1
|
Wu S, Guo N, Xu H, Li Y, Sun T, Jiang X, Fu D, You T, Diao S, Huang Y, Hu C. Caveolin-1 ameliorates hepatic injury in non-alcoholic fatty liver disease by inhibiting ferroptosis via the NOX4/ROS/GPX4 pathway. Biochem Pharmacol 2024; 230:116594. [PMID: 39490677 DOI: 10.1016/j.bcp.2024.116594] [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: 05/04/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease globally, with a complex and contentious pathogenesis. Caveolin-1 (CAV1) is an important regulator of liver function and can mitigate liver injury by scavenging reactive oxygen species (ROS). Evidence suggests that NOX4 is a source of ROS production, that oxidative stress and ferroptosis are closely related, and that both are involved in the onset and progression of NAFLD. However, whether CAV1 attenuates liver injury in NAFLD caused by high-fat diet via the NOX4/ROS/GPX4 pathway remains unclear. An in vivo fatty liver model was established by feeding mice with a high-fat diet for 16 weeks. In addition, an in vitro fatty liver model was established by incubating AML-12 cells with free fatty acids for 24 h using an in vitro culture method. In our study, it was observed that a high-fat diet induces mitochondrial damage and worsens oxidative stress in NAFLD. This diet also hinders GPX4 expression, leading to an escalation of ferroptosis and lipid accumulation. To counteract these effects, intraperitoneal administration of CSD peptide in mice attenuated the high-fat diet-induced liver mitochondrial damage and ferroptosis. Likewise, overexpression of CAV1 resulted in an increase in GPX4 expression and a reduction in levels of ROS-mediated iron metamorphosis, thus mitigating the progression of the disease. However, the effects of CAV1 on GPX4-mediated ferroptosis and lipid deposition could be reversed by CAV1 small interfering RNA (SiRNA). Finally, NOX4 inhibitor (GLX351322) treatment increased CAV1 siRNA-mediated GPX4 expression and decreased the level of ROS-mediated ferroptosis. These findings suggest a potential mechanism underlying the protective role of CAV1 against high-fat diet-induced hepatotoxicity in NAFLD, shedding new light on the interplay between CAV1, GPX4, and ferroptosis in liver pathology.
Collapse
Affiliation(s)
- Shuai Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Ning Guo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Hanlin Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Tianyin Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Xiangfu Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Dongdong Fu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Tingyu You
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Shaoxi Diao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
2
|
Aboy-Pardal MCM, Guadamillas MC, Guerrero CR, Català-Montoro M, Toledano-Donado M, Terrés-Domínguez S, Pavón DM, Jiménez-Jiménez V, Jimenez-Carretero D, Zamai M, Folgueira C, Cerezo A, Lolo FN, Nogueiras R, Sabio G, Sánchez-Álvarez M, Echarri A, Garcia R, Del Pozo MA. Plasma membrane remodeling determines adipocyte expansion and mechanical adaptability. Nat Commun 2024; 15:10102. [PMID: 39609408 PMCID: PMC11605069 DOI: 10.1038/s41467-024-54224-y] [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: 06/02/2022] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Adipocytes expand massively to accommodate excess energy stores and protect the organism from lipotoxicity. Adipose tissue expandability is at the center of disorders such as obesity and lipodystrophy; however, little is known about the relevance of adipocyte biomechanics on the etiology of these conditions. Here, we show in male mice in vivo that the adipocyte plasma membrane undergoes caveolar domain reorganization upon lipid droplet expansion. As the lipid droplet grows, caveolae disassemble to release their membrane reservoir and increase cell surface area, and transfer specific caveolar components to the LD surface. Adipose tissue null for caveolae is stiffer, shows compromised deformability, and is prone to rupture under mechanical compression. Mechanistically, phosphoacceptor Cav1 Tyr14 is required for caveolae disassembly: adipocytes bearing a Tyr14Phe mutation at this residue are stiffer and smaller, leading to decreased adiposity in vivo; exhibit deficient transfer of Cav1 and EHD2 to the LD surface, and show distinct Cav1 molecular dynamics and tension adaptation. These results indicate that Cav1 phosphoregulation modulates caveolar dynamics as a relevant component of the homeostatic mechanoadaptation of the differentiated adipocyte.
