Wnt/β-catenin pathway to be significantly inhibited, which attenuates the expression of Fut8. We further show that Cav-1-induced upregulation of Fut8 expression enhanced proliferation and invasion by mouse HCC cells in vitro. Our current findings provide molecular evidence that Cav-1 plays an important role in regulating glycosyltransferase expression and may participate in abnormal glycosylation, which mediates the proliferation and invasion of HCC.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Qiong Wu
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Huang Huang
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Xixi Chen
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Tianmiao Huang
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Wenli Li
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China.,School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Jianing Zhang
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Yubo Liu
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| |
Collapse
|
8
|
Eser Ocak P, Ocak U, Sherchan P, Gamdzyk M, Tang J, Zhang JH. Overexpression of Mfsd2a attenuates blood brain barrier dysfunction via Cav-1/Keap-1/Nrf-2/HO-1 pathway in a rat model of surgical brain injury. Exp Neurol 2020; 326:113203. [PMID: 31954682 PMCID: PMC7038791 DOI: 10.1016/j.expneurol.2020.113203] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Disruption of the blood brain barrier (BBB) and subsequent cerebral edema formation is one of the major adverse effects of brain surgery, leading to postoperative neurological dysfunction. Recently, Mfsd2a has been shown to have a crucial role for the maintenance of BBB functions. In this study, we aimed to evaluate the role of Mfsd2a on BBB disruption following surgical brain injury (SBI) in rats. MATERIALS AND METHODS Rats were subjected to SBI by partial resection of the right frontal lobe. To evaluate the effect of Mfsd2a on BBB permeability and neurobehavior outcome following SBI, Mfsd2a was either overexpressed or downregulated in the brain by administering Mfsd2a CRISPR activation or knockout plasmids, respectively. The potential mechanism of Mfsd2a-mediated BBB protection through the cav-1/Nrf-2/HO-1 signaling pathway was evaluated. RESULTS Mfsd2a levels were significantly decreased while cav-1, Nrf-2 and HO-1 levels were increased in the right frontal perisurgical area following SBI. When overexpressed, Mfsd2a attenuated brain edema and abolished neurologic impairment caused by SBI while downregulation of Mfsd2a expression further deteriorated BBB functions and worsened neurologic performance following SBI. The beneficial effect of Mfsd2a overexpression on BBB functions was associated with diminished expression of cav-1, increased Keap-1/Nrf-2 dissociation and further augmented levels of Nrf-2 and HO-1 in the right frontal perisurgical area, leading to enhanced levels of tight junction proteins following SBI. The BBB protective effect of Mfsd2a was blocked by selective inhibitors of Nrf-2 and HO-1. CONCLUSIONS Mfsd2a attenuates BBB disruption through cav-1/Nrf-2/HO-1 signaling pathway in rats subjected to experimental SBI.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA; Department of Neurosurgery, Uludag University School of Medicine, Bursa 16120, Turkey
| | - Umut Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA; Department of Emergency Medicine, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, Bursa 16310, Turkey; Department of Emergency Medicine, Bursa City Hospital, Bursa 16110, Turkey
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA; Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA; Department of Neurology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
9
|
Prosdocimi E, Checchetto V, Leanza L. Targeting the Mitochondrial Potassium Channel Kv1.3 to Kill Cancer Cells: Drugs, Strategies, and New Perspectives. SLAS DISCOVERY 2019; 24:882-892. [PMID: 31373829 DOI: 10.1177/2472555219864894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer is the consequence of aberrations in cell growth or cell death. In this scenario, mitochondria and ion channels play a critical role in regard to cell proliferation, malignant angiogenesis, migration, and metastasis. In this review, we focus on Kv1.3 and specifically on mitoKv1.3, which showed an aberrant expression in cancer cells compared with healthy tissues and which is involved in the apoptotic pathway. In recent years, mitoKv1.3 has become an oncological target since its pharmacological modulation has been demonstrated to reduce tumor growth and progression both in vitro and in vivo using preclinical mouse models of different types of tumors.
Collapse
Affiliation(s)
| | | | - Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Zhang Y, Fan W, Wu J, Dong J, Cui Z. Association of caveolin-1 protein expression with hepatocellular carcinoma: a meta-analysis and literature review. Cancer Manag Res 2019; 11:5113-5122. [PMID: 31239768 PMCID: PMC6553953 DOI: 10.2147/cmar.s194033] [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] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/07/2019] [Indexed: 01/08/2023] Open
Abstract
Background: Aberrant expression of caveolin-1 (CAV-1) is involved in the pathogenesis of hepatocellular carcinoma (HCC); however, the results have been inconsistent due to the small size of sample in the individual study. Methods: We performed a meta-analysis and evaluated the association of CAV-1 protein overexpression and clinicopathological significance by using Review Manager 5.2. Pooled ORs and HR with corresponding CIs were calculated. Results: Nine studies were included in the meta-analysis with 810 HCC and 172 cirrhosis patients. CAV-1 protein overexpression was correlated with the risk of cirrhosis; OR was 3.25, p=0.01. Furthermore, the rate of CAV-1 protein overexpression was significantly higher in HCC with cirrhosis than HCC without cirrhosis, suggesting that the CAV-1 protein overexpression likely initiated carcinogenesis in liver with cirrhosis and subsequently contributed to the progression of HCC. In addition, CAV-1 protein overexpression was strongly associated with poor differentiated HCC and invasion; ORs were 2.61 and 2.71, respectively. CAV-1 protein overexpression was strongly correlated with poor overall survival in patients with HCC; HR was 0.4, p=0.03. Conclusions: In summary, CAV-1 protein overexpression is at risk for liver cirrhosis and HCC derived from cirrhosis, and CAV-1 is also a promising prognostic predictor in HCC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng 475000, People's Republic of China
| | - Wenjuan Fan
- Medical Bioengineering Key Laboratory, Luohe Medical College, Luohe 462002, People's Republic of China
| | - Jiang Wu
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng 475000, People's Republic of China
| | - Jinglong Dong
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng 475000, People's Republic of China
| | - Zhanjun Cui
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, People's Republic of China
| |
Collapse
|
11
|
Nordzieke DE, Medraño-Fernandez I. The Plasma Membrane: A Platform for Intra- and Intercellular Redox Signaling. Antioxidants (Basel) 2018; 7:antiox7110168. [PMID: 30463362 PMCID: PMC6262572 DOI: 10.3390/antiox7110168] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/15/2018] [Accepted: 11/17/2018] [Indexed: 02/06/2023] Open
Abstract
Membranes are of outmost importance to allow for specific signal transduction due to their ability to localize, amplify, and direct signals. However, due to the double-edged nature of reactive oxygen species (ROS)—toxic at high concentrations but essential signal molecules—subcellular localization of ROS-producing systems to the plasma membrane has been traditionally regarded as a protective strategy to defend cells from unwanted side-effects. Nevertheless, specialized regions, such as lipid rafts and caveolae, house and regulate the activated/inhibited states of important ROS-producing systems and concentrate redox targets, demonstrating that plasma membrane functions may go beyond acting as a securing lipid barrier. This is nicely evinced by nicotinamide adenine dinucleotide phosphate (NADPH)-oxidases (NOX), enzymes whose primary function is to generate ROS and which have been shown to reside in specific lipid compartments. In addition, membrane-inserted bidirectional H2O2-transporters modulate their conductance precisely during the passage of the molecules through the lipid bilayer, ensuring time-scaled delivery of the signal. This review aims to summarize current evidence supporting the role of the plasma membrane as an organizing center that serves as a platform for redox signal transmission, particularly NOX-driven, providing specificity at the same time that limits undesirable oxidative damage in case of malfunction. As an example of malfunction, we explore several pathological situations in which an inflammatory component is present, such as inflammatory bowel disease and neurodegenerative disorders, to illustrate how dysregulation of plasma-membrane-localized redox signaling impacts normal cell physiology.
