1
|
Mauriello A, Correra A, Maratea AC, Caturano A, Liccardo B, Perrone MA, Giordano A, Nigro G, D’Andrea A, Russo V. Serum Lipids, Inflammation, and the Risk of Atrial Fibrillation: Pathophysiological Links and Clinical Evidence. J Clin Med 2025; 14:1652. [PMID: 40095683 PMCID: PMC11899858 DOI: 10.3390/jcm14051652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Dyslipidemia is a metabolic disorder characterized by quantitative and/or qualitative abnormalities in serum lipid levels. Elevated serum cholesterol levels can modify the turnover and recruitment of ionic channels in myocytes and cellular homeostasis, including those of inflammatory cells. Experimental and clinical data indicate that inflammation is implicated in the pathophysiology of atrial remodeling, which is the substrate of atrial fibrillation (AF). Data about the association between increased lipid serum levels and AF are few and contrasting. Lipoprotein (a), adiposity, and inflammation seem to be the main drivers of AF; in contrast, low-density lipoproteins, high-density lipoproteins and triglycerides are not directly involved in AF onset. The present review aimed to describe the pathophysiological link between dyslipidemia and AF, the efficacy of lipid-lowering therapies in atherosclerotic cardiovascular disease (ASCVD) patients with and without AF, and the impact of lipid-lowering therapies on AF incidence.
Collapse
Affiliation(s)
- Alfredo Mauriello
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.C.M.); (B.L.); (G.N.)
- Cardiology and Intensive Care Unit, Department of Cardiology, “Umberto I” Hospital, 84014 Nocera Inferiore, Italy;
- Intensive Cardiac Care Unit, “San Giuseppe Moscati” Hospital, ASL Caserta 81031 Aversa, Italy;
| | - Adriana Correra
- Intensive Cardiac Care Unit, “San Giuseppe Moscati” Hospital, ASL Caserta 81031 Aversa, Italy;
| | - Anna Chiara Maratea
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.C.M.); (B.L.); (G.N.)
| | - Alfredo Caturano
- Internal Medicine Unit, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy;
| | - Biagio Liccardo
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.C.M.); (B.L.); (G.N.)
| | - Marco Alfonso Perrone
- Department of Cardiology and CardioLab, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Gerardo Nigro
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.C.M.); (B.L.); (G.N.)
| | - Antonello D’Andrea
- Cardiology and Intensive Care Unit, Department of Cardiology, “Umberto I” Hospital, 84014 Nocera Inferiore, Italy;
| | - Vincenzo Russo
- Cardiology Unit, Department of Medical and Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80131 Naples, Italy; (A.M.); (A.C.M.); (B.L.); (G.N.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| |
Collapse
|
2
|
Mu C, Feng D, Khan M, Song H, Munir S, Hu Q, Yu L. Colorimetric, Quantitative, and Portable Liquid Crystal Elastomer Biosensing of Cholesterol and Malathion. Anal Chem 2025; 97:3926-3936. [PMID: 39939117 DOI: 10.1021/acs.analchem.4c05166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Cholesteric liquid crystal elastomers (CLCEs) possess a unique characteristic that allows the manipulation of the reflected color by adjusting the spacing between layers. This attribute can create a reliable and cost-effective colorimetric sensing platform for the on-site detection of various substances. In this study, CLCE films were coupled with cross-linked poly(acrylic acid) (CLCE-PAA) or poly(dimethylaminoethyl methacrylate) (CLCE-PDA) films to monitor cholesterol and malathion levels, respectively. In both cases, the color change is recorded by a mobile phone camera, and the reflectance wavelength is measured spectrophotometrically. For on-site detection, a smartphone application was used to capture the film's image, process the color data into hue (H) values, and calculate the corresponding concentration of the tested analyte via an analysis program. Cholesterol and malathion can be detected within a linear range of 0.2-1.0 mM and 1-10,000 ng/mL, respectively. The corresponding recoveries for actual sample analysis were 86-115% and 87-122% for cholesterol and malathion, respectively. This system offers a practical solution for on-site testing of cholesterol and malathion in biological and environmental samples.
Collapse
Affiliation(s)
- Chongyang Mu
- Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, Shandong University, Jinan 250100, China
| | - Dawei Feng
- Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, Shandong University, Jinan 250100, China
| | - Mashooq Khan
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Haoyang Song
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Sundas Munir
- Department of Chemistry, University of Rahimyar Khan, Mianwali 42200, Pakistan
| | - Qiongzheng Hu
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Li Yu
- Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, Shandong University, Jinan 250100, China
| |
Collapse
|
3
|
Liang X, Mounier N, Apfel N, Khalid S, Frayling TM, Bowden J. Using clustering of genetic variants in Mendelian randomization to interrogate the causal pathways underlying multimorbidity from a common risk factor. Genet Epidemiol 2025; 49:e22582. [PMID: 39138631 PMCID: PMC11647065 DOI: 10.1002/gepi.22582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/17/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024]
Abstract
Mendelian randomization (MR) is an epidemiological approach that utilizes genetic variants as instrumental variables to estimate the causal effect of an exposure on a health outcome. This paper investigates an MR scenario in which genetic variants aggregate into clusters that identify heterogeneous causal effects. Such variant clusters are likely to emerge if they affect the exposure and outcome via distinct biological pathways. In the multi-outcome MR framework, where a shared exposure causally impacts several disease outcomes simultaneously, these variant clusters can provide insights into the common disease-causing mechanisms underpinning the co-occurrence of multiple long-term conditions, a phenomenon known as multimorbidity. To identify such variant clusters, we adapt the general method of agglomerative hierarchical clustering to multi-sample summary-data MR setup, enabling cluster detection based on variant-specific ratio estimates. Particularly, we tailor the method for multi-outcome MR to aid in elucidating the causal pathways through which a common risk factor contributes to multiple morbidities. We show in simulations that our "MR-AHC" method detects clusters with high accuracy, outperforming the existing methods. We apply the method to investigate the causal effects of high body fat percentage on type 2 diabetes and osteoarthritis, uncovering interconnected cellular processes underlying this multimorbid disease pair.
Collapse
Affiliation(s)
- Xiaoran Liang
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Ninon Mounier
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Nicolas Apfel
- Department of EconomicsUniversity of SouthamptonSouthamptonUK
| | - Sara Khalid
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUK
| | - Timothy M. Frayling
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
- Department of Genetic Medicine and Development, Faculty of MedicineCMUGenevaSwitzerland
| | - Jack Bowden
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| |
Collapse
|
4
|
Shubhrasmita Sahu S, Sarkar P, Chattopadhyay A. Quantitation of F-actin in cytoskeletal reorganization: Context, methodology and implications. Methods 2024; 230:44-58. [PMID: 39074540 DOI: 10.1016/j.ymeth.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024] Open
Abstract
The actin cytoskeleton is involved in a large number of cellular signaling events in addition to providing structural integrity to the cell. Actin polymerization is a key event during cellular signaling. Although the role of actin cytoskeleton in cellular processes such as trafficking and motility has been extensively studied, the reorganization of the actin cytoskeleton upon signaling has been rarely explored due to lack of suitable assays. Keeping in mind this lacuna, we developed a confocal microscopy based approach that relies on high magnification imaging of cellular F-actin, followed by image reconstruction using commercially available software. In this review, we discuss the context and relevance of actin quantitation, followed by a detailed hands-on approach of the methodology involved with specific points on troubleshooting and useful precautions. In the latter part of the review, we elucidate the method by discussing applications of actin quantitation from our work in several important problems in contemporary membrane biology ranging from pathogen entry into host cells, to GPCR signaling and membrane-cytoskeleton interaction. We envision that future discovery of cell-permeable novel fluorescent probes, in combination with genetically encoded actin-binding reporters, would allow real-time visualization of actin cytoskeleton dynamics to gain deeper insights into active cellular processes in health and disease.
Collapse
Affiliation(s)
- Subhashree Shubhrasmita Sahu
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Department of Biochemistry, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Amitabha Chattopadhyay
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| |
Collapse
|
5
|
Santos-Miranda A, Joviano-Santos JV, Marques ILS, Cau S, Carvalho FA, Fraga JR, Alvarez-Leite JI, Roman-Campos D, Cruz JS. Electrocontractile remodeling of isolated cardiomyocytes induced during early-stage hypercholesterolemia. J Bioenerg Biomembr 2024; 56:373-387. [PMID: 38869808 DOI: 10.1007/s10863-024-10026-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Hypercholesterolemia is one of the most important risk factors for cardiovascular diseases. However, it is mostly associated with vascular dysfunction and atherosclerotic lesions, while evidence of direct effects of hypercholesterolemia on cardiomyocytes and heart function is still incomplete and controversial. In this study, we assessed the direct effects of hypercholesterolemia on heart function and the electro-contractile properties of isolated cardiomyocytes. After 5 weeks, male Swiss mice fed with AIN-93 diet added with 1.25% cholesterol (CHO), developed an increase in total serum cholesterol levels and cardiomyocytes cholesterol content. These changes led to altered electrocardiographic records, with a shortening of the QT interval. Isolated cardiomyocytes displayed a shortening of the action potential duration with increased rate of depolarization, which was explained by increased IK, reduced ICa.L and altered INa voltage-dependent inactivation. Also, reduced diastolic [Ca2+]i was found with preserved adrenergic response and cellular contraction function. However, contraction of isolated hearts is impaired in isolated CHO hearts, before and after ischemia/reperfusion, although CHO heart was less susceptible to arrhythmic contractions. Overall, our results demonstrate that early hypercholesterolemia-driven increase in cellular cholesterol content is associated with direct modulation of the heart and cardiomyocytes' excitability, Ca2+ handling, and contraction.
Collapse
Affiliation(s)
- Artur Santos-Miranda
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, Brazil.
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil.
| | - Julliane V Joviano-Santos
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Laboratório de Investigações NeuroCardíacas, Ciências Médicas de Minas Gerais (LINC CMMG), Minas Gerais, Brazil
| | - Ivan Lobo Sousa Marques
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Stefany Cau
- Department of Pharmacology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Fabrício A Carvalho
- Department of Pharmacology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Júlia R Fraga
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | - Danilo Roman-Campos
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| |
Collapse
|
6
|
Jane A, Rasher DB, Waller J, Annis E, Frederich M. Rearing condition influences gene expression in postlarval American lobster (Homarus americanus). PLoS One 2024; 19:e0307169. [PMID: 39024318 PMCID: PMC11257295 DOI: 10.1371/journal.pone.0307169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024] Open
Abstract
The American lobster (Homarus americanus) is an economically important species in the western Atlantic and its climate-driven range shift northward along the east coast of the United States is well documented. The thermal tolerance of lab-reared postlarvae of this species has been extensively investigated to better understand settlement and recruitment dynamics. However, there have been few studies focused on wild-caught postlarvae, and even fewer that have analyzed lab-rearing conditions in context of diet. In this study, we investigated gene transcriptional changes in postlarvae caught in the wild, as well as postlarvae reared in the laboratory on a brine shrimp diet or a wild-sourced zooplankton diet. We found between wild-caught and brine shrimp-reared larvae 3,682 differentially expressed genes, and between wild and zooplankton-reared postlarvae 3,939 differentially expressed genes. Between the two lab-reared groups fed different diets 2,603 genes were differentially expressed. We also exposed individuals in all rearing groups to chronic temperature treatments of 8°C and 26°C and found that both temperature extremes elicit 68-95% fewer transcriptional changes in wild postlarvae compared to either lab-reared group. In wild postlarvae, we identified differential expression of transcripts within the FoxO signaling pathway, a signaling pathway with a central role in cellular physiology, as potential molecular markers for cold tolerance in the American lobster. These findings contextualize the current literature on lab-reared postlarvae as containing conservative estimates for in situ organisms and can be used to inform population distribution modeling efforts. They also provide evidence for the need to adjust lab-rearing techniques or source wild larval crustaceans to augment studies of larval biology.