Collapse
Affiliation(s)
- María C M Aboy-Pardal
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta C Guadamillas
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Science and Agroforestal Technology and Genetics, Faculty of Biochemistry and Environmental Sciences, University of Castilla-La Mancha, Toledo, Spain
| | - Carlos R Guerrero
- ForceTool group, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid, Spain
| | - Mauro Català-Montoro
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mónica Toledano-Donado
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sara Terrés-Domínguez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Dácil M Pavón
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Allergy Therapeutics, Avenida Punto Es, 12, 28805 Alcalá de Henares, Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Health Science, Universidad Católica Santa Teresa de Jesús de Ávila, Ávila, Spain
| | - Daniel Jimenez-Carretero
- Cellomics Unit, Cell and Developmental Biology Area. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Moreno Zamai
- Microscopy and Dynamic Imaging Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cintia Folgueira
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas CIMUS, Santiago de Compostela, Spain
- Stress kinases in Diabetes, Cancer and Cardiovascular Disease lab. Cardiovascular risk factors & brain function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ana Cerezo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Molecular Pharmacology, Lilly Research Laboratories, Alcobendas, Spain
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Rubén Nogueiras
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas CIMUS, Santiago de Compostela, Spain
| | - Guadalupe Sabio
- Stress kinases in Diabetes, Cancer and Cardiovascular Disease lab. Cardiovascular risk factors & brain function program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Cell Compartmentalization, Homeostasis and Inflammation lab, Department of Metabolic and Inflammatory Diseases. Instituto de Investigaciones Biomédicas "Sols-Morreale"-CSIC, Madrid, Spain
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Mechanobiology of Organelles lab. Department of Cellular and Molecular Biology. Centro de Investigaciones Biológicas Margarita Salas - CSIC, Madrid, Spain
| | - Ricardo Garcia
- ForceTool group, Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid, Spain
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology lab, Novel mechanisms in atherosclerosis program. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
3
|
Safi R, Menéndez P, Pol A. Lipid droplets provide metabolic flexibility for cancer progression. FEBS Lett 2024; 598:1301-1327. [PMID: 38325881 DOI: 10.1002/1873-3468.14820] [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: 09/04/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
A hallmark of cancer cells is their remarkable ability to efficiently adapt to favorable and hostile environments. Due to a unique metabolic flexibility, tumor cells can grow even in the absence of extracellular nutrients or in stressful scenarios. To achieve this, cancer cells need large amounts of lipids to build membranes, synthesize lipid-derived molecules, and generate metabolic energy in the absence of other nutrients. Tumor cells potentiate strategies to obtain lipids from other cells, metabolic pathways to synthesize new lipids, and mechanisms for efficient storage, mobilization, and utilization of these lipids. Lipid droplets (LDs) are the organelles that collect and supply lipids in eukaryotes and it is increasingly recognized that the accumulation of LDs is a new hallmark of cancer cells. Furthermore, an active role of LD proteins in processes underlying tumorigenesis has been proposed. Here, by focusing on three major classes of LD-resident proteins (perilipins, lipases, and acyl-CoA synthetases), we provide an overview of the contribution of LDs to cancer progression and discuss the role of LD proteins during the proliferation, invasion, metastasis, apoptosis, and stemness of cancer cells.