Collapse
Affiliation(s)
- Daniela E Nordzieke
- Institute of Microbiology and Genetics, Department of Genetics of Eukaryotic Microorganisms, Georg August University Göttingen, Grisebachstr. 8, D-37077 Göttingen, Germany.
| | - Iria Medraño-Fernandez
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
12
|
Beyond the Matrix: The Many Non-ECM Ligands for Integrins. Int J Mol Sci 2018; 19:ijms19020449. [PMID: 29393909 PMCID: PMC5855671 DOI: 10.3390/ijms19020449] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/21/2018] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
The traditional view of integrins portrays these highly conserved cell surface receptors as mediators of cellular attachment to the extracellular matrix (ECM), and to a lesser degree, as coordinators of leukocyte adhesion to the endothelium. These canonical activities are indispensable; however, there is also a wide variety of integrin functions mediated by non-ECM ligands that transcend the traditional roles of integrins. Some of these unorthodox roles involve cell-cell interactions and are engaged to support immune functions such as leukocyte transmigration, recognition of opsonization factors, and stimulation of neutrophil extracellular traps. Other cell-cell interactions mediated by integrins include hematopoietic stem cell and tumor cell homing to target tissues. Integrins also serve as cell-surface receptors for various growth factors, hormones, and small molecules. Interestingly, integrins have also been exploited by a wide variety of organisms including viruses and bacteria to support infectious activities such as cellular adhesion and/or cellular internalization. Additionally, the disruption of integrin function through the use of soluble integrin ligands is a common strategy adopted by several parasites in order to inhibit blood clotting during hematophagy, or by venomous snakes to kill prey. In this review, we strive to go beyond the matrix and summarize non-ECM ligands that interact with integrins in order to highlight these non-traditional functions of integrins.
Collapse
|
13
|
Identification of the caveolae/raft-mediated endocytosis as the primary entry pathway for aquareovirus. Virology 2018; 513:195-207. [DOI: 10.1016/j.virol.2017.09.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 12/11/2022]
|
14
|
Jacques D, Abdel-Karim Abdel-Malak N, Abou Abdallah N, Al-Khoury J, Bkaily G. Difference in the response to angiotensin II between left and right ventricular endocardial endothelial cells. Can J Physiol Pharmacol 2017; 95:1271-1282. [PMID: 28727938 DOI: 10.1139/cjpp-2017-0280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Previous studies focused on the right ventricular endocardial endothelial cells (EECRs) and showed that angiotensin II (Ang II) induced increase in cytosolic and nuclear calcium via AT1 receptor activation. In the present study, we verified whether the response of left EECs (EECLs) to Ang II is different than that of EECRs. Our results showed that the EC50 of the Ang II-induced increase of cytosolic and nuclear calcium in EECLs was 10× higher (around 2 × 10-13 mol/L) than in EECRs (around 8 × 10-12 mol/L). The densities of both AT1 and AT2 receptors were also higher in EECLs than those previously reported in EECRs. The effect of Ang II was mediated in both cell types via the activation of AT1 receptors. Treatment with Ang II induced a significant increase of cytosolic and nuclear AT1 receptors in EECRs, whereas the opposite was found in EECLs. In both cell types, there was a transient increase of cytosolic and nuclear AT2 receptors following the Ang II treatment. In conclusion, our results showed that both AT1 and AT2 receptors densities are higher in both EECLs compared to what was reported in EECRs. The higher density of AT1 receptors in EECLs compared to REECs may explain, in part, the higher sensitivity of EECLs to Ang II.
Collapse
Affiliation(s)
- Danielle Jacques
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Nelly Abdel-Karim Abdel-Malak
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Nadia Abou Abdallah
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Johny Al-Khoury
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Ghassan Bkaily
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
15
|
Deng YF, Huang YY, Lu WS, Huang YH, Xian J, Wei HQ, Huang Q. The Caveolin-3 P104L mutation of LGMD-1C leads to disordered glucose metabolism in muscle cells. Biochem Biophys Res Commun 2017; 486:218-223. [PMID: 28232187 DOI: 10.1016/j.bbrc.2017.02.072] [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: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 11/30/2022]
Abstract
Caveolin-3 (CAV3) is a muscle specific protein that plays an important role in maintaining muscle health and glucose homeostasis in vivo. A novel autosomal dominant form of LGMD-1C in humans is due to a P104L mutation within the coding sequence of the human CAV3 gene. The mechanism by which the LGMD-1C mutation leads to muscle weakness remains unknown. Our objective was to determine whether muscle weakness was related to the imbalance of glucose metabolism. We found that when the P104L mutation was transiently transfected into C2C12 cells, there was decreased glucose uptake and glycogen synthesis after insulin stimulation. Immunoblotting analysis showed that the P104L mutation resulted in decreased expression of CAV3, CAV1 and pAkt. Confocal immunomicroscopy indicated that the P104L mutation reduced CAV3 and GLUT4 in the cell membrane, which accumulated mainly near the nucleus. This work is the first report of an association between muscle weakness due to LGMD-1C and energy metabolism. The P104L mutation led to a decrease in C2C12 muscle glucose uptake and glycogen synthesis and may be involved in the pathogenesis of LGMD-1C.
Collapse
Affiliation(s)
- Yu Feng Deng
- School of Nursing, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Yi Yuan Huang
- School of Nursing, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Wen Sheng Lu
- Department of Endocrinology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yuan Heng Huang
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Xian
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Qiao Wei
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi, China
| | - Qin Huang
- Department of Physiology, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
16
|
Mahdy MM, El-Ekiaby NM, Hashish RM, Salah RA, Hanafi RS, El-Said Azzazy HM, Abdelaziz AI. miR-29a Promotes Lipid Droplet and Triglyceride Formation in HCV Infection by Inducing Expression of SREBP-1c and CAV1. J Clin Transl Hepatol 2016; 4:293-299. [PMID: 28097097 PMCID: PMC5225148 DOI: 10.14218/jcth.2016.00046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/21/2016] [Accepted: 12/07/2016] [Indexed: 01/18/2023] Open
Abstract
Aims: To examine the regulation of SREBP-1c and CAV1 by microRNA-29a (miR-29a) in cells infected with hepatitis C virus (HCV) in an attempt to control HCV-induced non-alcoholic fatty liver disease. Methods: In order to examine the manipulation of SREBP-1c and CAV1 by miR-29a, oleic acid (OA)-treated JFH-I-infected Huh-7 cells were used. OA was added 24 h post-transfection and gene expression was investigated by qRT-PCR at 48 h post treatment. The functional impact of the observed alteration in SREBP-1c and CAV1 expression was analyzed by examining lipid droplet (LD) and triglyceride (TG) content at 72 h post-OA treatment using light microscopy and spectrophotometry, respectively. Viral load was quantified by qRT-PCR at 72 h post-transfection. Results: OA treatment induced the expression of miR-29a and SREBP-1c, as compared to untreated cells. Forced miR-29a expression led to a significant up-regulation of SREBP-1c as well as CAV1 compared to mock untransfected cells. Ectopic expression of miR-29a resulted in a marked increase in LDs and their respective TGs, while miR-29a antagomirs decreased both the LD and TG content compared to mock untransfected cells. Moreover, forcing the expression of miR-29a in JFH-1 HCV-infected Huh-7 cells resulted in 53% reduction in viral titers compared to mock untransfected Huh-7 cells. Conclusion: Inducing miR-29a expression significantly induces SREBP-1c and CAV1 expression, thereby increasing LDs as well as their respective TGs. Nonetheless, forcing the expression of miR-29a resulted in reduction of HCV RNA levels in Huh-7 cells.