Collapse
Affiliation(s)
- Aubrey Jane
- University of New England, School of Marine and Environmental Programs, Biddeford, Maine, United States of America
| | - Douglas B. Rasher
- Bigelow Laboratory for Ocean Sciences, East Boothbay, Maine, United States of America
| | - Jesica Waller
- Maine Department of Marine Resources, West Boothbay, Maine, United States of America
| | - Eric Annis
- Hood College, Frederick, Maryland, United States of America
| | - Markus Frederich
- University of New England, School of Marine and Environmental Programs, Biddeford, Maine, United States of America
| |
Collapse
|
7
|
Zhao Y, Yang HZ, Li H, Liang S, Wang M, Li CD, Zhuo D, Fan F, Guo M, Lv X, Zhang L, Chen X, Li SS, Jin X. Early statin exposure influences cardiac and skeletal development with implications for ion channel transcriptomes in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2024; 280:109905. [PMID: 38522713 DOI: 10.1016/j.cbpc.2024.109905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/26/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Statins, widely prescribed for cholesterol management by inhibiting HMG-CoA reductase in the cholesterol biosynthesis pathway, may also influence vertebrate development. In this study, we investigated the developmental effects of two widely used statins, atorvastatin (ATO) and pravastatin (PRA), on zebrafish offspring. For ATO, we administered doses classified as low (1 μM), medium (5 μM), and high (10 μM), while for PRA, the corresponding concentrations were set at low (18 μM), medium (180 μM), and high (270 μM). Our results showed significant reductions in birth and hatching rates, along with decreased body length in offspring at all ATO concentrations and medium to high PRA concentrations. A notable increase in malformation rates, especially in the spine and heart, was observed across all ATO treatments and in medium and high PRA groups. Additionally, we observed reduced heart contraction rates, decreased heart size, lower bone volumes, and diminished expression of mRNA osteogenic markers. Elevated venous sinus-artery bulb (SV-BA) ratios, increased thoracic area, and abnormal cartilage development were also prominent in all ATO-treated groups. Transcriptome analysis revealed alterations in genes predominantly associated with ion channels. These findings provide insights into the potential impacts of specific concentrations of statins on offspring development and highlight potential gene interactions with statins.
Collapse
Affiliation(s)
- Ying Zhao
- School of Medicine, Nankai University, Tianjin, China
| | | | - Huinan Li
- Department of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Shuang Liang
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Meng Wang
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Chun-Di Li
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Donghai Zhuo
- School of Medicine, Nankai University, Tianjin, China
| | - Feifei Fan
- School of Medicine, Nankai University, Tianjin, China
| | - Miao Guo
- School of Medicine, Nankai University, Tianjin, China
| | - Xinxin Lv
- School of Medicine, Nankai University, Tianjin, China
| | - Lingzhu Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Xu Chen
- School of Medicine, Nankai University, Tianjin, China; Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China; Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China.
| | - Shan-Shan Li
- School of Medicine, Nankai University, Tianjin, China.
| | - Xin Jin
- School of Medicine, Nankai University, Tianjin, China; Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China; Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China.
| |
Collapse
|
8
|
Kuntoji G, Kousar N, Gaddimath S, Koodlur Sannegowda L. Macromolecule-Nanoparticle-Based Hybrid Materials for Biosensor Applications. BIOSENSORS 2024; 14:277. [PMID: 38920581 PMCID: PMC11201996 DOI: 10.3390/bios14060277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024]
Abstract
Biosensors function as sophisticated devices, converting biochemical reactions into electrical signals. Contemporary emphasis on developing biosensor devices with refined sensitivity and selectivity is critical due to their extensive functional capabilities. However, a significant challenge lies in the binding affinity of biosensors to biomolecules, requiring adept conversion and amplification of interactions into various signal modalities like electrical, optical, gravimetric, and electrochemical outputs. Overcoming challenges associated with sensitivity, detection limits, response time, reproducibility, and stability is essential for efficient biosensor creation. The central aspect of the fabrication of any biosensor is focused towards forming an effective interface between the analyte electrode which significantly influences the overall biosensor quality. Polymers and macromolecular systems are favored for their distinct properties and versatile applications. Enhancing the properties and conductivity of these systems can be achieved through incorporating nanoparticles or carbonaceous moieties. Hybrid composite materials, possessing a unique combination of attributes like advanced sensitivity, selectivity, thermal stability, mechanical flexibility, biocompatibility, and tunable electrical properties, emerge as promising candidates for biosensor applications. In addition, this approach enhances the electrochemical response, signal amplification, and stability of fabricated biosensors, contributing to their effectiveness. This review predominantly explores recent advancements in utilizing macrocyclic and macromolecular conjugated systems, such as phthalocyanines, porphyrins, polymers, etc. and their hybrids, with a specific focus on signal amplification in biosensors. It comprehensively covers synthetic strategies, properties, working mechanisms, and the potential of these systems for detecting biomolecules like glucose, hydrogen peroxide, uric acid, ascorbic acid, dopamine, cholesterol, amino acids, and cancer cells. Furthermore, this review delves into the progress made, elucidating the mechanisms responsible for signal amplification. The Conclusion addresses the challenges and future directions of macromolecule-based hybrids in biosensor applications, providing a concise overview of this evolving field. The narrative emphasizes the importance of biosensor technology advancement, illustrating the role of smart design and material enhancement in improving performance across various domains.
Collapse
Affiliation(s)
| | | | | | - Lokesh Koodlur Sannegowda
- Department of Studies in Chemistry, Vijayanagara Sri Krishnadevaraya University, Jnanasagara, Vinayakanagara, Ballari 583105, India; (G.K.); (N.K.); (S.G.)
| |
Collapse
|
9
|
Casas M, Dickson EJ. Unraveling the Connection: Cholesterol, Calcium Signaling, and Neurodegeneration. Neurosci Insights 2024; 19:26331055241252772. [PMID: 38737295 PMCID: PMC11088808 DOI: 10.1177/26331055241252772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/19/2024] [Indexed: 05/14/2024] Open
Abstract
Cholesterol and calcium play crucial roles as integral structural components and functional signaling entities within the central nervous system. Disruption in cholesterol homeostasis has been linked to Alzheimer's, Parkinson's, and Huntington's Disease while alterations in calcium signaling is hypothesized to be a key substrate for neurodegeneration across many disorders. Despite the importance of regulated cholesterol and calcium homeostasis for brain health there has been an absence of research investigating the interdependence of these signaling molecules and how they can tune each other's abundance at membranes to influence membrane identity. Here, we discuss the role of cholesterol in shaping calcium dynamics in a neurodegenerative disorder that arises due to mutations in the lysosomal cholesterol transporter, Niemann Pick Type C1 (NPC1). We discuss the molecular mechanisms through which altered lysosomal cholesterol transport influences calcium signaling pathways through remodeling of ion channel distribution at organelle-organelle membrane contacts leading to neurodegeneration. This scientific inquiry not only sheds light on NPC disease but also holds implications for comprehending other cholesterol-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Maria Casas
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| |
Collapse
|
10
|
Sahranavard T, Soflaei SS, Alimi R, Pourali G, Nasrabadi M, Yadollahi A, Sharifi S, Alimi H, Shahri B, Ghalibaf AM, Metanat S, Ferns GA, Moohebati M, Ghayour-Mobarhan M. Factors associated with prolonged QTc interval in Iranian population: MASHAD cohort study. J Electrocardiol 2024; 84:112-122. [PMID: 38631278 DOI: 10.1016/j.jelectrocard.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
AIM QTc interval prolongation is a growing global issue which can cause torsades de pointes, a potentially fatal arrhythmia. We aimed to identify risk factors for prolonged QT interval in men and women. METHODS The Mashhad stroke and heart atherosclerotic disorder (MASHAD) cohort study collected electrocardiogram interval data. QT was corrected for heart rate using the Bazett's formula. Ordinal logistic regression with crude (univariable) and adjusted (multivariate) association analyses in the form of odds ratio and corresponding 95% confidence interval (CI) were used to identify the factors associated with QTc prolongation. RESULTS A total of 8878 individuals including 5318 females and 3560 males, aged 35 to 65 years, were included in this cross-sectional study. Participants with QTc prolongation were more likely to be older and have hypercholesterolemia, hypertension (HTN), and Type 2 diabetes mellitus (T2DM), but to have lower levels of physical activity (P < 0.05). Age (OR = 1.68, 95%CI = 1.18-2.39), hypercholesterolemia (OR = 1.77, 95%CI = 1.24-2.51), HTN (OR = 1.36, 95%CI = 1.06-1.73), T2DM (OR = 1.59, 95%CI = 1.19-2.13), severe anxiety (OR = 1.80, 95%CI = 1.05-3.11) and mild depression (OR = 1.38, 95%CI = 1.01-1.88) were independent risk factors for prolonged QTc interval in men. For women, only HTN (OR = 1.29, 95%CI = 1.02-1.63) and T2DM (OR = 1.50, 95%CI = 1.14-1.97) were independent risk factors. CONCLUSIONS Older age, Hypercholesterolemia, HTN, T2DM, severe anxiety and mild depression in men, and HTN and T2DM in women were associated with high risk of prolonged QTc interval. Healthcare practitioners should be aware of the risk factors of QTc interval prolongation and should exercise caution in the management of certain patients.
Collapse
Affiliation(s)
- Toktam Sahranavard
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Saffar Soflaei
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rasoul Alimi
- Department of Epidemiology and Biostatistics, School of Health, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohamad Nasrabadi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asal Yadollahi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Sharifi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hedieh Alimi
- Vascular and Endovascular Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahram Shahri
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sepehr Metanat
- Center for Orthopedic Trans-Disciplinary Applied Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex, UK
| | - Mohsen Moohebati
- Heart and Vascular Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Cardiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Ghayour-Mobarhan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Sallinger M, Grabmayr H, Humer C, Bonhenry D, Romanin C, Schindl R, Derler I. Activation mechanisms and structural dynamics of STIM proteins. J Physiol 2024; 602:1475-1507. [PMID: 36651592 DOI: 10.1113/jp283828] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
The family of stromal interaction molecules (STIM) includes two widely expressed single-pass endoplasmic reticulum (ER) transmembrane proteins and additional splice variants that act as precise ER-luminal Ca2+ sensors. STIM proteins mainly function as one of the two essential components of the so-called Ca2+ release-activated Ca2+ (CRAC) channel. The second CRAC channel component is constituted by pore-forming Orai proteins in the plasma membrane. STIM and Orai physically interact with each other to enable CRAC channel opening, which is a critical prerequisite for various downstream signalling pathways such as gene transcription or proliferation. Their activation commonly requires the emptying of the intracellular ER Ca2+ store. Using their Ca2+ sensing capabilities, STIM proteins confer this Ca2+ content-dependent signal to Orai, thereby linking Ca2+ store depletion to CRAC channel opening. Here we review the conformational dynamics occurring along the entire STIM protein upon store depletion, involving the transition from the quiescent, compactly folded structure into an active, extended state, modulation by a variety of accessory components in the cell as well as the impairment of individual steps of the STIM activation cascade associated with disease.
Collapse
Affiliation(s)
- Matthias Sallinger
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Herwig Grabmayr
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Christina Humer
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Daniel Bonhenry
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Nove Hrady, Czech Republic
| | - Christoph Romanin
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Rainer Schindl
- Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
12
|
Rout M, Mishra S, Panda S, Dehury B, Pati S. Lipid and cholesterols modulate the dynamics of SARS-CoV-2 viral ion channel ORF3a and its pathogenic variants. Int J Biol Macromol 2024; 254:127986. [PMID: 37944718 DOI: 10.1016/j.ijbiomac.2023.127986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
SARS-CoV-2 accessory protein, ORF3a is a putative ion channel which immensely contributes to viral pathogenicity by modulating host immune responses and virus-host interactions. Relatively high expression of ORF3a in diseased individuals and implication with inflammasome activation, apoptosis and autophagy inhibition, ratifies as an effective target for developing vaccines and therapeutics. Herein, we present the elusive dynamics of ORF3a-dimeric state using all-atoms molecular dynamics (MD) simulations at μ-seconds scale in a heterogeneous lipid-mimetic system in multiple replicates. Additionally, we also explore the effect of non-synonymous pathogenic mutations on ORF3a ion channel activity and viral pathogenicity in different SARS-CoV-2 variants using various structure-based protein stability (ΔΔG) tools and computational saturation mutagenesis. Our study ascertains the role of phosphatidylcholines and cholesterol in modulating the structure of ORF3a, which perturbs the size and flexibility of the polar cavity that allows permeation of large cations. Discrete trend in ion channel pore radius and area per lipid arises the premise that presence of lipids might also affect the overall conformation of ORF3a. MD structural-ensembles, in some replicates rationalize the crucial role of TM2 in maintaining the native structure of ORF3a. We also infer that loss of structural stability primarily grounds for pathogenicity in more than half of the pathogenic variants of ORF3a. Overall, the effect of mutation on alteration of ion permeability of ORF3a, proposed in this study brings mechanistic insights into variant consequences on viral membrane proteins of SARS-CoV-2, which can be utilized for the development of novel therapeutics to treat COVID-19 and other coronavirus diseases.