Collapse
Affiliation(s)
- Rémi Safi
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
| |
Collapse
|
4
|
Pang L, Cui M, Wu S, Kong J. Cav-1 regulates the bile salt export pump on the canalicular membrane of hepatocytes by PKCα-associated signalling under cholesterol stimulation. J Cell Mol Med 2024; 28:e18110. [PMID: 38164042 PMCID: PMC10844719 DOI: 10.1111/jcmm.18110] [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: 06/13/2023] [Revised: 10/25/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND AIMS The secretion of bile salts transported by the bile salt export pump (BSEP) is the primary driving force for the generation of bile flow; thus, it is closely related to the formation of cholesterol stones. Caveolin-1 (Cav-1), an essential player in cell signalling and endocytosis, is known to co-localize with cholesterol-rich membrane domains. This study illustrates the role of Cav-1 and BSEP in cholesterol stone formation. METHODS Adult male C57BL/6 mice were used as an animal model. HepG2 cells were cultured under different cholesterol concentrations and BSEP, Cav-1, p-PKCα and Hax-1 expression levels were determined via Western blotting. Expression levels of BSEP and Cav-1 mRNA were detected using real-time PCR. Immunofluorescence and immunoprecipitation assays were performed to study BSEP and Hax-1 distribution. Finally, an ATPase activity assay was performed to detect BSEP transport activity under different cholesterol concentrations in cells. RESULTS Under low-concentration stimulation with cholesterol, Cav-1 and BSEP protein and mRNA expression levels significantly increased, PKCα phosphorylation significantly decreased, BSEP binding capacity to Hax-1 weakened, and BSEP function increased. Under high-concentration stimulation with cholesterol, Cav-1 and BSEP protein and mRNA expression levels decreased, PKCα phosphorylation increased, BSEP binding capacity to Hax-1 rose, and BSEP function decreased. CONCLUSION Cav-1 regulates the bile salt export pump on the canalicular membrane of hepatocytes via PKCα-associated signalling under cholesterol stimulation.
Collapse
Affiliation(s)
- Liwei Pang
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Meiying Cui
- Department of Anesthesiology, Shengjing HospitalChina Medical UniversityShenyangChina
| | - Shuodong Wu
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jing Kong
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
5
|
Morales-Paytuví F, Fajardo A, Ruiz-Mirapeix C, Rae J, Tebar F, Bosch M, Enrich C, Collins BM, Parton RG, Pol A. Early proteostasis of caveolins synchronizes trafficking, degradation, and oligomerization to prevent toxic aggregation. J Cell Biol 2023; 222:e202204020. [PMID: 37526691 PMCID: PMC10394380 DOI: 10.1083/jcb.202204020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/05/2023] [Accepted: 06/09/2023] [Indexed: 08/02/2023] Open
Abstract
Caveolin-1 (CAV1) and CAV3 are membrane-sculpting proteins driving the formation of the plasma membrane (PM) caveolae. Within the PM mosaic environment, caveola assembly is unique as it requires progressive oligomerization of newly synthesized caveolins while trafficking through the biosynthetic-secretory pathway. Here, we have investigated these early events by combining structural, biochemical, and microscopy studies. We uncover striking trafficking differences between caveolins, with CAV1 rapidly exported to the Golgi and PM while CAV3 is initially retained in the endoplasmic reticulum and laterally moves into lipid droplets. The levels of caveolins in the endoplasmic reticulum are controlled by proteasomal degradation, and only monomeric/low oligomeric caveolins are exported into the cis-Golgi with higher-order oligomers assembling beyond this compartment. When any of those early proteostatic mechanisms are compromised, chemically or genetically, caveolins tend to accumulate along the secretory pathway forming non-functional aggregates, causing organelle damage and triggering cellular stress. Accordingly, we propose a model in which disrupted proteostasis of newly synthesized caveolins contributes to pathogenesis.
Collapse
Affiliation(s)
- Frederic Morales-Paytuví
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alba Fajardo
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carles Ruiz-Mirapeix
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - James Rae
- Institute for Molecular Bioscience (IMB), The University of Queensland (UQ) , Brisbane, Australia
| | - Francesc Tebar
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Enrich
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Brett M Collins
- Institute for Molecular Bioscience (IMB), The University of Queensland (UQ) , Brisbane, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience (IMB), The University of Queensland (UQ) , Brisbane, Australia
- Centre for Microscopy and Microanalysis (CMM), The University of Queensland (UQ), Brisbane, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA) , Barcelona, Spain
| |
Collapse
|
6
|
Safi R, Sánchez-Álvarez M, Bosch M, Demangel C, Parton RG, Pol A. Defensive-lipid droplets: Cellular organelles designed for antimicrobial immunity. Immunol Rev 2023; 317:113-136. [PMID: 36960679 DOI: 10.1111/imr.13199] [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] [Indexed: 03/25/2023]
Abstract
Microbes have developed many strategies to subvert host organisms, which, in turn, evolved several innate immune responses. As major lipid storage organelles of eukaryotes, lipid droplets (LDs) are an attractive source of nutrients for invaders. Intracellular viruses, bacteria, and protozoan parasites induce and physically interact with LDs, and the current view is that they "hijack" LDs to draw on substrates for host colonization. This dogma has been challenged by the recent demonstration that LDs are endowed with a protein-mediated antibiotic activity, which is upregulated in response to danger signals and sepsis. Dependence on host nutrients could be a generic "Achilles' heel" of intracellular pathogens and LDs a suitable chokepoint harnessed by innate immunity to organize a front-line defense. Here, we will provide a brief overview of the state of the conflict and discuss potential mechanisms driving the formation of the 'defensive-LDs' functioning as hubs of innate immunity.