Collapse
Affiliation(s)
| | - Nada Magdy El-Ekiaby
- Department of Pharmacology and Toxicology, German University in Cairo, New Cairo City, Egypt
- School of Medicine, NewGiza University, Cairo, Egypt
| | - Rana Mahmoud Hashish
- Department of Pharmaceutical Biology, German University in Cairo, New Cairo City, Egypt
| | - Radwa Ayman Salah
- Department of Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Rasha Sayed Hanafi
- Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Egypt
| | | | - Ahmed Ihab Abdelaziz
- Department of Pharmacology and Toxicology, German University in Cairo, New Cairo City, Egypt
- School of Medicine, NewGiza University, Cairo, Egypt
- *Correspondence to: Ahmed Ihab Abdelaziz, Department of Molecular Medicine, School of Medicine, Newgiza University, Cairo 11431, Egypt. Tel: +20-238277847, E-mail:
| |
Collapse
|
17
|
Caveolin proteins: a molecular insight into disease. Front Med 2016; 10:397-404. [DOI: 10.1007/s11684-016-0483-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 08/17/2016] [Indexed: 12/31/2022]
|
18
|
Chen J, Zeng W, Han C, Wu J, Zhang H, Tong X. Mutation in the caveolin-3 gene causes asymmetrical distal myopathy. Neuropathology 2016; 36:485-489. [PMID: 26947586 DOI: 10.1111/neup.12297] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 11/27/2022]
Abstract
Mutations in the gene encoding caveolin-3 (CAV3) can cause a broad spectrum of clinical phenotypes, including limb girdle muscular dystrophy, rippling muscle disease, distal myopathy (MD), idiopathic persistent elevation of serum creatine kinase and cardiomyopathy. MD is a relatively rare subtype of caveolinopathy. Here, we report a sporadic case of a middle-aged female Chinese patient with MD in which a CAV3 mutation was identical to that previously reported in cases of rippling muscle disease. T1-weighted enhanced skeletal muscle MRI of the lower limbs showed an abnormal signal in the distal and proximal muscles. A muscle biopsy revealed moderate dystrophic changes, and immunohistochemical staining showed reduced CAV-3 expression in the plasmalemma. Genetic analysis revealed a heterozygous c.136G > A (p.Ala46Thr) CAV3 mutation that appeared to be de novo because it was absent from the patient's parents. This study suggested that the CAV3 c.136G > A (p.Ala46Thr) mutation can cause MD as well as different phenotypes in different individuals, suggesting that additional unknown loci must affect the disease phenotypes.
Collapse
Affiliation(s)
- Juanjuan Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wenshuang Zeng
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chunxi Han
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Haiou Zhang
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaoxin Tong
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
19
|
Abstract
Spatial organization of the plasma membrane is an essential feature of the cellular response to external stimuli. Receptor organization at the cell surface mediates transmission of extracellular stimuli to intracellular signalling molecules and effectors that impact various cellular processes including cell differentiation, metabolism, growth, migration and apoptosis. Membrane domains include morphologically distinct plasma membrane invaginations such as clathrin-coated pits and caveolae, but also less well-defined domains such as lipid rafts and the galectin lattice. In the present chapter, we will discuss interaction between caveolae, lipid rafts and the galectin lattice in the control of cancer cell signalling.
Collapse
|
20
|
Farrokh S, Brillen AL, Haendeler J, Altschmied J, Schaal H. Critical regulators of endothelial cell functions: for a change being alternative. Antioxid Redox Signal 2015; 22:1212-29. [PMID: 25203279 DOI: 10.1089/ars.2014.6023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE The endothelium regulates vessel dilation and constriction, balances hemostasis, and inhibits thrombosis. In addition, pro- and anti-angiogenic molecules orchestrate proliferation, survival, and migration of endothelial cells. Regulation of all these processes requires fine-tuning of signaling pathways, which can easily be tricked into running the opposite direction when exogenous or endogenous signals get out of hand. Surprisingly, some critical regulators of physiological endothelial functions can turn malicious by mere alternative splicing, leading to the expression of protein isoforms with opposite functions. RECENT ADVANCES While reviewing the evidence of alternative splicing on cellular physiology, it became evident that expression of splice factors and their activities are regulated by externally triggered signaling cascades. Furthermore, genome-wide identification of RNA-binding sites of splicing regulatory proteins now offer a glimpse into the splicing code responsible for alternative splicing of molecules regulating endothelial functions. CRITICAL ISSUES Due to the constantly growing number of transcript and protein isoforms, it will become more and more important to identify and characterize all transcripts and proteins regulating endothelial cell functions. One critical issue will be a non-ambiguous nomenclature to keep consistency throughout different laboratories. FUTURE DIRECTIONS RNA-deep sequencing focusing on exon-exon junction needs to more reliably identify alternative splicing events combined with functional analyses that will uncover more splice variants contributing to or inhibiting proper endothelial functions. In addition, understanding the signals mediating alternative splicing and its regulation might allow us to derive new strategies to preserve endothelial function by suppressing or upregulating specific protein isoforms. Antioxid. Redox Signal. 22, 1212-1229.
Collapse
Affiliation(s)
- Sabrina Farrokh
- 1 Heisenberg-Group-Environmentally-Induced Cardiovascular Degeneration, IUF-Leibniz Research Institute for Environmental Medicine , Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
21
|
Ilina P, Partti S, Niklander J, Ruponen M, Lou YR, Yliperttula M. Effect of differentiation on endocytic profiles of endothelial and epithelial cell culture models. Exp Cell Res 2015; 332:89-101. [DOI: 10.1016/j.yexcr.2015.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/31/2022]
|
22
|
Peng Q, Jia SH, Parodo J, Ai Y, Marshall JC. Pre-B cell colony enhancing factor induces Nampt-dependent translocation of the insulin receptor out of lipid microdomains in A549 lung epithelial cells. Am J Physiol Endocrinol Metab 2015; 308:E324-33. [PMID: 25516545 DOI: 10.1152/ajpendo.00006.2014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pre-B cell colony-enhancing factor (PBEF) is a highly conserved pleiotropic protein reported to be an alternate ligand for the insulin receptor (IR). We sought to clarify the relationship between PBEF and insulin signaling by evaluating the effects of PBEF on the localization of the IRβ chain to lipid rafts in A549 epithelial cells. We isolated lipid rafts from A549 cells and detected the IR by immunoprecipitation from raft fractions or whole cell lysates. Cells were treated with rPBEF, its enzymatic product nicotinamide adenine dinucleotide (NAD), or the Nampt inhibitor daporinad to study the effect of PBEF on IRβ movement. We used coimmunoprecipitation studies in cells transfected with PBEF and IRβ constructs to detect interactions between PBEF, the IRβ, and caveolin-1 (Cav-1). PBEF was present in both lipid raft and nonraft fractions, whereas the IR was found only in lipid raft fractions of resting A549 cells. The IR-, PBEF-, and Cav-1-coimmunoprecipitated rPBEF treatment resulted in the movement of IRβ- and tyrosine-phosphorylated Cav-1 from lipid rafts to nonrafts, an effect that could be blocked by daporinad, suggesting that this effect was facilitated by the Nampt activity of PBEF. The addition of PBEF to insulin-treated cells resulted in reduced Akt phosphorylation of both Ser⁴⁷³ and Thr³⁰⁸. We conclude that PBEF can inhibit insulin signaling through the IR by Nampt-dependent promotion of IR translocation into the nonraft domains of A549 epithelial cells. PBEF-induced alterations in the spatial geometry of the IR provide a mechanistic explanation for insulin resistance in inflammatory states associated with upregulation of PBEF.