Collapse
Affiliation(s)
- Madhusmita Rout
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar 751023, Odisha, India
| | - Sarbani Mishra
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar 751023, Odisha, India
| | - Sunita Panda
- Mycology Division, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar 751023, Odisha, India
| | - Budheswar Dehury
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar 751023, Odisha, India.
| | - Sanghamitra Pati
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar 751023, Odisha, India.
| |
Collapse
|
13
|
Iwamoto M, Morito M, Oiki S, Nishitani Y, Yamamoto D, Matsumori N. Cardiolipin binding enhances KcsA channel gating via both its specific and dianion-monoanion interchangeable sites. iScience 2023; 26:108471. [PMID: 38077151 PMCID: PMC10709135 DOI: 10.1016/j.isci.2023.108471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/29/2023] [Accepted: 11/13/2023] [Indexed: 01/17/2024] Open
Abstract
KcsA is a potassium channel with a plethora of structural and functional information, but its activity in the KcsA-producing actinomycete membranes remains elusive. To determine lipid species involved in channel-modulation, a surface plasmon resonance (SPR)-based methodology, characterized by immobilization of membrane proteins under a membrane environment, was applied. Dianionic cardiolipin (CL) showed extremely higher affinity for KcsA than monoanionic lipids. The SPR experiments further demonstrated that CL bound not only to the N-terminal M0 helix, a lipid-sensor domain, but to the M0 helix-deleted mutant. In contrast, monoanionic lipids interacted primarily with the M0 helix. This indicates the presence of an alternative CL-binding site, plausibly in the transmembrane domain. Single-channel recordings demonstrated that CL enhanced channel opening in an M0-independent manner. Taken together, the action of monoanionic lipids is exclusively mediated by the M0 helix, while CL binds both the M0 helix and its specific site, further enhancing the channel activity.
Collapse
Affiliation(s)
- Masayuki Iwamoto
- Department of Molecular Neuroscience, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Masayuki Morito
- Department of Chemistry, Graduate School of Science, Kyushu University, Fukuoka 819-0395 Japan
| | - Shigetoshi Oiki
- Biomedial Imaging Research Center, University of Fukui, Fukui 910-1193, Japan
| | - Yudai Nishitani
- Department of Applied Physics, Faculty of Science, Fukuoka University, Fukuoka 814-0180, Japan
| | - Daisuke Yamamoto
- Department of Applied Physics, Faculty of Science, Fukuoka University, Fukuoka 814-0180, Japan
| | - Nobuaki Matsumori
- Department of Chemistry, Graduate School of Science, Kyushu University, Fukuoka 819-0395 Japan
| |
Collapse
|
14
|
Fuselier C, Dufay E, Berquand A, Terryn C, Bonnomet A, Molinari M, Martiny L, Schneider C. Dynamized ultra-low dilution of Ruta graveolens disrupts plasma membrane organization and decreases migration of melanoma cancer cell. Cell Adh Migr 2023; 17:1-13. [PMID: 36503402 PMCID: PMC9746621 DOI: 10.1080/19336918.2022.2154732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Cutaneous melanoma is a cancer with a very poor prognosis mainly because of metastatic dissemination and therefore a deregulation of cell migration. Current therapies can benefit from complementary medicines as supportive care in oncology. In our study, we show that a dynamized ultra-low dilution of Ruta Graveolens leads to an in vitro inhibition of migration on fibronectin of B16F10 melanoma cells, as well as a decrease in metastatic dissemination in vivo. These effects appear to be due to a disruption of plasma membrane organization, with a change in cell and membrane stiffness, associated with a disorganization of the actin cytoskeleton and a modification of the lipid composition of the plasma membrane. Together, these results demonstrate, in in vitro and in vivo models of cutaneous melanoma, an anti-cancer and anti-metastatic activity of ultra-low dynamized dilution of Ruta graveolens and reinforce its interest as complementary medicine in oncology.
Collapse
Affiliation(s)
- Camille Fuselier
- Center Armand-Frappier Santé Biotechnologie of the INRS, University of Quebec, Laval, Quebec, Canada
| | - Eleonore Dufay
- CNRS UMR 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | | | - Christine Terryn
- Platform PICT, University of Reims Champagne-Ardenne, Reims, France
| | - Arnaud Bonnomet
- Platform PICT, University of Reims Champagne-Ardenne, Reims, France
| | - Michael Molinari
- Institute of Chemistry & Biology of Membranes & Nano-objects, Bordeaux, France
| | - Laurent Martiny
- CNRS UMR 7369 MEDyC, University of Reims Champagne-Ardenne, Reims, France
| | | |
Collapse
|
15
|
Yamamoto K, Shimogonya Y, Maeno R, Kawabe K, Ando J. Endothelial cells differentially sense laminar and disturbed flows by altering the lipid order of their plasma and mitochondrial membranes. Am J Physiol Cell Physiol 2023; 325:C1532-C1544. [PMID: 37927239 PMCID: PMC10861177 DOI: 10.1152/ajpcell.00393.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/19/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
Endothelial cells (ECs) experience two different blood flow patterns: laminar and disturbed flow. Their responses to laminar flow contribute to vascular homeostasis, whereas their responses to disturbed flow result in EC dysfunction and vascular diseases. However, it remains unclear how ECs differentially sense laminar and disturbed flow and trigger signaling that elicits different responses. Here, we showed that ECs differentially sense laminar and disturbed flows by altering the lipid order of their plasma and mitochondrial membranes in opposite directions. This results in distinct changes in mitochondrial function, namely, increased adenosine triphosphate (ATP) production for laminar flow and increased hydrogen peroxide (H2O2) release for disturbed flow, leading to ATP- and H2O2-mediated signaling, respectively. When cultured human aortic ECs were subjected to laminar or disturbed flow in flow-loading devices, the lipid order of their plasma membranes immediately decreased in response to laminar flow and increased in response to disturbed flow. Laminar flow also decreased the lipid order of mitochondrial membranes and increased mitochondrial ATP production. In contrast, disturbed flow increased the lipid order of mitochondrial membranes and increased the release of H2O2 from the mitochondria. The addition of cholesterol to the cells increased the lipid order of both membranes and abrogated laminar flow-induced ATP production, while treatment of the cells with a cholesterol-depleting reagent, methyl-β cyclodextrin, decreased the lipid order of both membranes and abolished disturbed flow-induced H2O2 release, indicating that changes in the membrane lipid order and/or cholesterol content are closely linked to flow-induced changes in mitochondrial functions.NEW & NOTEWORTHY How vascular endothelial cells (ECs) differentially sense laminar and disturbed flows and trigger intracellular signaling remains unclear. Here, we show that EC plasma membranes act as mechanosensors to discriminate between laminar and disturbed flows by undergoing opposite changes in their lipid order. Similar lipid order changes occur simultaneously in the mitochondrial membranes, which are linked to changes in mitochondrial function, that is, increased ATP production for laminar flow and increased H2O2 release for disturbed flow.
Collapse
Affiliation(s)
- Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuji Shimogonya
- Department of Mechanical Engineering, College of Engineering, Nihon University, Koriyama, Japan
| | - Ryohei Maeno
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Vascular Surgery, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenshiroh Kawabe
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Vascular Surgery, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Joji Ando
- Laboratory of Biomedical Engineering, School of Medicine, Dokkyo Medical University, Tochigi, Japan
| |
Collapse
|
16
|
Talvio K, Wagner VA, Minkeviciene R, Kirkwood JS, Kulinich AO, Umemori J, Bhatia A, Hur M, Käkelä R, Ethell IM, Castrén ML. An iPSC-derived astrocyte model of fragile X syndrome exhibits dysregulated cholesterol homeostasis. Commun Biol 2023; 6:789. [PMID: 37516746 PMCID: PMC10387075 DOI: 10.1038/s42003-023-05147-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 07/14/2023] [Indexed: 07/31/2023] Open
Abstract
Cholesterol is an essential membrane structural component and steroid hormone precursor, and is involved in numerous signaling processes. Astrocytes regulate brain cholesterol homeostasis and they supply cholesterol to the needs of neurons. ATP-binding cassette transporter A1 (ABCA1) is the main cholesterol efflux transporter in astrocytes. Here we show dysregulated cholesterol homeostasis in astrocytes generated from human induced pluripotent stem cells (iPSCs) derived from males with fragile X syndrome (FXS), which is the most common cause of inherited intellectual disability. ABCA1 levels are reduced in FXS human and mouse astrocytes when compared with controls. Accumulation of cholesterol associates with increased desmosterol and polyunsaturated phospholipids in the lipidome of FXS mouse astrocytes. Abnormal astrocytic responses to cytokine exposure together with altered anti-inflammatory and cytokine profiles of human FXS astrocyte secretome suggest contribution of inflammatory factors to altered cholesterol homeostasis. Our results demonstrate changes of astrocytic lipid metabolism, which can critically regulate membrane properties and affect cholesterol transport in FXS astrocytes, providing target for therapy in FXS.
Collapse
Affiliation(s)
- Karo Talvio
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Victoria A Wagner
- Division of Biomedical Sciences, and Neuroscience Graduate Program, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Rimante Minkeviciene
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jay S Kirkwood
- Metabolomics Core Facility, Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, USA
| | - Anna O Kulinich
- Division of Biomedical Sciences, and Neuroscience Graduate Program, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Juzoh Umemori
- Gene and Cell Technology, A.I.Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Anil Bhatia
- Metabolomics Core Facility, Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, USA
| | - Manhoi Hur
- Metabolomics Core Facility, Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, USA
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, Biocenter Finland (Metabolomics), and Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Iryna M Ethell
- Division of Biomedical Sciences, and Neuroscience Graduate Program, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Maija L Castrén
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
17
|
Casas M, Murray KD, Hino K, Vierra NC, Simó S, Trimmer JS, Dixon RE, Dickson EJ. NPC1-dependent alterations in K V2.1-Ca V1.2 nanodomains drive neuronal death in models of Niemann-Pick Type C disease. Nat Commun 2023; 14:4553. [PMID: 37507375 PMCID: PMC10382591 DOI: 10.1038/s41467-023-39937-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Lysosomes communicate through cholesterol transfer at endoplasmic reticulum (ER) contact sites. At these sites, the Niemann Pick C1 cholesterol transporter (NPC1) facilitates the removal of cholesterol from lysosomes, which is then transferred to the ER for distribution to other cell membranes. Mutations in NPC1 result in cholesterol buildup within lysosomes, leading to Niemann-Pick Type C (NPC) disease, a progressive and fatal neurodegenerative disorder. The molecular mechanisms connecting NPC1 loss to NPC-associated neuropathology remain unknown. Here we show both in vitro and in an animal model of NPC disease that the loss of NPC1 function alters the distribution and activity of voltage-gated calcium channels (CaV). Underlying alterations in calcium channel localization and function are KV2.1 channels whose interactions drive calcium channel clustering to enhance calcium entry and fuel neurotoxic elevations in mitochondrial calcium. Targeted disruption of KV2-CaV interactions rescues aberrant CaV1.2 clustering, elevated mitochondrial calcium, and neurotoxicity in vitro. Our findings provide evidence that NPC is a nanostructural ion channel clustering disease, characterized by altered distribution and activity of ion channels at membrane contacts, which contribute to neurodegeneration.
Collapse
Affiliation(s)
- Maria Casas
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Karl D Murray
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
- Department of Psychiatry & Behavioral Sciences, School of Medicine, University of California, Davis, CA, USA
| | - Keiko Hino
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - Nicholas C Vierra
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - James S Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
18
|
Banerjee KK, Maity P, Das S, Karmakar S. Effect of cholesterol on the ion-membrane interaction: Zeta potential and dynamic light scattering study. Chem Phys Lipids 2023; 254:105307. [PMID: 37182823 DOI: 10.1016/j.chemphyslip.2023.105307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/11/2023] [Indexed: 05/16/2023]
Abstract
Cholesterol in a bio-membrane plays a significant role in many cellular event and is known to regulate the functional activity of protein and ion channel. In this study we report a significant effect of cholesterol on the ion-membrane interaction. We prepare large unilamellar vesicles, composed of zwitterionic lipid DOPC and anionic lipid DOPG with different cholesterol concentration. Electrostatics of anionic membranes containing cholesterol in the presence of NaCl has systematically been explored using dynamic light scattering and zeta potential. Negative zeta potential of the membrane decreases its negative value with increasing ion concentration for all cholesterol concentrations. However, zeta potential itself decreases with increasing cholesterol content even in the absence of monovalent ions. Electrostatic behaviour of the membrane is determined from well-known Gouy Chapmann model. Negative surface charge density of the membrane decreases with increasing cholesterol content. Binding constant, estimated from the electrostatic double layer theory, is found to increase significantly in the presence of cholesterol. Comparison of electrostatic parameters of the membrane in the presence and absence of cholesterol suggests that cholesterol significantly alter the electrostatic behaviour of the membrane.