Collapse
Affiliation(s)
- Rémi Safi
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Miguel Sánchez-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (IIB), Madrid, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Caroline Demangel
- Immunobiology and Therapy Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Robert G Parton
- Institute for Molecular Bioscience (IMB), Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis (CMM), University of Queensland, Brisbane, Queensland, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
7
|
Schüssele DS, Haller PK, Haas ML, Hunter C, Sporbeck K, Proikas-Cezanne T. Autophagy profiling in single cells with open source CellProfiler-based image analysis. Autophagy 2023; 19:338-351. [PMID: 35435815 PMCID: PMC9809960 DOI: 10.1080/15548627.2022.2065617] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Single cell-based analysis of macroautophagy/autophagy is largely achieved through the use of fluorescence microscopy to detect autophagy-related proteins that associate with autophagic membranes and therefore can be quantified as fluorescent puncta. In this context, an automated analysis of the number and size of recognized puncta is preferable to a manual count, because more reliable results can be generated in a short time. Here we present a method for open source CellProfiler software-based analysis for quantitative autophagy assessments using GFP-tagged WIPI1 (WD repeat domain, phosphoinositide interacting 1) images acquired with Airyscan or confocal laser-scanning microscopy. The CellProfiler protocol is provided as a ready-to-use software pipeline, and the creation of this pipeline is detailed in both text and video formats. In addition, we provide CellProfiler pipelines for endogenous SQSTM1/p62 (sequestosome 1) or intracellular lipid droplet (LD) analysis, suitable to assess forms of selective autophagy. All protocols and software pipelines can be quickly and easily adapted for the use of alternative autophagy markers or cell types, and can also be used for high-throughput purposes.Abbreviations: AF Alexa Fluor ATG autophagy related BafA1 bafilomycin A1 BSA bovine serum albumin DAPI 4,6-diamidino-2-phenylindole DMEM Dulbecco's modified Eagle's medium DMSO dimethyl sulfoxide EDTA ethylenediaminetetraacetic acid EBSS Earle's balanced salt solution FBS fetal bovine serum GFP green fluorescent protein LD lipid droplet LSM laser scanning microscope MAP1LC3B microtubule associated protein 1 light chain 3 beta MTOR mechanistic target of rapamycin kinase PBS phosphate-buffered saline PIK3C3/VPS34 phosphatidylinositol 3-kinase catalytic subunit type 3 SQSTM1 sequestosome 1 TIFF tagged image file format U2OS U-2 OS cell line WIPI WD repeat domain, phosphoinositide interacting.