Collapse
Affiliation(s)
- Qianyi Peng
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Song Hui Jia
- Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - Jean Parodo
- Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - Yuhang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - John C Marshall
- Department of Surgery, Department of Critical Care Medicine, and Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
23
|
Smith-Jackson K, Hentschke MR, Poli-de-Figueiredo CE, Pinheiro da Costa BE, Kurlak LO, Broughton Pipkin F, Czajka A, Mistry HD. Placental expression of eNOS, iNOS and the major protein components of caveolae in women with pre-eclampsia. Placenta 2015; 36:607-10. [PMID: 25707739 DOI: 10.1016/j.placenta.2015.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/21/2015] [Accepted: 02/02/2015] [Indexed: 01/08/2023]
Abstract
Caveolae regulate many cardiovascular functions and thus could be of interest in relation to pre-eclampsia, a pregnancy specific disorder characterised by hypertension and proteinuria. We examined placental mRNA and protein expression/localisation of the caveolae components Caveolin 1-3, Cavin 1-4 as well as eNOS/iNOS in normotensive control (n = 24) and pre-eclamptic pregnancies (n = 19). Placental mRNA expression of caveolin-1, cavin 1-3, was lower and eNOS expression was increased in pre-eclampsia (P < 0.05 for all). Additionally Caveolin-1 protein expression was also reduced in pre-eclampsia (P = 0.007); this could be an adaptive response in pre-eclampsia, possibly to attenuate the oxidative stress/inflammation.
Collapse
Affiliation(s)
- K Smith-Jackson
- Division of Women's Health, King's College London, Women's Health Academic Centre, UK; Department of Obstetrics & Gynaecology, School of Medicine, University of Nottingham, UK
| | - M R Hentschke
- Division of Women's Health, King's College London, Women's Health Academic Centre, UK; Department of Obstetrics & Gynaecology, School of Medicine, University of Nottingham, UK; Laboratory of Nephrology - IPB, School of Medicine, PUCRS, Brazil
| | | | | | - L O Kurlak
- Department of Obstetrics & Gynaecology, School of Medicine, University of Nottingham, UK
| | - F Broughton Pipkin
- Department of Obstetrics & Gynaecology, School of Medicine, University of Nottingham, UK
| | - A Czajka
- Division of Diabetes and Nutritional Sciences, Diabetes Research Group, King's College London, UK
| | - H D Mistry
- Division of Women's Health, King's College London, Women's Health Academic Centre, UK; Department of Obstetrics & Gynaecology, School of Medicine, University of Nottingham, UK.
| |
Collapse
|
24
|
Ma Q, Han Y, Chen C, Cao Y, Wang S, Shen W, Zhang H, Li Y, van Dongen MA, He B, Yu M, Xu L, Banaszak Holl MM, Liu G, Zhang Q, Qi R. Oral absorption enhancement of probucol by PEGylated G5 PAMAM dendrimer modified nanoliposomes. Mol Pharm 2015; 12:665-74. [PMID: 25587935 DOI: 10.1021/mp500388m] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Probucol (PB), an antioxidant drug, is commonly used as a lipid concentration lowering drug to reduce blood plasma cholesterol levels in the clinic. However, the therapeutic effects of this drug are negatively impacted by its poor water solubility and low oral absorption efficiency. In this study, a PEGylated G5 PAMAM dendrimer (G5-PEG) modified nanoliposome was employed to increase water solubility, transepithelial transport, and oral absorption of PB. The uptake mechanism was explored in vitro in Caco-2 cells with the results suggesting that the absorption improvement of G5-PEG modified PB-liposome (PB-liposome/G5-PEG) was related to P-glycoprotein (P-gp) efflux pump but was independent of caveolae endocytosis pathways. Additionally, plasma lipid concentration lowering effects of PB-liposome/G5-PEG were evaluated in vivo in a LDLR-/- hyperlipidemia mouse model. Compared with saline treated group, treatment with PB-liposome/G5-PEG significantly inhibited the increase of plasma total cholesterol (TC) and triglyceride (TG) of mice induced by a high fat diet. Moreover, its lipid concentration lowering effects and plasma drug concentration were greater than PB alone or commercial PB tablets. Our results demonstrated that PB-liposome/G5-PEG significantly increased the oral absorption of PB and therefore significantly improved its pharmacodynamic effects.
Collapse
Affiliation(s)
- Qian Ma
- Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, Peking University , Beijing 100191, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Lee SH, Woo TG, Lee SJ, Kim JS, Ha NC, Park BJ. Extracellular p53 fragment re-enters K-Ras mutated cells through the caveolin-1 dependent early endosomal system. Oncotarget 2014; 4:2523-31. [PMID: 24344114 PMCID: PMC3926846 DOI: 10.18632/oncotarget.1550] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
K-Ras mutation is detected in over 30% of human malignancies. In particular, 90% of human pancreatic cancers are initiated by K-Ras mutation. Thus, selective elimination of K-Ras mutated cells would be a plausible strategy to prevent or cure the malignancies. In our previous reports, it has been revealed that oncogenic K-Ras promotes the exocytosis of p53 with Snail. In this study, we have followed the final destination of extracellular p53, which is secreted by the Snail complex. Here we provide evidences that p53, exported from K-Ras-mutated cells, is specifically re-endocytosed by oncogenic K-Ras-containing cancer cells. The p53 DNA-binding domain directly associates with caveolin-1 and enters K-Ras mutated cells through early endosome-mediated endocytosis. Using a serial deletion approach, we revealed that a fragment of human p53 extending from 93-143 amino acids (AA) is responsible for binding with caveolin-1 and for endocytosis. In contrast, p53-Snail binding occurs at the 143-193 aa region. Finally, through in vivo study, we confirmed that injected recombinant p53 could be up-taken by tumor tissues, constructed by oncogenic K-Ras transformed MEF cells. In contrast, the tumors formed by H-Ras mutated MEF cells did not accumulate the injected p53 protein. These results indicate that the p53 fragment might be useful as a specific delivery tool into K- Ras mutated cells as well as a diagnostic method.