Collapse
Affiliation(s)
- Kalyan Kumar Banerjee
- Soft matter and Biophysics Laboratory, Department of Physics, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Pabitra Maity
- Soft matter and Biophysics Laboratory, Department of Physics, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Surajit Das
- Soft matter and Biophysics Laboratory, Department of Physics, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata 700032, India
| | - Sanat Karmakar
- Soft matter and Biophysics Laboratory, Department of Physics, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata 700032, India.
| |
Collapse
|
19
|
Milton EM, Cartolano MC, McDonald MD. A multi-targeted investigation of Deepwater Horizon crude oil exposure impacts on the marine teleost stress axis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 257:106444. [PMID: 36848692 DOI: 10.1016/j.aquatox.2023.106444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/09/2023] [Accepted: 02/18/2023] [Indexed: 06/18/2023]
Abstract
The toxicity of the polycyclic aromatic hydrocarbons (PAHs) in Deepwater Horizon (DWH) oil is well-established, but a knowledge gap exists regarding how this combination of PAHs affects the vertebrate stress axis. We hypothesized that (1) marine vertebrates exposed to DWH PAHs experience stress axis impairment, and co-exposure to an additional chronic stressor may exacerbate these effects, (2) serotonin (5-hydroxytryptamine; 5-HT) may act as a secondary cortisol secretagogue in DWH PAH-exposed fish to compensate for impairment, and (3) the mechanism of stress axis impairment may involve downregulation of cyclic adenosine monophosphate (cAMP; as proxy for melanocortin 2 receptor (MC2R) functionality), total cholesterol, and/or mRNA expression of CYP1A and steroidogenic proteins StAR, P450scc, and 11β-h at the level of the kidney. We found that in vivo plasma cortisol and plasma adrenocorticotropic hormone (ACTH) concentrations in Gulf toadfish exposed to an environmentally relevant DWH PAH concentration (ΣPAH50= 4.6 ± 1.6 μg/L) for 7 days were not significantly different from controls, whether fish were chronically stressed or not. However, the rate of cortisol secretion by isolated kidneys after acute stimulation with ACTH was significantly lower in PAH-exposed toadfish compared to clean seawater (SW) controls. 5-HT does not appear to be acting as a secondary cortisol secretagogue, rather, PAH-exposed + stressed toadfish exhibited significantly lower plasma 5-HT concentrations than clean SW + stressed fish as well as a reduced sensitivity to 5-HT at the level of the kidney. There was a tendency for kidney cAMP concentrations to be lower in PAH-exposed fish (p = 0.069); however, mRNA expression of steroidogenic proteins between control and PAH-exposed toadfish were not significantly different and a significant elevation in total cholesterol concentration in PAH-exposed toadfish compared to controls was measured. Future work is needed to establish whether the slower cortisol secretion rate by isolated kidneys of PAH-exposed fish is detrimental, to determine the potential role of other secretagogues in compensating for the impaired kidney interrenal cell function, and to determine whether there is a reduction in MC2R mRNA expression or an impairment in the function of steroidogenic proteins.
Collapse
Affiliation(s)
- Emily M Milton
- Department of Marine Biology and Ecology, University of Miami Rosenstiel School of Marine, Atmospheric, and Earth Science, 4600 Rickenbacker Causeway, Miami, FL 33149-1098, USA
| | - Maria C Cartolano
- Department of Marine Biology and Ecology, University of Miami Rosenstiel School of Marine, Atmospheric, and Earth Science, 4600 Rickenbacker Causeway, Miami, FL 33149-1098, USA
| | - M Danielle McDonald
- Department of Marine Biology and Ecology, University of Miami Rosenstiel School of Marine, Atmospheric, and Earth Science, 4600 Rickenbacker Causeway, Miami, FL 33149-1098, USA.
| |
Collapse
|
20
|
Arreola J, López-Romero AE, Pérez-Cornejo P, Rodríguez-Menchaca AA. Phosphatidylinositol 4,5-Bisphosphate and Cholesterol Regulators of the Calcium-Activated Chloride Channels TMEM16A and TMEM16B. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:279-304. [PMID: 36988885 DOI: 10.1007/978-3-031-21547-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Chloride fluxes through homo-dimeric calcium-activated channels TMEM16A and TMEM16B are critical to blood pressure, gastrointestinal motility, hormone, fluid and electrolyte secretion, pain sensation, sensory transduction, and neuronal and muscle excitability. Their gating depends on the voltage-dependent binding of two intracellular calcium ions to a high-affinity site formed by acidic residues from α-helices 6-8 in each monomer. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), a low-abundant lipid of the inner leaflet, supports TMEM16A function; it allows TMEM16A to evade the down-regulation induced by calcium, poly-L-lysine, or PI(4,5)P2 5-phosphatase. In stark contrast, adding or removing PI(4,5)P2 diminishes or increases TMEM16B function, respectively. PI(4,5)P2-binding sites on TMEM16A, and presumably on TMEM16B, are on the cytosolic side of α-helices 3-5, opposite the calcium-binding sites. This modular structure suggested that PI(4,5)P2 and calcium cooperate to maintain the conductive state in TMEM16A. Cholesterol, the second-largest constituent of the plasma membrane, also regulates TMEM16A though the mechanism, functional outcomes, binding site(s), and effects on TMEM16A and TMEM16B remain unknown.
Collapse
Affiliation(s)
- Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | | | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Aldo A Rodríguez-Menchaca
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| |
Collapse
|
21
|
Enrich C, Lu A, Tebar F, Rentero C, Grewal T. Ca 2+ and Annexins - Emerging Players for Sensing and Transferring Cholesterol and Phosphoinositides via Membrane Contact Sites. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:393-438. [PMID: 36988890 DOI: 10.1007/978-3-031-21547-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Maintaining lipid composition diversity in membranes from different organelles is critical for numerous cellular processes. However, many lipids are synthesized in the endoplasmic reticulum (ER) and require delivery to other organelles. In this scenario, formation of membrane contact sites (MCS) between neighbouring organelles has emerged as a novel non-vesicular lipid transport mechanism. Dissecting the molecular composition of MCS identified phosphoinositides (PIs), cholesterol, scaffolding/tethering proteins as well as Ca2+ and Ca2+-binding proteins contributing to MCS functioning. Compelling evidence now exists for the shuttling of PIs and cholesterol across MCS, affecting their concentrations in distinct membrane domains and diverse roles in membrane trafficking. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) at the plasma membrane (PM) not only controls endo-/exocytic membrane dynamics but is also critical in autophagy. Cholesterol is highly concentrated at the PM and enriched in recycling endosomes and Golgi membranes. MCS-mediated cholesterol transfer is intensely researched, identifying MCS dysfunction or altered MCS partnerships to correlate with de-regulated cellular cholesterol homeostasis and pathologies. Annexins, a conserved family of Ca2+-dependent phospholipid binding proteins, contribute to tethering and untethering events at MCS. In this chapter, we will discuss how Ca2+ homeostasis and annexins in the endocytic compartment affect the sensing and transfer of cholesterol and PIs across MCS.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Albert Lu
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel⋅lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Dickson EJ. Role of Lysosomal Cholesterol in Regulating PI(4,5)P 2-Dependent Ion Channel Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:193-215. [PMID: 36988882 DOI: 10.1007/978-3-031-21547-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Lysosomes are central regulators of cellular growth and signaling. Once considered the acidic garbage can of the cell, their ever-expanding repertoire of functions include the regulation of cell growth, gene regulation, metabolic signaling, cell migration, and cell death. In this chapter, we detail how another of the lysosome's crucial roles, cholesterol transport, plays a vital role in the control of ion channel function and neuronal excitability through its ability to influence the abundance of the plasma membrane signaling lipid phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). This chapter will introduce the biosynthetic pathways of cholesterol and PI(4,5)P2, discuss the molecular mechanisms through which each lipid distinctly regulates ion channels, and consider the interdependence of these lipids in the control of ion channel function.
Collapse
Affiliation(s)
- Eamonn J Dickson
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.
| |
Collapse
|
23
|
Weber-Boyvat M, Kroll J, Trimbuch T, Olkkonen VM, Rosenmund C. The lipid transporter ORP2 regulates synaptic neurotransmitter release via two distinct mechanisms. Cell Rep 2022; 41:111882. [PMID: 36577376 DOI: 10.1016/j.celrep.2022.111882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/26/2022] [Accepted: 12/02/2022] [Indexed: 12/28/2022] Open
Abstract
Cholesterol is crucial for neuronal synaptic transmission, assisting in the molecular and structural organization of lipid rafts, ion channels, and exocytic proteins. Although cholesterol absence was shown to result in impaired neurotransmission, how cholesterol locally traffics and its route of action are still under debate. Here, we characterized the lipid transfer protein ORP2 in murine hippocampal neurons. We show that ORP2 preferentially localizes to the presynapse. Loss of ORP2 reduces presynaptic cholesterol levels by 50%, coinciding with a profoundly reduced release probability, enhanced facilitation, and impaired presynaptic calcium influx. In addition, ORP2 plays a cholesterol-transport-independent role in regulating vesicle priming and spontaneous release, likely by competing with Munc18-1 in syntaxin1A binding. To conclude, we identified a dual function of ORP2 as a physiological modulator of the synaptic cholesterol content and a regulator of neuronal exocytosis.
Collapse
Affiliation(s)
- Marion Weber-Boyvat
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jana Kroll
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Thorsten Trimbuch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Christian Rosenmund
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
24
|
Tiffner A, Hopl V, Derler I. CRAC and SK Channels: Their Molecular Mechanisms Associated with Cancer Cell Development. Cancers (Basel) 2022; 15:101. [PMID: 36612099 PMCID: PMC9817886 DOI: 10.3390/cancers15010101] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer represents a major health burden worldwide. Several molecular targets have been discovered alongside treatments with positive clinical outcomes. However, the reoccurrence of cancer due to therapy resistance remains the primary cause of mortality. Endeavors in pinpointing new markers as molecular targets in cancer therapy are highly desired. The significance of the co-regulation of Ca2+-permeating and Ca2+-regulated ion channels in cancer cell development, proliferation, and migration make them promising molecular targets in cancer therapy. In particular, the co-regulation of the Orai1 and SK3 channels has been well-studied in breast and colon cancer cells, where it finally leads to an invasion-metastasis cascade. Nevertheless, many questions remain unanswered, such as which key molecular components determine and regulate their interplay. To provide a solid foundation for a better understanding of this ion channel co-regulation in cancer, we first shed light on the physiological role of Ca2+ and how this ion is linked to carcinogenesis. Then, we highlight the structure/function relationship of Orai1 and SK3, both individually and in concert, their role in the development of different types of cancer, and aspects that are not yet known in this context.
Collapse
Affiliation(s)
- Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| |
Collapse
|
25
|
Szczurowska E, Szánti-Pintér E, Randáková A, Jakubík J, Kudova E. Allosteric Modulation of Muscarinic Receptors by Cholesterol, Neurosteroids and Neuroactive Steroids. Int J Mol Sci 2022; 23:13075. [PMID: 36361865 PMCID: PMC9656441 DOI: 10.3390/ijms232113075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2023] Open
Abstract
Muscarinic acetylcholine receptors are membrane receptors involved in many physiological processes. Malfunction of muscarinic signaling is a cause of various internal diseases, as well as psychiatric and neurologic conditions. Cholesterol, neurosteroids, neuroactive steroids, and steroid hormones are molecules of steroid origin that, besides having well-known genomic effects, also modulate membrane proteins including muscarinic acetylcholine receptors. Here, we review current knowledge on the allosteric modulation of muscarinic receptors by these steroids. We give a perspective on the research on the non-genomic effects of steroidal compounds on muscarinic receptors and drug development, with an aim to ultimately exploit such knowledge.
Collapse
Affiliation(s)
- Ewa Szczurowska
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 6, 166 10 Prague, Czech Republic
| | - Eszter Szánti-Pintér
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 6, 166 10 Prague, Czech Republic
| | - Alena Randáková
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Jan Jakubík
- Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Namesti 2, Prague 6, 166 10 Prague, Czech Republic
| |
Collapse
|
26
|
Schneider EH, Fitzgerald AC, Ponnapula SS, Dopico AM, Bukiya AN. Differential distribution of cholesterol pools across arteries under high-cholesterol diet. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159235. [PMID: 36113825 DOI: 10.1016/j.bbalip.2022.159235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/26/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
Excessive cholesterol constitutes a major risk factor for vascular disease. Within cells, cholesterol is distributed in detergent-sensitive and detergent-resistant fractions, with the largest amount of cholesterol residing in cellular membranes. We set out to determine whether various arteries differ in their ability to accumulate esterified and non-esterified cholesterol in detergent-sensitive versus detergent-resistant fractions throughout the course of a high-cholesterol diet. Male Sprague-Dawley rats were placed on 2 % cholesterol diet while a control group was receiving iso-caloric standard chow. Liver, aorta, and pulmonary, mesenteric, and cerebral arteries were collected at 2-6, 8-12, 14-18, and 20-24 weeks from the start of high-cholesterol diet. After fraction separation, esterified and free non-esterified cholesterol levels were measured. In all arteries, largest cholesterol amounts were present in detergent-sensitive fractions in the non-esterified form. Overall, cholesterol in aorta and cerebral arteries was elevated during 14-18 weeks of high-cholesterol diet. Cerebral arteries also exhibited increase in esterified cholesterol within detergent-sensitive domains, as well as increase in cholesterol level in the detergent-resistant fraction at earlier time-points of diet. Pulmonary artery and mesenteric artery were largely resistant to cholesterol accumulation. Quantitative polymerase chain reaction (qPCR) analysis revealed up-regulation of low-density lipoprotein receptor (Ldlr) and low-density lipoprotein receptor-related protein 1 (Lrp1) gene expression in cerebral arteries when compared to mesenteric and pulmonary arteries, respectively. In summary, we unveiled the differential ability of arteries to accumulate cholesterol over the course of a high-cholesterol diet. The differential accumulation of cholesterol seems to correlate with the up-regulated gene expression of proteins responsible for cholesterol uptake.