Collapse
Affiliation(s)
- David S. Schüssele
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Patricia K. Haller
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany,International Max Planck Research School ‘From Molecules to Organisms’, Max Planck Institute for Developmental Biology and Eberhard Karls University Tübingen, Tübingen, Germany
| | - Maximilian L. Haas
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Catherine Hunter
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany,International Max Planck Research School ‘From Molecules to Organisms’, Max Planck Institute for Developmental Biology and Eberhard Karls University Tübingen, Tübingen, Germany
| | - Katharina Sporbeck
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany,International Max Planck Research School ‘From Molecules to Organisms’, Max Planck Institute for Developmental Biology and Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tassula Proikas-Cezanne
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, Tübingen, Germany,International Max Planck Research School ‘From Molecules to Organisms’, Max Planck Institute for Developmental Biology and Eberhard Karls University Tübingen, Tübingen, Germany,CONTACT Tassula Proikas-Cezanne Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076Tübingen, Germany
| |
Collapse
|
8
|
Moreno-Echeverri AM, Susnik E, Vanhecke D, Taladriz-Blanco P, Balog S, Petri-Fink A, Rothen-Rutishauser B. Pitfalls in methods to study colocalization of nanoparticles in mouse macrophage lysosomes. J Nanobiotechnology 2022; 20:464. [PMID: 36309696 PMCID: PMC9618187 DOI: 10.1186/s12951-022-01670-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background In the field of nanoscience there is an increasing interest to follow dynamics of nanoparticles (NP) in cells with an emphasis on endo-lysosomal pathways and long-term NP fate. During our research on this topic, we encountered several pitfalls, which can bias the experimental outcome. We address some of these pitfalls and suggest possible solutions. The accuracy of fluorescence microscopy methods has an important role in obtaining insights into NP interactions with lysosomes at the single cell level including quantification of NP uptake in a specific cell type. Methods Here we use J774A.1 cells as a model for professional phagocytes. We expose them to fluorescently-labelled amorphous silica NP with different sizes and quantify the colocalization of fluorescently-labelled NP with lysosomes over time. We focus on confocal laser scanning microscopy (CLSM) to obtain 3D spatial information and follow live cell imaging to study NP colocalization with lysosomes. Results We evaluate different experimental parameters that can bias the colocalization coefficients (i.e., Pearson’s and Manders’), such as the interference of phenol red in the cell culture medium with the fluorescence intensity and image post-processing (effect of spatial resolution, optical slice thickness, pixel saturation and bit depth). Additionally, we determine the correlation coefficients for NP entering the lysosomes under four different experimental set-ups. First, we found out that not only Pearson’s, but also Manders’ correlation coefficient should be considered in lysosome-NP colocalization studies; second, there is a difference in NP colocalization when using NP of different sizes and fluorescence dyes and last, the correlation coefficients might change depending on live-cell and fixed-cell imaging set-up. Conclusions The results summarize detailed steps and recommendations for the experimental design, staining, sample preparation and imaging to improve the reproducibility of colocalization studies between the NP and lysosomes. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01670-9.
Collapse
|
9
|
Santoscoy MC, Jarboe LR. Production of cholesterol-like molecules impacts Escherichia coli robustness, production capacity, and vesicle trafficking. Metab Eng 2022; 73:134-143. [PMID: 35842218 DOI: 10.1016/j.ymben.2022.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/26/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
The economic viability of bioprocesses is constrained by the limited range of operating conditions that can be tolerated by the cell factory. Engineering of the microbial cell membrane is one strategy that can increase robustness and thus alter this range. In this work, we targeted cellular components that contribute to maintenance of appropriate membrane function, such as: flotillin-like proteins, membrane structural proteins, and membrane lipids. Specifically, we exploited the promiscuity of squalene hopene cyclase (SHC) to produce polycyclic terpenoids with properties analogous to cholesterol. Strains producing these cholesterol-like molecules were visualized by AFM and height features were observed. Production of these cholesterol-like molecules was associated with increased tolerance towards a diversity of chemicals, particularly alcohols, and membrane trafficking processes such as lipid droplet accumulation and production of extracellular vesicles. This engineering approach improved the production titers for wax-esters and ethanol by 80- and 10-fold, respectively. Expression of SHC resulted in the production of steroids. Strains engineered to also express truncated squalene synthase (tERG9) produced diplopterol and generally did not perform as well. Increased expression of several membrane-associated proteins, such as YqiK, was observed to impact vesicle trafficking and further improve tolerance relative to SHC alone, but did not improve bio-production. Deletion of YbbJ increased lipid droplet accumulation as well as production of intracellular wax esters. This work serves as a proof of concept for engineering strategies targeting membrane physiology and trafficking to expand the production capacity of microbial cell factories.