Collapse
Affiliation(s)
- Sun-Hye Lee
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | | | | | | | | | | |
Collapse
|
27
|
Wang S, Zhang C, Liu Y, Xu C, Chen Z. Functional polymorphisms of caveolin-1 variants as potential biomarkers of esophageal squamous cell carcinoma. Biomarkers 2014; 19:652-9. [PMID: 25271040 DOI: 10.3109/1354750x.2014.968210] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate the association of caveolin-1 (CAV1) genetic variants (C239A (rs1997623), G14713A (rs3807987), G21985A (rs12672038), T29107A (rs7804372)) with esophageal squamous cell carcinoma (ESCC) susceptibility. METHODS A total of 427 patients with ESCC and 427 healthy controls were genotyped using the polymerase chain reaction and restriction fragment length polymorphism (PCR-RFLP) method. RESULTS There were significant differences between patients and controls in distributions of their genotypes and allelic frequencies in G14713A and T29107A polymorphisms. Furthermore, haplotype analysis revealed that haplotypes CAAT and CAGT were associated with high risk for ESCC, while haplotype CGGA was protective against ESCC. Stratified analysis showed the associations between the SNPs (G14713A and T29107A) and ESCC risk were noteworthy among female patients and patients who never smoke or drank alcohol. CONCLUSIONS Genetic polymorphisms of CAV1 G14713A and T29107A might affect an individual's susceptibility in developing ESCC, making them efficient potential genetic biomarkers for early detection of ESCC.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Gastroenterology, Qianfoshan Hospital, Shandong University , Jinan, Shandong Province , China and
| | | | | | | | | |
Collapse
|
28
|
The estrogen receptor β-PI3K/Akt pathway mediates the cytoprotective effects of tocotrienol in a cellular Parkinson's disease model. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1303-12. [DOI: 10.1016/j.bbadis.2014.04.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/20/2014] [Accepted: 04/07/2014] [Indexed: 11/18/2022]
|
29
|
Kinoshita M, Matsumori N, Murata M. Coexistence of two liquid crystalline phases in dihydrosphingomyelin and dioleoylphosphatidylcholine binary mixtures. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:1372-81. [DOI: 10.1016/j.bbamem.2014.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 11/30/2022]
|
30
|
Kozlov MM, Campelo F, Liska N, Chernomordik LV, Marrink SJ, McMahon HT. Mechanisms shaping cell membranes. Curr Opin Cell Biol 2014; 29:53-60. [PMID: 24747171 DOI: 10.1016/j.ceb.2014.03.006] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 01/08/2023]
Abstract
Membranes of intracellular organelles are characterized by large curvatures with radii of the order of 10-30nm. While, generally, membrane curvature can be a consequence of any asymmetry between the membrane monolayers, generation of large curvatures requires the action of mechanisms based on specialized proteins. Here we discuss the three most relevant classes of such mechanisms with emphasis on the physical requirements for proteins to be effective in generation of membrane curvature. We provide new quantitative estimates of membrane bending by shallow hydrophobic insertions and compare the efficiency of the insertion mechanism with those of the protein scaffolding and crowding mechanisms.
Collapse
Affiliation(s)
- Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel.
| | - Felix Campelo
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Nicole Liska
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Leonid V Chernomordik
- Section on Membrane Biology, Program of Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States
| | - Siewert J Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Harvey T McMahon
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK.
| |
Collapse
|
31
|
Royce SG, Le Saux CJ. Role of caveolin-1 in asthma and chronic inflammatory respiratory diseases. Expert Rev Respir Med 2014; 8:339-47. [PMID: 24742020 DOI: 10.1586/17476348.2014.905915] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Caveolin-1 (Cav-1) is the major protein present in invaginations of the plasma membrane of cells known as caveolae. Cav-1 is expressed in numerous resident and inflammatory cells implicated in the pathogenesis of asthma and chronic inflammatory respiratory diseases including chronic obstructive pulmonary disease. A remarkable repertoire of functions has been identified for Cav-1 and these extend to, and have relevance to, asthma and chronic inflammatory respiratory diseases. Important processes influenced by Cav-1 include inflammation, fibrosis, smooth muscle contractility, regulation of apoptosis and cell senescence as well as epithelial barrier function and homeostasis. A better understanding of Cav-1 may be useful in developing new therapies for chronic inflammatory respiratory diseases.
Collapse
Affiliation(s)
- Simon G Royce
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
32
|
Arndt L, Castonguay J, Arlt E, Meyer D, Hassan S, Borth H, Zierler S, Wennemuth G, Breit A, Biel M, Wahl-Schott C, Gudermann T, Klugbauer N, Boekhoff I. NAADP and the two-pore channel protein 1 participate in the acrosome reaction in mammalian spermatozoa. Mol Biol Cell 2014; 25:948-64. [PMID: 24451262 PMCID: PMC3952862 DOI: 10.1091/mbc.e13-09-0523] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A TPCN1 gene–deficient mouse strain is used to show that two convergent working NAADP-dependent pathways with nonoverlapping activation and self-inactivation profiles for distinct NAADP concentrations drive acrosomal exocytosis, by which TPC1 is central for the pathway activated by low-micromolar NAADP concentrations. The functional relationship between the formation of hundreds of fusion pores during the acrosome reaction in spermatozoa and the mobilization of calcium from the acrosome has been determined only partially. Hence, the second messenger NAADP, promoting efflux of calcium from lysosome-like compartments and one of its potential molecular targets, the two-pore channel 1 (TPC1), were analyzed for its involvement in triggering the acrosome reaction using a TPCN1 gene–deficient mouse strain. The present study documents that TPC1 and NAADP-binding sites showed a colocalization at the acrosomal region and that treatment of spermatozoa with NAADP resulted in a loss of the acrosomal vesicle that showed typical properties described for TPCs: Registered responses were not detectable for its chemical analogue NADP and were blocked by the NAADP antagonist trans-Ned-19. In addition, two narrow bell-shaped dose-response curves were identified with maxima in either the nanomolar or low micromolar NAADP concentration range, where TPC1 was found to be responsible for activating the low affinity pathway. Our finding that two convergent NAADP-dependent pathways are operative in driving acrosomal exocytosis supports the concept that both NAADP-gated cascades match local NAADP concentrations with the efflux of acrosomal calcium, thereby ensuring complete fusion of the large acrosomal vesicle.
Collapse
Affiliation(s)
- Lilli Arndt
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians University, 81377 München, Germany Department of Pharmacy, Ludwig-Maximilians University, 81377 München, Germany Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University, 79104 Freiburg, Germany Institute for Anatomy, University of Duisburg-Essen, 45141 Essen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
de Freitas AC, Coimbra EC, Leitão MDCG. Molecular targets of HPV oncoproteins: potential biomarkers for cervical carcinogenesis. Biochim Biophys Acta Rev Cancer 2014; 1845:91-103. [PMID: 24388872 DOI: 10.1016/j.bbcan.2013.12.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 12/10/2013] [Accepted: 12/27/2013] [Indexed: 12/17/2022]
Abstract
Cervical cancer is the second most common cancer among women worldwide and is responsible for 275,000 deaths each year. Persistent infection with high-risk human papillomavirus (HR-HPV) is an essential factor for the development of cervical cancer. Although the process is not fully understood, molecular mechanisms caused by HPV infection are necessary for its development and reveal a large number of potential biomarkers for diagnosis and prognosis. These molecules are host genes and/or proteins, and cellular microRNAs involved in cell cycle regulation that result from disturbed expression of HR-HPV E5, E6 and E7 oncoproteins. One of the current challenges in medicine is to discover potent biomarkers that can correctly diagnose cervical premalignant lesions and standardize clinical management. Currently, studies are showing that some of these molecules are potential biomarkers of cervical carcinogenesis, and it is possible to carry out a more accurate diagnosis and provide more appropriate follow-up treatment for women with cervical dysplasia. In this paper, we review recent research studies on cell cycle molecules deregulated by HPV infections, as well as their potential use for cervical cancer screening.