Collapse
Affiliation(s)
- Elizabeth H Schneider
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Amanda C Fitzgerald
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Supriya Suzy Ponnapula
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
27
|
Rezende L, Couto NFD, Fernandes-Braga W, Epshtein Y, Alvarez-Leite JI, Levitan I, Andrade LDO. OxLDL induces membrane structure rearrangement leading to biomechanics alteration and migration deficiency in macrophage. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183951. [PMID: 35504320 DOI: 10.1016/j.bbamem.2022.183951] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022]
Abstract
Cholesterol sequestration from plasma membrane has been shown to induce lipid packing disruption, causing actin cytoskeleton reorganization and polymerization, increasing cell stiffness and inducing lysosomal exocytosis in non-professional phagocytes. Similarly, oxidized form of low-density lipoprotein (oxLDL) has also been shown to disrupt lipid organization and packing in endothelial cells, leading to biomechanics alterations that interfere with membrane injury and repair. For macrophages, much is known about oxLDL effects in cell activation, cytokine production and foam cell formation. However, little is known about its impact in the organization of macrophage membrane structured domains and cellular mechanics, the focus of the present study. Treatment of bone marrow-derived macrophages (BMDM) with oxLDL not only altered membrane structure, and potentially the distribution of raft domains, but also induced actin rearrangement, diffuse integrin distribution and cell shrinkage, similarly to observed upon treatment of these cells with MβCD. Those alterations led to decreased migration efficiency. For both treatments, higher co-localization of actin cytoskeleton and GM1 was observed, indicating a similar mechanism of action involving raft-like domain dynamics. Lastly, like MβCD treatment, oxLDL also induced lysosomal spreading in BMDM. We propose that OxLDL induced re-organization of membrane/cytoskeleton complex in macrophages can be attributed to the insertion of oxysterols into the membrane, which lead to changes in lipid organization and disruption of membrane structure, similar to the effect of cholesterol depletion by MβCD treatment. These results indicate that oxLDL can induce physical alterations in the complex membrane/cytoskeleton of macrophages, leading to significant biomechanical changes that compromise cell behavior.
Collapse
Affiliation(s)
- Luisa Rezende
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natalia Fernanda Do Couto
- Department of Morphology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Weslley Fernandes-Braga
- Department of Biochemistry and Immunology/Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Yulia Epshtein
- Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | | | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | | |
Collapse
|
28
|
Kim HS, Suh JS, Jang YK, Ahn SH, Choi GH, Yang JY, Lim GH, Jung Y, Jiang J, Sun J, Suk M, Wang Y, Kim TJ. Förster Resonance Energy Transfer-Based Single-Cell Imaging Reveals Piezo1-Induced Ca 2+ Flux Mediates Membrane Ruffling and Cell Survival. Front Cell Dev Biol 2022; 10:865056. [PMID: 35646889 PMCID: PMC9136143 DOI: 10.3389/fcell.2022.865056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/25/2022] [Indexed: 01/18/2023] Open
Abstract
A mechanosensitive ion channel, Piezo1 induces non-selective cation flux in response to various mechanical stresses. However, the biological interpretation and underlying mechanisms of cells resulting from Piezo1 activation remain elusive. This study elucidates Piezo1-mediated Ca2+ influx driven by channel activation and cellular behavior using novel Förster Resonance Energy Transfer (FRET)-based biosensors and single-cell imaging analysis. Results reveal that extracellular Ca2+ influx via Piezo1 requires intact caveolin, cholesterol, and cytoskeletal support. Increased cytoplasmic Ca2+ levels enhance PKA, ERK, Rac1, and ROCK activity, which have the potential to promote cancer cell survival and migration. Furthermore, we demonstrate that Piezo1-mediated Ca2+ influx upregulates membrane ruffling, a characteristic feature of cancer cell metastasis, using spatiotemporal image correlation spectroscopy. Thus, our findings provide new insights into the function of Piezo1, suggesting that Piezo1 plays a significant role in the behavior of cancer cells.
Collapse
Affiliation(s)
- Heon-Su Kim
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea,Institute of Systems Biology, Pusan National University, Pusan, South Korea
| | - Jung-Soo Suh
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Yoon-Kwan Jang
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Sang-Hyun Ahn
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Gyu-Ho Choi
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Jin-Young Yang
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Gah-Hyun Lim
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Youngmi Jung
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea
| | - Jie Jiang
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Sun
- Department of Cell Biology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Myungeun Suk
- Department of Mechanical Engineering, Dong-Eui University, Pusan, South Korea
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Tae-Jin Kim
- Department of Integrated Biological Science, Pusan National University, Pusan, South Korea,Institute of Systems Biology, Pusan National University, Pusan, South Korea,Department of Biological Sciences, Pusan National University, Pusan, South Korea,*Correspondence: Tae-Jin Kim,
| |
Collapse
|
29
|
Hudgins EC, Bonar AM, Nguyen T, Fancher IS. Targeting Lipid—Ion Channel Interactions in Cardiovascular Disease. Front Cardiovasc Med 2022; 9:876634. [PMID: 35600482 PMCID: PMC9120415 DOI: 10.3389/fcvm.2022.876634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
General lipid-lowering strategies exhibit clinical benefit, however, adverse effects and low adherence of relevant pharmacotherapies warrants the investigation into distinct avenues for preventing dyslipidemia-induced cardiovascular disease. Ion channels play an important role in the maintenance of vascular tone, the impairment of which is a critical precursor to disease progression. Recent evidence suggests that the dysregulation of ion channel function in dyslipidemia is one of many contributors to the advancement of cardiovascular disease thus bringing to light a novel yet putative therapeutic avenue for preventing the progression of disease mechanisms. Increasing evidence suggests that lipid regulation of ion channels often occurs through direct binding of the lipid with the ion channel thereby creating a potential therapeutic target wherein preventing specific lipid-ion channel interactions, perhaps in combination with established lipid lowering therapies, may restore ion channel function and the proper control of vascular tone. Here we first detail specific examples of lipid-ion channel interactions that promote vascular dysfunction and highlight the benefits of preventing such interactions. We next discuss the putative therapeutic avenues, such as peptides, monoclonal antibodies, and aspects of nanomedicine that may be utilized to prevent pathological lipid-ion channel interactions. Finally, we discuss the experimental challenges with identifying lipid-ion channel interactions as well as the likely pitfalls with developing the aforementioned putative strategies.
Collapse
|
30
|
Dong C, Liu W, Zhang Y, Song Y, Du J, Huang Z, Wang T, Yu Z, Ma X. Identification of Common Hub Genes in Human Dermal Fibroblasts Stimulated by Mechanical Stretch at Both the Early and Late Stages. Front Surg 2022; 9:846161. [PMID: 35510126 PMCID: PMC9058084 DOI: 10.3389/fsurg.2022.846161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Background Mechanical stretch is vital for soft tissue regeneration and development and is utilized by plastic surgeons for tissue expansion. Identifying the common hub genes in human dermal fibroblasts (HDFs) stimulated by mechanical stretch at different stages will help elucidate the mechanisms involved and improve the efficiency of tissue expansion. Methods A gene expression dataset (GSE58389) was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) in HDFs between cyclic mechanical stretching and static samples were identified at 5 and 24 h. Common DEGs overlapped in both the 5 h and 24 h groups. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to determine the functions of the DEGs. Protein-protein interaction networks were constructed using the STRING database. The top 10 hub genes were selected using the plug-in Cytohubba within Cytoscape. The regulatory network of hub genes was predicted using NetworkAnalyst. Results A total of 669 and 249 DEGs were identified at the early (5 h) and late stages (24 h), respectively. Of these, 152 were present at both stages and were designated as common DEGs. The top enriched GO terms were “regulation of autophagy” at the early stage, and “sterol biosynthetic processes” at the late stage. The top KEGG terms were “pyrimidine metabolism” and “synaptic vesicle cycle” at the early and late stages, respectively. Seven common DEGs [DEAD-box helicase 17 (DDX17), exocyst complex component 7 (EXOC7), CASK interacting protein 1 (CASKIN1), ribonucleoprotein PTB-binding 1 (RAVER1), late cornified envelope 1D (LCE1D), LCE1C, and polycystin 1, transient receptor potential channel interacting (PKD1)] and three common DEGs [5′-3′ exoribonuclease 2 (XRN2), T-complex protein 1 (TCP1), and syntaxin 3 (STX3)] were shown to be upregulated and downregulated hub genes, respectively. The GO terms of the common hub genes were “skin development” and “mRNA processing.” After constructing the regulatory network, hsa-mir-92a-3p, hsa-mir-193b-3p, RNA polymerase II subunit A (POLR2A), SMAD family member 5 (SMAD5), and MYC-associated zinc finger protein (MAZ) were predicted as potential targets in both stages. Conclusion At the early stage, there were clear changes in gene expression related to DNA and chromatin alterations; at late stages, gene expression associated with cholesterol metabolism was suppressed. Common DEGs related to skin development, transcriptional regulation, and cytoskeleton rearrangement identified in both stages were found to be potential targets for promoting HDF growth and alignment under mechanical stretch.
Collapse
|
31
|
North KC, Zhang M, Singh AK, Zaytseva D, Slayden AV, Bukiya AN, Dopico AM. Cholesterol Inhibition of Slo1 Channels Is Calcium-Dependent and Can Be Mediated by Either High-Affinity Calcium-Sensing Site in the Slo1 Cytosolic Tail. Mol Pharmacol 2022; 101:132-143. [PMID: 34969832 PMCID: PMC8969144 DOI: 10.1124/molpharm.121.000392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/27/2021] [Indexed: 11/22/2022] Open
Abstract
Calcium- and voltage-gated K+ channels of large conductance (BKs) are expressed in the cell membranes of all excitable tissues. Currents mediated by BK channel-forming slo1 homotetramers are consistently inhibited by increases in membrane cholesterol (CLR). The molecular mechanisms leading to this CLR action, however, remain unknown. Slo1 channels are activated by increases in calcium (Ca2+) nearby Ca2+-recognition sites in the slo1 cytosolic tail: one high-affinity and one low-affinity site locate to the regulator of conductance for K+ (RCK) 1 domain, whereas another high-affinity site locates within the RCK2 domain. Here, we first evaluated the crosstalking between Ca2+ and CLR on the function of slo1 (cbv1 isoform) channels reconstituted into planar lipid bilayers. CLR robustly reduced channel open probability while barely decreasing unitary current amplitude, with CLR maximal effects being observed at 10-30 µM internal Ca2+ CLR actions were not only modulated by internal Ca2+ levels but also disappeared in absence of this divalent. Moreover, in absence of Ca2+, BK channel-activating concentrations of magnesium (10 mM) did not support CLR action. Next, we evaluated CLR actions on channels where the different Ca2+-sensing sites present in the slo1 cytosolic domain became nonfunctional via mutagenesis. CLR still reduced the activity of low-affinity Ca2+ (RCK1:E379A, E404A) mutants. In contrast, CLR became inefficacious when both high-affinity Ca2+ sites were mutated (RCK1:D367A,D372A and RCK2:D899N,D900N,D901N,D902N,D903N), yet still was able to decrease the activity of each high-affinity site mutant. Therefore, BK channel inhibition by CLR selectively requires optimal levels of Ca2+ being recognized by either of the slo1 high-affinity Ca2+-sensing sites. SIGNIFICANCE STATEMENT: Results reveal that inhibition of calcium/voltage-gated K+ channel of large conductance (BK) (slo1) channels by membrane cholesterol requires a physiologically range of internal calcium (Ca2+) and is selectively linked to the two high-affinity Ca2+-sensing sites located in the cytosolic tail domain, which underscores that Ca2+ and cholesterol actions are allosterically coupled to the channel gate. Cholesterol modification of BK channel activity likely contributes to disruption of normal physiology by common health conditions that are triggered by disruption of cholesterol homeostasis.