Collapse
Affiliation(s)
- Miguel C Santoscoy
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Laura R Jarboe
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
10
|
Abstract
Several studies have reported a significant association between the metabolic syndrome (MetS) and mortality around the world. Caveolin-1 (CAV-1) has been widely studied in dyslipidaemia, and several studies have indicated that CAV-1 genetic variations may correlate with dietary intake of fatty acids. This study aimed to investigate the interaction of CAV-1 rs3807992 with types of dietary fatty acid in the MetS risk. This cross-sectional study was carried out on 404 overweight and obese females. Dietary intake was obtained from a 147-item FFQ. The CAV-1 genotype was measured using the PCR-restriction fragment length polymorphism method. Anthropometric values and serum levels (TC, LDL, HDL, TAG and FBS) were measured by standard methods. It was observed that the (AA + AG) group had significantly higher BMI, waist circumference and DBP (P = 0·02, P = 0·02, and P = 0·01, respectively) and lower serum LDL, HDL and TC (P < 0·05) than the GG group. It was found that A allele carriers were at higher odds of the MetS (P = 0·01), abdominal obesity (P = 0·06), increased TAG concentration (P = 0·01), elevated blood pressure (BP) (P = 0·01), increased glucose concentration (P = 0·45) and decreased HDL-cholesterol concentration (P = 0·03). Moreover, the interaction of CAV-1 and SFA intake was significant in terms of the MetS (P = 0·03), LDL (P = 0·03) and BP (P = 0·01). Additionally, the (AA + AG) group was significantly related to PUFA intake in terms of the MetS (P = 0·04), TAG (P = 0·02), glucose (P = 0·02) and homoeostasis model assessment insulin resistance (P = 0·01). Higher PUFA consumption might attenuate the CAV-1 rs3807992 associations with the MetS, and individuals with greater genetic predisposition appeared to have a higher risk of the MetS, associated with higher SFA consumption.
Collapse
|
11
|
Schneider Alves AC, Cardoso RS, de Oliveira Neto XA, Kawano DF. Uncovering the Potential of Lipid Drugs: A Focus on Transient Membrane Microdomain-Targeted Lipid Therapeutics. Mini Rev Med Chem 2022; 22:2318-2331. [PMID: 35264091 DOI: 10.2174/1389557522666220309162203] [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: 10/04/2021] [Revised: 10/27/2021] [Accepted: 01/27/2022] [Indexed: 11/22/2022]
Abstract
Membrane lipids are generally viewed as inert physical barriers, but many vital cellular processes greatly rely on the interaction with these structures, as expressed by the membrane hypothesis that explain the genesis of schizophrenia, Alzheimer's and autoimmune diseases, chronic fatigue or cancer, among others. The concept that the cell membrane displays transient membrane microdomains with distinct lipid composition provide the basis for the development of selective lipid-targeted therapies, the membrane-lipid therapies (MLTs). In this concern, medicinal chemists may design therapeutically valuable compounds 1) with a higher affinity for the lipids in these microdomains to restore the normal physiological conditions, 2) that can directly or 3) indirectly (via enzyme inhibition/activation) replace damaged lipids or restore the regular lipid levels in the whole membrane or microdomain, 4) that alter the expression of genes related to lipid genesis/metabolism or 5) that modulate the pathways related to the membrane binding affinity of lipid-anchored proteins. In this context, this mini-review aims to explore the structural diversity and clinical applications of some of the main membrane and microdomain-targeted lipid drugs.
Collapse
Affiliation(s)
- Anna Carolina Schneider Alves
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP 200 Cândido Portinari Street, Campinas, SP 13083871. Brazil
| | - Raquel Soares Cardoso
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP. 200 Cândido Portinari Street, Campinas, SP 13083871. Brazil
| | - Xisto Antonio de Oliveira Neto
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP. 200 Cândido Portinari Street, Campinas, SP 13083871. Brazil
| | - Daniel Fábio Kawano
- Faculty of Pharmaceutical Sciences, University of Campinas - UNICAMP. 200 Cândido Portinari Street, Campinas, SP 13083871. Brazil
| |
Collapse
|
12
|
Luo S, Yang M, Zhao H, Han Y, Jiang N, Yang J, Chen W, Li C, Liu Y, Zhao C, Sun L. Caveolin-1 Regulates Cellular Metabolism: A Potential Therapeutic Target in Kidney Disease. Front Pharmacol 2021; 12:768100. [PMID: 34955837 PMCID: PMC8703113 DOI: 10.3389/fphar.2021.768100] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/08/2021] [Indexed: 01/09/2023] Open
Abstract
The kidney is an energy-consuming organ, and cellular metabolism plays an indispensable role in kidney-related diseases. Caveolin-1 (Cav-1), a multifunctional membrane protein, is the main component of caveolae on the plasma membrane. Caveolae are represented by tiny invaginations that are abundant on the plasma membrane and that serve as a platform to regulate cellular endocytosis, stress responses, and signal transduction. However, caveolae have received increasing attention as a metabolic platform that mediates the endocytosis of albumin, cholesterol, and glucose, participates in cellular metabolic reprogramming and is involved in the progression of kidney disease. It is worth noting that caveolae mainly depend on Cav-1 to perform the abovementioned cellular functions. Furthermore, the mechanism by which Cav-1 regulates cellular metabolism and participates in the pathophysiology of kidney diseases has not been completely elucidated. In this review, we introduce the structure and function of Cav-1 and its functions in regulating cellular metabolism, autophagy, and oxidative stress, focusing on the relationship between Cav-1 in cellular metabolism and kidney disease; in addition, Cav-1 that serves as a potential therapeutic target for treatment of kidney disease is also described.