Collapse
Affiliation(s)
- Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Center for Biological Sciences, Federal University of Pernambuco, Recife, Brazil.
| | - Eliane Campos Coimbra
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Center for Biological Sciences, Federal University of Pernambuco, Recife, Brazil.
| | - Maria da Conceição Gomes Leitão
- Laboratory of Molecular Studies and Experimental Therapy, Department of Genetics, Center for Biological Sciences, Federal University of Pernambuco, Recife, Brazil.
| |
Collapse
|
34
|
Qin H, Bollag WB. The caveolin-1 scaffolding domain peptide decreases phosphatidylglycerol levels and inhibits calcium-induced differentiation in mouse keratinocytes. PLoS One 2013; 8:e80946. [PMID: 24236206 PMCID: PMC3827482 DOI: 10.1371/journal.pone.0080946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 10/14/2013] [Indexed: 11/19/2022] Open
Abstract
Phospholipase D2 (PLD2) has been found localized in low-density caveolin-rich membrane microdomains. Our previous study suggested that PLD2 and aquaporin 3 (AQP3) interact in these domains to inhibit keratinocyte proliferation and promote differentiation by cooperating to produce phosphatidylglycerol. To examine the effect of membrane microdomain localization on the PLD2/AQP3 signaling module and keratinocyte proliferation and differentiation, we treated mouse keratinocytes with 3 µM cell-permeable caveolin-1 scaffolding domain peptide or a negative control peptide and stimulated cell differentiation using a moderately elevated extracellular calcium concentration (125 uM) to maximally promote differentiation and phosphatidylglycerol production. Cell proliferation, differentiation, total PLD activity, phosphatidylglycerol levels, and AQP3 activity were monitored. The caveolin-1 scaffolding domain peptide itself had no effect on phosphatidylglycerol levels or keratinocyte proliferation or differentiation but prevented the changes induced by a moderately elevated calcium concentration, whereas a negative control did not. The caveolin-1 scaffolding domain peptide had little effect on total PLD activity or glycerol uptake (AQP3 activity). We conclude that the caveolin-1 scaffolding domain peptide disrupts the functional association between AQP3 and PLD2 and prevents both the inhibited proliferation and the stimulated differentiation in response to elevated extracellular calcium levels. The interaction of caveolin-1 and PLD2 is indirect (i.e., lipid mediated); together with the proliferation-promoting effects of caveolin-1 knockout on epidermal keratinocytes, we propose that the caveolin-1 scaffolding domain pepetide exerts a dominant-negative effect on caveolin-1 to alter lipid rafts in these cells.
Collapse
Affiliation(s)
- Haixia Qin
- Department of Physiology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
| | - Wendy B. Bollag
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
- Department of Physiology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- Department of Medicine (Dermatology), Medical College of Georgia at Georgia Regents University, Augusta, Georgia, United States of America
- Departments of Orthopaedic Surgery, Oral Biology and Cell Biology and Anatomy, Georgia Regents University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
35
|
Hedley PL, Kanters JK, Dembic M, Jespersen T, Skibsbye L, Aidt FH, Eschen O, Graff C, Behr ER, Schlamowitz S, Corfield V, McKenna WJ, Christiansen M. The role of CAV3 in long-QT syndrome: clinical and functional assessment of a caveolin-3/Kv11.1 double heterozygote versus caveolin-3 single heterozygote. ACTA ACUST UNITED AC 2013; 6:452-61. [PMID: 24021552 DOI: 10.1161/circgenetics.113.000137] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Mutations in CAV3, coding for caveolin-3, the major constituent scaffolding protein of cardiac caveolae, have been associated with skeletal muscle disease, cardiomyopathy, and most recently long-QT syndrome (LQTS) and sudden infant death syndrome. We examined the occurrence of CAV3 mutations in a large cohort of patients with LQTS. METHODS AND RESULTS Probands with LQTS (n=167) were screened for mutations in CAV3 using direct DNA sequencing. A single proband (0.6%) was found to be a heterozygous carrier of a previously described missense mutation, caveolin-3:p.T78M. The proband was also a heterozygous carrier of the trafficking-deficient Kv11.1:p.I400N mutation. The caveolin-3:p.T78M mutation was found isolated in 3 family members, none of whom had a prolonged QTc interval. Coimmunoprecipitations of caveolin-3 and the voltage-gated potassium channel subunit (Kv11.1) were performed, and the electrophysiological classification of the Kv11.1 mutant was carried out by patch-clamp technique in human embryonic kidney 293 cells. Furthermore, the T-wave morphology was assessed in mutation carriers, double mutation carriers, and nonmutation carriers by applying a morphology combination score. The morphology combination score was normal for isolated caveolin-3:p.T78M carriers and of LQT2 type in double heterozygotes. CONCLUSIONS Mutations in CAV3 are rare in LQTS. Furthermore, caveolin-3:p.T78M did not exhibit a LQTS phenotype. Because no association has ever been found between LQTS and isolated CAV3 mutations, we suggest that LQTS9 is considered a provisional entity.
Collapse
Affiliation(s)
- Paula L Hedley
- Department of Clinical Biochemistry, Immunology, and Genetics, Statens Serum Institut, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Trans-resveratrol down-regulates caveolin-1, up-regulates endothelial NO synthase and reduces their interaction in vascular smooth muscle and endothelial cells. FOOD BIOSCI 2013. [DOI: 10.1016/j.fbio.2013.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
37
|
Basu Roy UK, Henkhaus RS, Loupakis F, Cremolini C, Gerner EW, Ignatenko NA. Caveolin-1 is a novel regulator of K-RAS-dependent migration in colon carcinogenesis. Int J Cancer 2013; 133:43-57. [PMID: 23280667 DOI: 10.1002/ijc.28001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 11/12/2012] [Accepted: 12/05/2012] [Indexed: 12/13/2022]
Abstract
Caveolin-1 is an essential component of membrane caveolae. It is an important regulator of cellular processes such as signal transduction and endocytosis. We report here, for the first time, that caveolin-1 is a target of the K-RAS oncogene in colon carcinogenesis. Caveolin-1 is induced in colon cancer cells and in human colon tumor samples, in response to K-RAS activating mutations. An activated K-RAS oncogene transcriptionally induces caveolin-1 expression in human colon cancer cells and this effect is not restricted to the type of activating K-RAS mutation. Inhibition of the P-I3 Kinase-AKT pathway, but not the ERK MAPK pathway, both important K-RAS effectors, leads to a decrease in caveolin-1 expression indicating that the AKT pathway is involved in caveolin-1 expression in response to an activated K-RAS. Increased AKT signaling induces caveolin-1 expression by increasing the activity of the transcription factor, Sp1. Interestingly; caveolin-1 depletion alters K-RAS-dependent signaling by decreasing Grb2-SOS activity. Consistent with these finding, caveolin-1-depleted cells shows decreased migration in vitro. However, caveolin-1 overexpression by itself does not increase migration whereas an activated Src can increase migration in a caveolin-1-dependent manner. This increased migration is highly dependent on the RhoA GTPase, indicating that an activated K-RAS modulates migration in part via caveolin-1 induction, and increasing RhoA activity via phospho-caveolin-1. Our findings indicate that K-RAS regulates both caveolin-1 expression and other factors affecting caveolin-1 functions in colon cancer-derived cell migration.