Collapse
Affiliation(s)
- Kelsey C North
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Man Zhang
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Aditya K Singh
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Dasha Zaytseva
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alexandria V Slayden
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
32
|
Ando J, Yamamoto K. Hemodynamic Forces, Endothelial Mechanotransduction, and Vascular Diseases. Magn Reson Med Sci 2022; 21:258-266. [PMID: 34024868 PMCID: PMC9680547 DOI: 10.2463/mrms.rev.2021-0018] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/23/2021] [Indexed: 11/09/2022] Open
Abstract
Cells in the tissues and organs of a living body are subjected to mechanical forces, such as pressure, friction, and tension from their surrounding environment. Cells are equipped with a mechanotransduction mechanism by which they perceive mechanical forces and transmit information into the cell interior, thereby causing physiological or pathogenetic mechano-responses. Endothelial cells (ECs) lining the inner surface of blood vessels are constantly exposed to shear stress caused by blood flow and a cyclic strain caused by intravascular pressure. A number of studies have shown that ECs are sensitive to changes in these hemodynamic forces and alter their morphology and function, sometimes by modifying gene expression. The mechanism of endothelial mechanotransduction has been elucidated, and the plasma membrane has recently been shown to act as a mechanosensor. The lipid order and cholesterol content of plasma membranes change immediately upon the exposure of ECs to hemodynamic forces, resulting in a change in membrane fluidity. These changes in a plasma membrane's physical properties affect the conformation and function of various ion channels, receptors, and microdomains (such as caveolae and primary cilia), thereby activating a wide variety of downstream signaling pathways. Such endothelial mechanotransduction works to maintain circulatory homeostasis; however, errors in endothelial mechanotransduction can cause abnormalities in vascular physiological function, leading to the initiation and progression of various vascular diseases, such as hypertension, thrombosis, aneurysms, and atherosclerosis. Recent advances in detailed imaging technology and computational fluid dynamics analysis have enabled us to evaluate the hemodynamic forces acting on vascular tissue accurately, contributing greatly to our understanding of vascular mechanotransduction and the pathogenesis of vascular diseases, as well as the development of new therapies for vascular diseases.
Collapse
Affiliation(s)
- Joji Ando
- Laboratory of Biomedical Engineering, School of Medicine, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
33
|
Martínez-Campelo L, Cruz R, Blanco-Verea A, Moscoso I, Ramos-Luis E, Lage R, Álvarez-Barredo M, Sabater-Molina M, Peñafiel-Verdú P, Jiménez-Jáimez J, Rodríguez-Mañero M, Brion M. Searching for genetic modulators of the phenotypic heterogeneity in Brugada syndrome. PLoS One 2022; 17:e0263469. [PMID: 35231055 PMCID: PMC8887717 DOI: 10.1371/journal.pone.0263469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/20/2022] [Indexed: 11/19/2022] Open
Abstract
In Brugada syndrome, even within the same family where all affected individuals share the same mutation, phenotypic variation is prominent, with variable penetrance and expressivity, presenting different degrees of involvement. It is difficult to establish a direct correlation between genotype and phenotype to predict prognosis in complications and risk of sudden death. The factors that modulate this inter- and intra-familial phenotypic variability remain to be determined. With the intention of testing whether other genetic factors, in addition to the causal mutation in SCN5A, may have a modulating effect on the Brugada phenotype and the risk of sudden death, we have studied 8 families with a causal variant in SCN5A with at least two affected individuals, one of whom has suffered cardiac arrest or sudden death. Whole exome sequencing was performed looking for additional variants that modify the phenotype and allow us to predict a better or worse prognosis for the evolution of the disease. The results did not show any clear genetic modifier; nevertheless, highlight the possible implication of the cholesterol and fibrosis pathways, as well as the circadian rhythm, as possible modulators of Brugada syndrome phenotype.
Collapse
Affiliation(s)
- Laura Martínez-Campelo
- Cardiovascular Genetics, Santiago de Compostela Health Research Institute, Santiago de Compostela, Spain
- Genomic Medicine Group, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Raquel Cruz
- Genomic Medicine Group, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- CIBER of Rare Diseases, Carlos III Health Institute, Madrid, Spain
| | - Alejandro Blanco-Verea
- Cardiovascular Genetics, Santiago de Compostela Health Research Institute, Santiago de Compostela, Spain
- Genomic Medicine Group, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Isabel Moscoso
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Eva Ramos-Luis
- Cardiovascular Genetics, Santiago de Compostela Health Research Institute, Santiago de Compostela, Spain
- Genomic Medicine Group, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ricardo Lage
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María Álvarez-Barredo
- Cardiovascular Genetics, Santiago de Compostela Health Research Institute, Santiago de Compostela, Spain
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
| | - María Sabater-Molina
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
- Cardiogenetics Laboratory, Murcian Institute for Biosanitary Research, Cardiology Service, Virgen de la Arrixaca University Clinical Hospital, Murcia, Spain
| | - Pablo Peñafiel-Verdú
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
- Cardiogenetics Laboratory, Murcian Institute for Biosanitary Research, Cardiology Service, Virgen de la Arrixaca University Clinical Hospital, Murcia, Spain
| | - Juan Jiménez-Jáimez
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
- Arrhythmia Unit, Virgen de las Nieves University Hospital, Granada, Spain
| | - Moisés Rodríguez-Mañero
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
- Cardiology Service, Santiago de Compostela University Hospital, Santiago de Compostela, Spain
| | - María Brion
- Cardiovascular Genetics, Santiago de Compostela Health Research Institute, Santiago de Compostela, Spain
- Genomic Medicine Group, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Cardiovascular CIBER, Carlos III Health Institute, Madrid, Spain
- Family Heart Disease Unit, Cardiology Service, Santiago de Compostela University Hospital, Santiago de Compostela, Spain
| |
Collapse
|
34
|
Maltan L, Andova AM, Derler I. The Role of Lipids in CRAC Channel Function. Biomolecules 2022; 12:biom12030352. [PMID: 35327543 PMCID: PMC8944985 DOI: 10.3390/biom12030352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/12/2022] [Accepted: 02/20/2022] [Indexed: 11/28/2022] Open
Abstract
The composition and dynamics of the lipid membrane define the physical properties of the bilayer and consequently affect the function of the incorporated membrane transporters, which also applies for the prominent Ca2+ release-activated Ca2+ ion channel (CRAC). This channel is activated by receptor-induced Ca2+ store depletion of the endoplasmic reticulum (ER) and consists of two transmembrane proteins, STIM1 and Orai1. STIM1 is anchored in the ER membrane and senses changes in the ER luminal Ca2+ concentration. Orai1 is the Ca2+-selective, pore-forming CRAC channel component located in the plasma membrane (PM). Ca2+ store-depletion of the ER triggers activation of STIM1 proteins, which subsequently leads to a conformational change and oligomerization of STIM1 and its coupling to as well as activation of Orai1 channels at the ER-PM contact sites. Although STIM1 and Orai1 are sufficient for CRAC channel activation, their efficient activation and deactivation is fine-tuned by a variety of lipids and lipid- and/or ER-PM junction-dependent accessory proteins. The underlying mechanisms for lipid-mediated CRAC channel modulation as well as the still open questions, are presented in this review.
Collapse
|
35
|
Brain cell type-specific cholesterol metabolism and implications for learning and memory. Trends Neurosci 2022; 45:401-414. [DOI: 10.1016/j.tins.2022.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/06/2022] [Accepted: 01/25/2022] [Indexed: 12/21/2022]
|
36
|
Luo M, Cai G, Ho KKY, Wen K, Tong Z, Deng L, Liu AP. Compression enhances invasive phenotype and matrix degradation of breast Cancer cells via Piezo1 activation. BMC Mol Cell Biol 2022; 23:1. [PMID: 34979904 PMCID: PMC8722159 DOI: 10.1186/s12860-021-00401-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
Background Uncontrolled growth in solid breast cancer generates mechanical compression that may drive the cancer cells into a more invasive phenotype, but little is known about how such compression affects the key events and corresponding regulatory mechanisms associated with invasion of breast cancer cells including cellular behaviors and matrix degradation. Results Here we show that compression enhanced invasion and matrix degradation of breast cancer cells. We also identified Piezo1 as the putative mechanosensitive cellular component that transmitted compression to not only enhance the invasive phenotype, but also induce calcium influx and downstream Src signaling. Furthermore, we demonstrated that Piezo1 was mainly localized in caveolae, and both Piezo1 expression and compression-enhanced invasive phenotype of the breast cancer cells were reduced when caveolar integrity was compromised by either knocking down caveolin1 expression or depleting cholesterol content. Conclusions Taken together, our data indicate that mechanical compression activates Piezo1 channels to mediate enhanced breast cancer cell invasion, which involves both cellular events and matrix degradation. This may be a critical mechanotransduction pathway during breast cancer metastasis, and thus potentially a novel therapeutic target for the disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00401-6.
Collapse
Affiliation(s)
- Mingzhi Luo
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, People's Republic of China.,Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Kenneth K Y Ho
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.,Present address: Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Kang Wen
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, People's Republic of China
| | - Zhaowen Tong
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, People's Republic of China.
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA. .,Applied Physics Program, University of Michigan, Ann Arbor, MI, USA. .,Department of Biophysics, University of Michigan, Ann Arbor, MI, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA. .,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
37
|
Pan X. Cholesterol Metabolism in Chronic Kidney Disease: Physiology, Pathologic Mechanisms, and Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:119-143. [PMID: 35503178 PMCID: PMC11106795 DOI: 10.1007/978-981-19-0394-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High plasma levels of lipids and/or lipoproteins are risk factors for atherosclerosis, nonalcoholic fatty liver disease (NAFLD), obesity, and diabetes. These four conditions have also been identified as risk factors leading to the development of chronic kidney disease (CKD). Although many pathways that generate high plasma levels of these factors have been identified, most clinical and physiologic dysfunction results from aberrant assembly and secretion of lipoproteins. The results of several published studies suggest that elevated levels of low-density lipoprotein (LDL)-cholesterol are a risk factor for atherosclerosis, myocardial infarction, coronary artery calcification associated with type 2 diabetes, and NAFLD. Cholesterol metabolism has also been identified as an important pathway contributing to the development of CKD; clinical treatments designed to alter various steps of the cholesterol synthesis and metabolism pathway are currently under study. Cholesterol synthesis and catabolism contribute to a multistep process with pathways that are regulated at the cellular level in renal tissue. Cholesterol metabolism may also be regulated by the balance between the influx and efflux of cholesterol molecules that are capable of crossing the membrane of renal proximal tubular epithelial cells and podocytes. Cellular accumulation of cholesterol can result in lipotoxicity and ultimately kidney dysfunction and failure. Thus, further research focused on cholesterol metabolism pathways will be necessary to improve our understanding of the impact of cholesterol restriction, which is currently a primary intervention recommended for patients with dyslipidemia.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
38
|
Ahn SJ, Fancher IS, Granados ST, Do Couto NF, Hwang CL, Phillips SA, Levitan I. Cholesterol-Induced Suppression of Endothelial Kir Channels Is a Driver of Impairment of Arteriolar Flow-Induced Vasodilation in Humans. Hypertension 2022; 79:126-138. [PMID: 34784737 PMCID: PMC8845492 DOI: 10.1161/hypertensionaha.121.17672] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dyslipidemia-induced endothelial dysfunction is an important factor in the progression of cardiovascular disease; however, the underlying mechanisms are unclear. Our recent studies demonstrated that flow-induced vasodilation (FIV) is regulated by inwardly rectifying K+ channels (Kir2.1) in resistance arteries. Furthermore, we showed that hypercholesterolemia inhibits Kir2.1-dependent vasodilation. In this study, we introduced 2 new mouse models: (1) endothelial-specific deletion of Kir2.1 to demonstrate the role of endothelial Kir2.1 in FIV and (2) cholesterol-insensitive Kir2.1 mutant to determine the Kir2.1 regulation in FIV under hypercholesterolemia. FIV was significantly reduced in endothelial-specific Kir2.1 knock-out mouse mesenteric arteries compared with control groups. In cholesterol-insensitive Kir2.1 mutant mice, Kir2.1 currents were not affected by cyclodextrin and FIV was restored in cells and arteries, respectively, with a hypercholesterolemic background. To extend our observations to humans, 16 healthy subjects were recruited with LDL (low-density lipoprotein)-cholesterol ranging from 51 to 153 mg/dL and FIV was assessed in resistance arteries isolated from gluteal adipose. Resistance arteries from participants with >100 mg/dL LDL (high-LDL) exhibited reduced FIV as compared with those participants with <100 mg/dL LDL (low-LDL). A significant negative correlation was observed between LDL cholesterol and FIV in high-LDL. Expressing dominant-negative Kir2.1 in endothelium blunted FIV in arteries from low-LDL but had no further effect on FIV in arteries from high-LDL. The Kir2.1-dependent vasodilation more negatively correlated to LDL cholesterol in high-LDL. Overexpressing wild-type Kir2.1 in endothelium fully recovered FIV in arteries from participants with high-LDL. Our data suggest that cholesterol-induced suppression of Kir2.1 is a major mechanism underlying endothelial dysfunction in hypercholesterolemia.
Collapse
Affiliation(s)
- Sang Joon Ahn
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep, College of Medicine, University of Illinois at Chicago
| | - Ibra S. Fancher
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep, College of Medicine, University of Illinois at Chicago,Department of Kinesiology and Applied Physiology, University of Delaware
| | - Sara T. Granados
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep, College of Medicine, University of Illinois at Chicago
| | - Natalia F. Do Couto
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep, College of Medicine, University of Illinois at Chicago,Department of Physical Therapy, College of Applied Health Science, University of Illinois at Chicago
| | - Chueh-Lung Hwang
- Department of Physical Therapy, College of Applied Health Science, University of Illinois at Chicago
| | - Shane A. Phillips
- Department of Physical Therapy, College of Applied Health Science, University of Illinois at Chicago
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep, College of Medicine, University of Illinois at Chicago
| |
Collapse
|
39
|
Hakim MA, Behringer EJ. Methyl-Beta-Cyclodextrin Restores K IR Channel Function in Brain Endothelium of Female Alzheimer's Disease Mice. J Alzheimers Dis Rep 2021; 5:693-703. [PMID: 34755043 PMCID: PMC8543374 DOI: 10.3233/adr-210016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background: As the sixth-leading cause of death in the United States, Alzheimer’s disease (AD) entails deteriorating endothelial control of blood flow throughout the brain. In particular, reduced inward-rectifying K+ (KIR) channel function in animal models of aging and AD compromises endothelial function and optimal perfusion of brain parenchyma. Deficient endothelial KIR channels may result from aberrant interaction with plasma membrane cholesterol as a primary regulator of membrane fluidity and ion channels. Objective: We tested the hypothesis that mild methyl-β-cyclodextrin (MβCD) treatment to reduce membrane cholesterol may restore endothelial KIR channel function in brain endothelium of old AD mice. Methods: Membrane potential was continuously measured in isolated endothelial tubes from posterior cerebral arteries of young (1 to 3 months) and old (16 to 19 months) female 3xTg-AD mice before and after mild treatment with the cholesterol-removing agent MβCD (1 mmol/L). Elevated extracellular potassium ([K+]E; 15 mmol/L) and NS309 (1μmol/L) activated KIR and Ca2+-activated K+ (SKCa/IKCa) channels respectively before and after MβCD treatment. Results: SKCa/IKCa channel function for producing hyperpolarization remained stable regardless of age group and MβCD treatment (ΔVm: ∼–33 mV). However, as deficient during AD, KIR channel function was restored (ΔVm: –9±1 mV) versus pre-MβCD conditions (–5±1 mV); a progressive effect that reached –14±1 mV hyperpolarization at 60 min following MβCD washout. Conclusion: In female animals, MβCD treatment of brain endothelium selectively restores KIR versus SKCa/IKCa channel function during AD. Thus, the endothelial cholesterol-KIR channel interface is a novel target for ameliorating perfusion of the AD brain.
Collapse
Affiliation(s)
- Md A Hakim
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | | |
Collapse
|
40
|
Pipatpolkai T, Quetschlich D, Stansfeld PJ. From Bench to Biomolecular Simulation: Phospholipid Modulation of Potassium Channels. J Mol Biol 2021; 433:167105. [PMID: 34139216 PMCID: PMC8361781 DOI: 10.1016/j.jmb.2021.167105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/05/2022]
Abstract
Potassium (K+) ion channels are crucial in numerous cellular processes as they hyperpolarise a cell through K+ conductance, returning a cell to its resting potential. K+ channel mutations result in multiple clinical complications such as arrhythmia, neonatal diabetes and migraines. Since 1995, the regulation of K+ channels by phospholipids has been heavily studied using a range of interdisciplinary methods such as cellular electrophysiology, structural biology and computational modelling. As a result, K+ channels are model proteins for the analysis of protein-lipid interactions. In this review, we will focus on the roles of lipids in the regulation of K+ channels, and how atomic-level structures, along with experimental techniques and molecular simulations, have helped guide our understanding of the importance of phospholipid interactions.
Collapse
Affiliation(s)
- Tanadet Pipatpolkai
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology Anatomy and Genetics, Parks Road, Oxford OX1 3PT, UK; OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PT, UK
| | - Daniel Quetschlich
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK; Department of Chemistry, South Parks Road, Oxford OX1 3QZ, UK
| | - Phillip J Stansfeld
- School of Life Sciences & Department of Chemistry, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
41
|
Cottrill KA, Peterson RJ, Lewallen CF, Koval M, Bridges RJ, McCarty NA. Sphingomyelinase decreases transepithelial anion secretion in airway epithelial cells in part by inhibiting CFTR-mediated apical conductance. Physiol Rep 2021; 9:e14928. [PMID: 34382377 PMCID: PMC8358481 DOI: 10.14814/phy2.14928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is an anion channel whose dysfunction causes cystic fibrosis (CF). The loss of CFTR function in pulmonary epithelial cells causes surface dehydration, mucus build-up, inflammation, and bacterial infections that lead to lung failure. Little has been done to evaluate the effects of lipid perturbation on CFTR activity, despite CFTR residing in the plasma membrane. This work focuses on the acute effects of sphingomyelinase (SMase), a bacterial virulence factor secreted by CF relevant airway bacteria which degrades sphingomyelin into ceramide and phosphocholine, on the electrical circuitry of pulmonary epithelial monolayers. We report that basolateral SMase decreases CFTR-mediated transepithelial anion secretion in both primary bronchial and tracheal epithelial cells from explant tissue, with current CFTR modulators unable to rescue this effect. Focusing on primary cells, we took a holistic ion homeostasis approach to determine a cause for reduced anion secretion following SMase treatment. Using impedance analysis, we determined that basolateral SMase inhibits apical and basolateral conductance in non-CF primary cells without affecting paracellular permeability. In CF primary airway cells, correction with clinically relevant CFTR modulators did not prevent SMase-mediated inhibition of CFTR currents. Furthermore, SMase was found to inhibit only apical conductance in these cells. Future work should determine the mechanism for SMase-mediated inhibition of CFTR currents, and further explore the clinical relevance of SMase and sphingolipid imbalances.
Collapse
Affiliation(s)
- Kirsten A. Cottrill
- Molecular and Systems Pharmacology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
| | - Raven J. Peterson
- Biochemistry, Cell, and Developmental Biology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
| | - Colby F. Lewallen
- Georgia Institute of TechnologyG.W. Woodruff School of Mechanical EngineeringAtlantaGeorgiaUSA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep MedicineDepartment of MedicineEmory UniversityAtlantaGeorgiaUSA
- Department of Cell BiologyEmory UniversityAtlantaGeorgiaUSA
| | - Robert J. Bridges
- Department of Physiology and BiophysicsCenter for Genetic DiseasesChicago Medical SchoolNorth Chicago, IllinoisUSA
| | - Nael A. McCarty
- Molecular and Systems Pharmacology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
- Department of Pediatrics and Children’s Healthcare of AtlantaCenter for Cystic Fibrosis and Airways Disease ResearchEmory University School of MedicineAtlantaGeorgiaUSA
| |
Collapse
|
42
|
Capera J, Pérez-Verdaguer M, Peruzzo R, Navarro-Pérez M, Martínez-Pinna J, Alberola-Die A, Morales A, Leanza L, Szabó I, Felipe A. A novel mitochondrial Kv1.3-caveolin axis controls cell survival and apoptosis. eLife 2021; 10:e69099. [PMID: 34196606 PMCID: PMC8248986 DOI: 10.7554/elife.69099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/22/2021] [Indexed: 12/16/2022] Open
Abstract
The voltage-gated potassium channel Kv1.3 plays an apparent dual physiological role by participating in activation and proliferation of leukocytes as well as promoting apoptosis in several types of tumor cells. Therefore, Kv1.3 is considered a potential pharmacological target for immunodeficiency and cancer. Different cellular locations of Kv1.3, at the plasma membrane or the mitochondria, could be responsible for such duality. While plasma membrane Kv1.3 facilitates proliferation, the mitochondrial channel modulates apoptotic signaling. Several molecular determinants of Kv1.3 drive the channel to the cell surface, but no information is available about its mitochondrial targeting. Caveolins, which are able to modulate cell survival, participate in the plasma membrane targeting of Kv1.3. The channel, via a caveolin-binding domain (CDB), associates with caveolin 1 (Cav1), which localizes Kv1.3 to lipid raft membrane microdomains. The aim of our study was to understand the role of such interactions not only for channel targeting but also for cell survival in mammalian cells. By using a caveolin association-deficient channel (Kv1.3 CDBless), we demonstrate here that while the Kv1.3-Cav1 interaction is responsible for the channel localization in the plasma membrane, a lack of such interaction accumulates Kv1.3 in the mitochondria. Kv1.3 CDBless severely affects mitochondrial physiology and cell survival, indicating that a functional link of Kv1.3 with Cav1 within the mitochondria modulates the pro-apoptotic effects of the channel. Therefore, the balance exerted by these two complementary mechanisms fine-tune the physiological role of Kv1.3 during cell survival or apoptosis. Our data highlight an unexpected role for the mitochondrial caveolin-Kv1.3 axis during cell survival and apoptosis.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| | - Mireia Pérez-Verdaguer
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| | | | - María Navarro-Pérez
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| | - Juan Martínez-Pinna
- Dept de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicanteSpain
| | - Armando Alberola-Die
- Dept de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicanteSpain
| | - Andrés Morales
- Dept de Fisiología, Genética y Microbiología, Universidad de AlicanteAlicanteSpain
| | - Luigi Leanza
- Department of Biology, University of PadovaPadovaItaly
| | - Ildiko Szabó
- Department of Biology, University of PadovaPadovaItaly
| | - Antonio Felipe
- Molecular Physiology Laboratory, Dpt. de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de BarcelonaBarcelonaSpain
| |
Collapse
|
43
|
Alvira-Iraizoz F, Gillard BT, Lin P, Paterson A, Pauža AG, Ali MA, Alabsi AH, Burger PA, Hamadi N, Adem A, Murphy D, Greenwood MP. Multiomic analysis of the Arabian camel (Camelus dromedarius) kidney reveals a role for cholesterol in water conservation. Commun Biol 2021; 4:779. [PMID: 34163009 PMCID: PMC8222267 DOI: 10.1038/s42003-021-02327-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 06/06/2021] [Indexed: 02/05/2023] Open
Abstract
The Arabian camel (Camelus dromedarius) is the most important livestock animal in arid and semi-arid regions and provides basic necessities to millions of people. In the current context of climate change, there is renewed interest in the mechanisms that enable camelids to survive in arid conditions. Recent investigations described genomic signatures revealing evolutionary adaptations to desert environments. We now present a comprehensive catalogue of the transcriptomes and proteomes of the dromedary kidney and describe how gene expression is modulated as a consequence of chronic dehydration and acute rehydration. Our analyses suggested an enrichment of the cholesterol biosynthetic process and an overrepresentation of categories related to ion transport. Thus, we further validated differentially expressed genes with known roles in water conservation which are affected by changes in cholesterol levels. Our datasets suggest that suppression of cholesterol biosynthesis may facilitate water retention in the kidney by indirectly facilitating the AQP2-mediated water reabsorption.
Collapse
Affiliation(s)
- Fernando Alvira-Iraizoz
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK.
| | - Benjamin T Gillard
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Panjiao Lin
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Alex Paterson
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Audrys G Pauža
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Mahmoud A Ali
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, AL Ain, United Arab Emirates
| | - Ammar H Alabsi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Pamela A Burger
- Department of Interdisciplinary Life Sciences, Research Institute of Wildlife Ecology, Vetmeduni Vienna, Vienna, Austria
| | - Naserddine Hamadi
- Department of Life and Environmental Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Abdu Adem
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, AL Ain, United Arab Emirates.
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Michael P Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
44
|
McGruer V, Tanabe P, Vliet SMF, Dasgupta S, Qian L, Volz DC, Schlenk D. Effects of Phenanthrene Exposure on Cholesterol Homeostasis and Cardiotoxicity in Zebrafish Embryos. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:1586-1595. [PMID: 33523501 DOI: 10.1002/etc.5002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/29/2020] [Accepted: 01/27/2021] [Indexed: 06/12/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are pervasive pollutants in aquatic ecosystems, and developing fish embryos are especially sensitive to PAH exposure. Exposure to crude oil or phenanthrene (a reference PAH found in oil) produces an array of gross morphological abnormalities in developing fish embryos, including cardiotoxicity. Recently, studies utilizing transcriptomic analyses in several oil-exposed fish embryos found significant changes in the abundance of transcripts involved in cholesterol biosynthesis. Given the vital role of cholesterol availability in embryonic heart development, we hypothesized that cholesterol dysregulation in early development contributes to phenanthrene-induced cardiotoxicity. We exposed zebrafish embryos to 12 or 15 µM phenanthrene from 6 to 72 h post fertilization (hpf) and demonstrated that, in conjunction with pericardial edema and bradycardia, several genes (fdft1 and hmgcra) in the cholesterol biosynthetic pathway were significantly altered. When embryos were pretreated with a cholesterol solution from 6 to 24 hpf followed by exposure to phenanthrene from 24 to 48 hpf, the effects of phenanthrene on heart rate were partially mitigated. Despite changes in gene expression, whole-mount in situ staining of cholesterol was not significantly affected in embryos exposed to phenanthrene ranging in stage from 24 to 72 hpf. However, the 2-dimensional yolk area was significantly increased with phenanthrene exposure at 72 hpf, suggesting that lipid transport from the yolk to the developing embryo was impaired. Environ Toxicol Chem 2021;40:1586-1595. © 2021 SETAC.