Collapse
Affiliation(s)
- Upal K Basu Roy
- Department of Biochemistry and Molecular Biophysics, Biochemistry and Molecular and Cellular Biology Graduate Program, University of Arizona, Tucson, AZ, USA
| | | | | | | | | | | |
Collapse
|
38
|
The Role of Cholesterol in Prostate Cancer. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
39
|
Lokappa SB, Nagaraj R. Interaction of peptides spanning the transmembrane domain of caveolin-1 with model membranes. J Pept Sci 2012; 18:696-703. [DOI: 10.1002/psc.2457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 08/29/2012] [Accepted: 08/30/2012] [Indexed: 01/17/2023]
Affiliation(s)
- Sowmya Bekshe Lokappa
- CSIR-Centre for Cellular and Molecular Biology; Uppal Road; Hyderabad; 500 007; India
| | - Ramakrishnan Nagaraj
- CSIR-Centre for Cellular and Molecular Biology; Uppal Road; Hyderabad; 500 007; India
| |
Collapse
|
40
|
Abstract
Entry of reovirus virions has been well studied in several tissue culture systems. After attachment to junctional adhesion molecule A (JAM-A), virions undergo clathrin-mediated endocytosis followed by proteolytic disassembly of the capsid and penetration to the cytoplasm. However, during in vivo infection of the intestinal tract, and likely in the tumor microenvironment, capsid proteolysis (uncoating) is initiated extracellularly. We used multiple approaches to determine if uncoated reovirus particles, called intermediate subviral particles (ISVPs), enter cells by directly penetrating the limiting membrane or if they take advantage of endocytic pathways to establish productive infection. We found that entry and infection by reovirus ISVPs was inhibited by dynasore, an inhibitor of dynamin-dependent endocytosis, as well as by genistein and dominant-negative caveolin-1, which block caveolar endocytosis. Inhibition of caveolar endocytosis also reduced infection by reovirus virions. Extraction of membrane cholesterol with methyl-β-cyclodextrin inhibited infection by virions but had no effect when infection was initiated with ISVPs. We found this pathway to be independent of both clathrin and caveolin. Together, these data suggest that reovirus virions can use both dynamin-dependent and dynamin-independent endocytic pathways during cell entry, and they reveal that reovirus ISVPs can take advantage of caveolar endocytosis to establish productive infection.
Collapse
|
41
|
Shao C, Kendall E, DeVoe DL. Electro-optical BLM chips enabling dynamic imaging of ordered lipid domains. LAB ON A CHIP 2012; 12:3142-9. [PMID: 22728885 PMCID: PMC3411933 DOI: 10.1039/c2lc40077d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Studies of lipid rafts, ordered microdomains of sphingolipids and cholesterol within cell membranes, are essential in probing the relationships between membrane organization and cellular function. While in vitro studies of lipid phase separation are commonly performed using spherical vesicles as model membranes, the utility of these models is limited by a number of factors. Here we present a microfluidic device that supports simultaneous electrical measurements and confocal imaging of on-chip bilayer lipid membranes (BLMs), enabling real-time multi-domain imaging of membrane organization. The chips further support closed microfluidic access to both sides of the membrane, allowing the membrane boundary conditions to be rapidly changed and providing a mechanism for dynamically adjusting membrane curvature through application of a transmembrane pressure gradient. Here we demonstrate the platform through the study of dynamic generation and dissolution of ordered lipid domains as membrane components are transported to and from the supporting annulus containing solvated lipids and cholesterol.
Collapse
Affiliation(s)
- Chenren Shao
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| | - Eric Kendall
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| | - Don L. DeVoe
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
42
|
Yang XY, Huang CC, Kan QM, Li Y, Liu D, Zhang XC, Sato T, Yamagata S, Yamagata T. Calcium regulates caveolin-1 expression at the transcriptional level. Biochem Biophys Res Commun 2012; 426:334-41. [PMID: 22940132 DOI: 10.1016/j.bbrc.2012.08.079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 08/15/2012] [Indexed: 11/29/2022]
Abstract
Caveolin-1, an indispensable component of caveolae serving as a transformation suppressor protein, is highly expressed in poorly metastatic mouse osteosarcoma FBJ-S1 cells while highly metastatic FBJ-LL cells express low levels of caveolin-1. Calcium concentration is higher in FBJ-S1 cells than in FBJ-LL cells; therefore, we investigated the possibility that calcium signaling positively regulates caveolin-1 in mouse FBJ-S1 cells. When cells were treated with the calcium channel blocker nifedipine, cyclosporin A (a calcineurin inhibitor), or INCA-6 (a nuclear factor of activated T-cells [NFAT] inhibitor), caveolin-1 expression at the mRNA and protein levels decreased. RNA silencing of voltage-dependent L-type calcium channel subunit alpha-1C resulted in suppression of caveolin-1 expression. This novel caveolin-1 regulation pathway was also identified in mouse NIH 3T3 cells and Lewis lung carcinoma cells. These results indicate that caveolin-1 is positively regulated at the transcriptional level through a novel calcium signaling pathway mediated by L-type calcium channel/Ca(2+)/calcineurin/NFAT.
Collapse
Affiliation(s)
- Xiao-Yan Yang
- Laboratory of Tumor Biology and Glycobiology, Department of Life Sciences, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ricci G, Scionti I, Tupler R, Siciliano G. Response. Neuromuscul Disord 2012. [DOI: 10.1016/j.nmd.2012.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
44
|
Arielly SS, Ariel M, Yehuda R, Scigelova M, Yehezkel G, Khalaila I. Quantitative analysis of caveolin-rich lipid raft proteins from primary and metastatic colorectal cancer clones. J Proteomics 2012; 75:2629-37. [PMID: 22484058 DOI: 10.1016/j.jprot.2012.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 01/26/2012] [Accepted: 03/08/2012] [Indexed: 12/26/2022]
Abstract
Caveolin-rich lipid rafts (CLRs) are thickened sections of the cell membrane that are composed of the integral membrane proteins caveolins together with saturated long chain fatty acids, cholesterol and lipids. Membrane proteins - lipid raft proteins in particular - may play important roles in cell signaling and cell-cell interaction. Due to their unique structure, CLRs seem to be the preferred docking site for specific proteins involved in focal adhesion and cancer metastasis. Our objective was thus to identify and quantify CLR proteins from primary and metastatic colorectal cancer (CRC) clones. We found differential expression of nine CLR proteins from primary and metastatic CRC clones. Among the identified proteins, an immune system inhibiting protein was significantly overexpressed in the metastatic clone, while cell adhesion and transport molecules were among the overexpressed proteins in the primary clone. All the identified CRL proteins are involved in tumorigenesis, specifically metastasis, and may thus serve as therapeutic targets. A novel concept for identification and quantification of CLR proteins with label-free mass spectrometry method was specifically examined in this study. Validation of the method against immunoblotting and FACS analysis indicates that it can be applied for the identification of novel biomarkers for cancer and metastasis.
Collapse
Affiliation(s)
- Shirley S Arielly
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | | | | | | | | | | |
Collapse
|
45
|
Elucidation of the Rotavirus NSP4-Caveolin-1 and -Cholesterol Interactions Using Synthetic Peptides. JOURNAL OF AMINO ACIDS 2012; 2012:575180. [PMID: 22500212 PMCID: PMC3303745 DOI: 10.1155/2012/575180] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 11/16/2011] [Indexed: 01/19/2023]
Abstract
Rotavirus (RV) NSP4, the first described viral enterotoxin, is a multifunctional glycoprotein that contributes to viral pathogenesis, morphogenesis, and replication. NSP4 binds both termini of caveolin-1 and is isolated from caveolae fractions that are rich in anionic phospholipids and cholesterol. These interactions indicate that cholesterol/caveolin-1 plays a role in NSP4 transport to the cell surface, which is essential to its enterotoxic activity. Synthetic peptides were utilized to identify target(s) of intervention by exploring the NSP4-caveolin-1 and -cholesterol interactions. NSP4112–140 that overlaps the caveolin-1 binding domain and a cholesterol recognition amino acid consensus (CRAC) motif and both termini of caveolin-1 (N-caveolin-12–20, 19–40 and C-caveolin-1161–180) were synthesized. Direct fluorescence-binding assays were employed to determine binding affinities of the NSP4-caveolin-1 peptides and cholesterol. Intracellular cholesterol alteration revealed a redistribution of NSP4 and disintegration of viroplasms. These data further imply interruption of NSP4112–140-N-caveolin-119–40 and cholesterol interactions may block NSP4 intracellular transport, hence enterotoxicity.