Collapse
Affiliation(s)
- Victoria McGruer
- Environmental Toxicology Graduate Program, University of California, Riverside, California, USA
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Philip Tanabe
- Environmental Toxicology Graduate Program, University of California, Riverside, California, USA
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Sara M F Vliet
- Environmental Toxicology Graduate Program, University of California, Riverside, California, USA
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Subham Dasgupta
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Le Qian
- College of Sciences, China Agricultural University, Beijing, China
| | - David C Volz
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, California, USA
| |
Collapse
|
45
|
Cai YX, Zhang BL, Yu M, Yang YC, Ao X, Zhu D, Wang QS, Lou J, Liang C, Tang LL, Wu MM, Zhang ZR, Ma HP. Cholesterol Stimulates the Transient Receptor Potential Melastatin 4 Channel in mpkCCD c14 Cells. Front Pharmacol 2021; 12:627875. [PMID: 34054517 PMCID: PMC8160378 DOI: 10.3389/fphar.2021.627875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/21/2021] [Indexed: 11/24/2022] Open
Abstract
We have shown that cholesterol regulates the activity of ion channels in mouse cortical collecting duct (CCD) mpkCCDc14 cells and that the transient receptor potential melastatin 4 (TRPM4) channel is expressed in these cells. However, whether TRPM4 channel is regulated by cholesterol remains unclear. Here, we performed inside-out patch-clamp experiments and found that inhibition of cholesterol biosynthesis by lovastatin significantly decreased, whereas enrichment of cholesterol with exogenous cholesterol significantly increased, TRPM4 channel open probability (Po) by regulating its sensitivity to Ca2+ in mpkCCDc14 cells. In addition, inside-out patch-clamp data show that acute depletion of cholesterol in the membrane inner leaflet by methyl-β-cyclodextrin (MβCD) significantly reduced TRPM4 Po, which was reversed by exogenous cholesterol. Moreover, immunofluorescence microscopy, Western blot, cell-surface biotinylation, and patch clamp analysis show that neither inhibition of intracellular cholesterol biosynthesis with lovastatin nor application of exogenous cholesterol had effect on TRPM4 channel protein abundance in the plasma membrane of mpkCCDc14 cells. Sucrose density gradient centrifugation studies demonstrate that TRPM4 was mainly located in cholesterol-rich lipid rafts. Lipid-protein overlay experiments show that TRPM4 directly interacted with several anionic phospholipids, including PI(4,5)P2. Depletion of PI(4,5)P2 with either wortmannin or PGE2 abrogated the stimulatory effects of exogenous cholesterol on TRPM4 activity, whereas exogenous PI(4,5)P2 (diC8-PI(4,5)P2, a water-soluble analog) increased the effects. These results suggest that cholesterol stimulates TRPM4 via a PI(4,5)P2-dependent mechanism.
Collapse
Affiliation(s)
- Yong-Xu Cai
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Bao-Long Zhang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Miao Yu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Yan-Chao Yang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Xue Ao
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Di Zhu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Qiu-Shi Wang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Jie Lou
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Chen Liang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Liang-Liang Tang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Ming-Ming Wu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zhi-Ren Zhang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key laboratory for Metabolic disorder and cancer related cardiovascular diseases, and Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - He-Ping Ma
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
46
|
Ramal-Sanchez M, Bernabò N, Valbonetti L, Cimini C, Taraschi A, Capacchietti G, Machado-Simoes J, Barboni B. Role and Modulation of TRPV1 in Mammalian Spermatozoa: An Updated Review. Int J Mol Sci 2021; 22:4306. [PMID: 33919147 PMCID: PMC8122410 DOI: 10.3390/ijms22094306] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/26/2022] Open
Abstract
Based on the abundance of scientific publications, the polymodal sensor TRPV1 is known as one of the most studied proteins within the TRP channel family. This receptor has been found in numerous cell types from different species as well as in spermatozoa. The present review is focused on analyzing the role played by this important channel in the post-ejaculatory life of spermatozoa, where it has been described to be involved in events such as capacitation, acrosome reaction, calcium trafficking, sperm migration, and fertilization. By performing an exhaustive bibliographic search, this review gathers, for the first time, all the modulators of the TRPV1 function that, to our knowledge, were described to date in different species and cell types. Moreover, all those modulators with a relationship with the reproductive process, either found in the female tract, seminal plasma, or spermatozoa, are presented here. Since the sperm migration through the female reproductive tract is one of the most intriguing and less understood events of the fertilization process, in the present work, chemotaxis, thermotaxis, and rheotaxis guiding mechanisms and their relationship with TRPV1 receptor are deeply analyzed, hypothesizing its (in)direct participation during the sperm migration. Last, TRPV1 is presented as a pharmacological target, with a special focus on humans and some pathologies in mammals strictly related to the male reproductive system.
Collapse
Affiliation(s)
- Marina Ramal-Sanchez
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Nicola Bernabò
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Monterotondo Scalo, 00015 Rome, Italy
| | - Luca Valbonetti
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Monterotondo Scalo, 00015 Rome, Italy
| | - Costanza Cimini
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Angela Taraschi
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
- Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale”, Via Campo Boario 1, 64100 Teramo, Italy
| | - Giulia Capacchietti
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Juliana Machado-Simoes
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Barbara Barboni
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| |
Collapse
|
47
|
Dharmaraj K, Dattler D, Kahlert H, Lendeckel U, Nagel F, Delcea M, Scholz F. The effects of the chemical environment of menaquinones in lipid monolayers on mercury electrodes on the thermodynamics and kinetics of their electrochemistry. EUROPEAN BIOPHYSICS JOURNAL 2021; 50:731-743. [PMID: 33730176 PMCID: PMC8260536 DOI: 10.1007/s00249-021-01512-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/08/2021] [Indexed: 11/30/2022]
Abstract
AbstractThe effects of the chemical environment of menaquinones (all-trans MK-4 and all-trans MK-7) incorporated in lipid monolayers on mercury electrodes have been studied with respect to the thermodynamics and kinetics of their electrochemistry. The chemical environment relates to the composition of lipid films as well as the adjacent aqueous phase. It could be shown that the addition of all-trans MK-4 to TMCL does not change the phase transition temperatures of TMCL. In case of DMPC monolayers, the presence of cholesterol has no effect on the thermodynamics (formal redox potentials) of all-trans MK-7, but the kinetics are affected. Addition of an inert electrolyte (sodium perchlorate; change of ionic strength) to the aqueous phase shifts the redox potentials of all-trans MK-7 only slightly. The formal redox potentials of all-trans MK-4 were determined in TMCL and nCL monolayers and found to be higher in nCL monolayers than in TMCL monolayers. The apparent electron transfer rate constants, transfer coefficients and activation energies of all-trans MK-4 in cardiolipins have been also determined. Most surprisingly, the apparent electron transfer rate constants of all-trans MK-4 exhibit an opposite pH dependence for TMCL and nCL films: the rate constants increase in TMCL films with increasing pH, but in nCL films they increase with decreasing pH. This study is a contribution to understand environmental effects on the redox properties of membrane bond redox systems.
Graphical abstract
Collapse
Affiliation(s)
- Karuppasamy Dharmaraj
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Dirk Dattler
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Heike Kahlert
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, University of Greifswald, Ferdinand-Sauerbruch-Str, 17475, Greifswald, Germany
| | - Felix Nagel
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Mihaela Delcea
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Fritz Scholz
- Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany.
| |
Collapse
|
48
|
Abstract
All living cells interact dynamically with a constantly changing world. Eukaryotes, in particular, evolved radically new ways to sense and react to their environment. These advances enabled new and more complex forms of cellular behaviour in eukaryotes, including directional movement, active feeding, mating, and responses to predation. But what are the key events and innovations during eukaryogenesis that made all of this possible? Here we describe the ancestral repertoire of eukaryotic excitability and discuss five major cellular innovations that enabled its evolutionary origin. The innovations include a vastly expanded repertoire of ion channels, the emergence of cilia and pseudopodia, endomembranes as intracellular capacitors, a flexible plasma membrane and the relocation of chemiosmotic ATP synthesis to mitochondria, which liberated the plasma membrane for more complex electrical signalling involved in sensing and reacting. We conjecture that together with an increase in cell size, these new forms of excitability greatly amplified the degrees of freedom associated with cellular responses, allowing eukaryotes to vastly outperform prokaryotes in terms of both speed and accuracy. This comprehensive new perspective on the evolution of excitability enriches our view of eukaryogenesis and emphasizes behaviour and sensing as major contributors to the success of eukaryotes. This article is part of the theme issue 'Basal cognition: conceptual tools and the view from the single cell'.
Collapse
Affiliation(s)
- Kirsty Y. Wan
- Living Systems Institute, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Gáspár Jékely
- Living Systems Institute, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
49
|
Sahranavard T, Carbone F, Montecucco F, Xu S, Al-Rasadi K, Jamialahmadi T, Sahebkar A. The role of potassium in atherosclerosis. Eur J Clin Invest 2021; 51:e13454. [PMID: 33216974 DOI: 10.1111/eci.13454] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is a chronic progressive inflammatory condition with a leading prevalence worldwide. Endothelial dysfunction leads to low-density lipoprotein trafficking into subendothelial space and the subsequent form of oxidized LDL (ox-LDL) within intimal layer, perpetuating the vicious cycle of endothelial dysfunction. K+ exerts beneficial effects in vascular wall by reducing LDL oxidization, vascular smooth muscle cells (VSMCs) proliferation, and free radical generation. K+ also modulates vascular tone through a regulatory effect on cell membrane potential. MATERIALS AND METHODS The most relevant papers on the association between 'potassium channels' and 'atherosclerosis' were selected among those deposited on PubMed from 1990 to 2020. RESULTS Here, we provide a short narrative review that elaborates on the role of K+ in atherosclerosis. This review also update the current knowledge about potential pharmacological agents targeting K+ channels with a special focus on pleiotropic activities of agents such as statins, sulfonylureas and dihydropyridines. CONCLUSION In this review, the mechanism of different K+ channels on vascular endothelium will be summarized, mainly focusing on their pathophysiological role in atherosclerosis and potential therapeutic application.
Collapse
Affiliation(s)
- Toktam Sahranavard
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Suowen Xu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | | | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| |
Collapse
|
50
|
Guo Y, Zou G, Qi K, Jin J, Yao L, Pan Y, Xiong W. Simvastatin impairs hippocampal synaptic plasticity and cognitive function in mice. Mol Brain 2021; 14:41. [PMID: 33627175 PMCID: PMC7905661 DOI: 10.1186/s13041-021-00758-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Lipophilic statins which are blood brain barrier (BBB) permeable are speculated to affect the cholesterol synthesis and neural functions in the central nervous system. However, whether these statins can affect cholesterol levels and synaptic plasticity in hippocampus and the in vivo consequence remain unclear. Here, we report that long-term subcutaneous treatments of simvastatin significantly impair mouse hippocampal synaptic plasticity, reflected by the attenuated long-term potentiation of field excitatory postsynaptic potentials. The simvastatin administration causes a deficiency in recognition and spatial memory but fails to affect motor ability and anxiety behaviors in the mice. Mass spectrometry imaging indicates a significant decrease in cholesterol intensity in hippocampus of the mice receiving chronic simvastatin treatments. Such effects of simvastatin are transient because drug discontinuation can restore the hippocampal cholesterol level and synaptic plasticity and the memory function. These findings may provide further clues to elucidate the mechanisms of neurological side effects, especially the brain cognitive function impairment, caused by long-term usage of BBB-permeable statins.
Collapse
Affiliation(s)
- Yujun Guo
- Department of Neurosurgery, Institute On Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences At the Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Guichang Zou
- Department of Neurosurgery, Institute On Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences At the Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Keke Qi
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230029, China
| | - Jin Jin
- Department of Neurosurgery, Institute On Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences At the Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Lei Yao
- Department of Neurosurgery, Institute On Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences At the Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Yang Pan
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230029, China
| | - Wei Xiong
- Department of Neurosurgery, Institute On Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences At the Microscale, University of Science and Technology of China, Hefei, 230026, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|