Collapse
|
46
|
Rippling muscle disease and facioscapulohumeral dystrophy-like phenotype in a patient carrying a heterozygous CAV3 T78M mutation and a D4Z4 partial deletion: Further evidence for "double trouble" overlapping syndromes. Neuromuscul Disord 2012; 22:534-40. [PMID: 22245016 PMCID: PMC3359497 DOI: 10.1016/j.nmd.2011.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 11/11/2011] [Accepted: 12/01/2011] [Indexed: 01/12/2023]
Abstract
We report the first case of a heterozygous T78M mutation in the caveolin-3 gene (CAV3) associated with rippling muscle disease and proximal myopathy. The patient displayed also bilateral winged scapula with limited abduction of upper arms and marked asymmetric atrophy of leg muscles shown by magnetic resonance imaging. Immunohistochemistry on the patient’s muscle biopsy demonstrated a reduction of caveolin-3 staining, compatible with the diagnosis of caveolinopathy. Interestingly, consistent with the possible diagnosis of FSHD, the patient carried a 35 kb D4Z4 allele on chromosome 4q35. We discuss the hypothesis that the two genetic mutations may exert a synergistic effect in determining the phenotype observed in this patient.
Collapse
|
47
|
Abstract
Despite the progress in medical treatment sepsis remains one of the major causes of death in pediatric and elderly patients. Understanding signaling pathways associated with sepsis may be of key significance for designing more efficient therapeutic approaches which could alleviate sepsis outcome. Earlier studies suggested that cholesteroland sphingolipid-rich lipid rafts and their morphologically distinct subset, caveolaecan be utilized by certain bacterial pathogens to enter and invade host cells. Moreover, there is also evidence that the expression levels of the major caveolar coat proteincaveolin-1 can be regulated by the major component of the outer membrane of Gram-negative bacteria,lipopolysaccharide (LPS) in various cell types involved in sepsis. In particular recent studies using caveolin-1 knockout mice and cells have revealed that caveolin-1 is directly involved in regulating numerous signalingpathways and functions in various cell types of the immune system and other cell types involved in sepsis. Moreover, the most recent report implies that in addition to extensively studied caveolin-1, caveolin-2 is also important in regulating LPS-induced sepsis and might possibly play an opposite role to caveolin-1 in regulating certain pro-inflammatory signaling pathways. The purpose of this review is to discuss these new exciting discoveries relatedto the specific role of caveolin-1 and the less studiedcaveolin-2in regulating signaling and outcome associated with sepsis induced by LPS and pathogenic bacteria at molecular, cellular and systemic levels.
Collapse
Affiliation(s)
- Grzegorz Sowa
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia,USA
| |
Collapse
|
48
|
Sowa G. Caveolae, caveolins, cavins, and endothelial cell function: new insights. Front Physiol 2012; 2:120. [PMID: 22232608 PMCID: PMC3252561 DOI: 10.3389/fphys.2011.00120] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 12/19/2011] [Indexed: 12/29/2022] Open
Abstract
Caveolae are cholesterol and glycosphingolipid-rich flask-shaped invaginations of the plasma membrane which are particularly abundant in vascular endothelium and present in all other cell types of the cardiovascular system, including vascular smooth-muscle cells, macrophages, cardiac myocytes, and fibroblasts. Caveolins and the more recently discovered cavins are the major protein components of caveolae. When caveolae were discovered, their functional role was believed to be limited to transport across the endothelial cell barrier. Since then, however, a large body of evidence has accumulated, suggesting that these microdomains are very important in regulating many other important endothelial cell functions, mostly due to their ability to concentrate and compartmentalize various signaling molecules. Over the course of several years, multiple studies involving knockout mouse and small interfering RNA approaches have considerably enhanced our understanding of the role of caveolae and caveolin-1 in regulating many cardiovascular functions. New findings have been reported implicating other caveolar protein components in endothelial cell signaling and function, such as the understudied caveolin-2 and newly discovered cavin proteins. The aim of this review is to focus primarily on molecular and cellular aspects of the role of caveolae, caveolins, and cavins in endothelial cell signaling and function. In addition, where appropriate, the possible implications for the cardiovascular and pulmonary physiology and pathophysiology will be discussed.
Collapse
Affiliation(s)
- Grzegorz Sowa
- Department of Medical Pharmacology and Physiology, University of MissouriColumbia, MO, USA
| |
Collapse
|
49
|
Sowa G. Regulation of Cell Signaling and Function by Endothelial Caveolins: Implications in Disease. TRANSLATIONAL MEDICINE (SUNNYVALE, CALIF.) 2012; Suppl 8:001. [PMID: 26605130 PMCID: PMC4655115 DOI: 10.4172/2161-1025.s8-001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Caveolae are cholesterol- and glycosphingolipid-rich omega-shaped invaginations of the plasma membrane that are very abundant in vascular endothelial cells and present in most cell types. Caveolins are the major coat protein components of caveolae. Multiple studies using knockout mouse, small interfering RNA, and cell-permeable peptide delivery approaches have significantly enhanced our understanding of the role of endothelial caveolae and caveolin-1 in physiology and disease. Several postnatal pulmonary and cardiovascular pathologies have been reported in caveolin-1 knockout mice, many of which have been recently rescued by selective re-expression of caveolin-1 in endothelium of these mice. A large body of experimental evidence mostly using caveolin-1 knockout mice suggests that, depending on the disease model, endothelial caveolin-1 may play either a protective or a detrimental role. For instance, physiological or higher expression levels of caveolin-1 in endothelium might be beneficial in such diseases as pulmonary hypertension, cardiac hypertrophy, or ischemic injury. On the other hand, endothelial caveolin-1 might contribute to acute lung injury and inflammation, atherosclerosis or pathological angiogenesis associated with inflammatory bowel disease. Moreover, depending on the specific model, endothelial caveolin-1 may either promote or suppress tumor-induced angiogenesis. In addition to overwhelming evidence for the role of endothelial caveolin-1, more recent studies also suggest that endothelial caveolin-2 could possibly play a role in pulmonary disease. The purpose of this review is to focus on how caveolin-1 expressed in endothelial cells regulates endothelial cell signaling and function. The review places particular emphasis on relevance to disease, including but not limited to Pulmonary and cardiovascular disorders as well as cancer. In addition to caveolin-1, possible importance of the less-studied endothelial caveolin-2 in pulmonary diseases will be also discussed.
Collapse
Affiliation(s)
- Grzegorz Sowa
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, 65212, USA
| |
Collapse
|
50
|
Reeves VL, Thomas CM, Smart EJ. Lipid rafts, caveolae and GPI-linked proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 729:3-13. [PMID: 22411310 DOI: 10.1007/978-1-4614-1222-9_1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lipid rafts and caveolae are specialized membrane microdomains enriched in sphingolipids and cholesterol. They function in a variety of cellular processes including but not limited to endocytosis, transcytosis, signal transduction and receptor recycling. Here, we outline the similarities and differences between lipid rafts and caveolae as well as discuss important components and functions of each.
Collapse
|