Collapse
Affiliation(s)
- Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Hao Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Na Jiang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Jinfei Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Wei Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Chenrui Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yan Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Chanyue Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| |
Collapse
|
13
|
Hello from the other side: Membrane contact of lipid droplets with other organelles and subsequent functional implications. Prog Lipid Res 2021; 85:101141. [PMID: 34793861 DOI: 10.1016/j.plipres.2021.101141] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Lipid droplets (LDs) are ubiquitous organelles that play crucial roles in response to physiological and environmental cues. The identification of several neutral lipid synthesizing and regulatory protein complexes have propelled significant advance on the mechanisms of LD biogenesis in the endoplasmic reticulum (ER). Increasing evidence suggests that distinct proteins and regulatory factors, which localize to membrane contact sites (MCS), are involved not only in interorganellar lipid exchange and transport, but also function in other important cellular processes, including autophagy, mitochondrial dynamics and inheritance, ion signaling and inter-regulation of these MCS. More and more tethers and molecular determinants are associated to MCS and to a diversity of cellular and pathophysiological processes, demonstrating the dynamics and importance of these junctions in health and disease. The conjugation of lipids with proteins in supramolecular complexes is known to be paramount for many biological processes, namely membrane biosynthesis, cell homeostasis, regulation of organelle division and biogenesis, and cell growth. Ultimately, this physical organization allows the contact sites to function as crucial metabolic hubs that control the occurrence of chemical reactions. This leads to biochemical and metabolite compartmentalization for the purposes of energetic efficiency and cellular homeostasis. In this review, we will focus on the structural and functional aspects of LD-organelle interactions and how they ensure signaling exchange and metabolites transfer between organelles.
Collapse
|
14
|
Hallberg I, Persson S, Olovsson M, Sirard MA, Damdimopoulou P, Rüegg J, Sjunnesson YCB. Perfluorooctane sulfonate (PFOS) exposure of bovine oocytes affects early embryonic development at human-relevant levels in an in vitro model. Toxicology 2021; 464:153028. [PMID: 34762985 DOI: 10.1016/j.tox.2021.153028] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/20/2021] [Accepted: 11/04/2021] [Indexed: 02/02/2023]
Abstract
Perfluorooctane sulfonate (PFOS) has been added to Stockholm Convention for global phase out, but will continue to contribute to the chemical burden in humans for a long time to come due to extreme persistence in the environment. In the body, PFOS is transferred into to the ovarian follicular fluid that surrounds the maturing oocyte. In the present study, bovine cumulus oocyte complexes were exposed to PFOS during 22 h in vitro maturation. Concentrations of 2 ng g-1 (PFOS-02) representing average human exposure and 53 ng g-1 (PFOS-53) relevant to highly exposed groups were used. After exposure, developmental competence was recorded until day 8 after fertilisation. Blastocysts were fixed and either stained to evaluate blastomere number and lipid distribution using confocal microscopy or frozen and pooled for microarray-based gene expression and DNA methylation analyses. PFOS-53 delayed the first cleavage to two-cell stage and beyond at 44 h after fertilisation (p < .01). No reduction of proportion blastocysts were seen at day 8 in either of the groups, but PFOS-53 exposure resulted in delayed development into more advanced stages of blastocysts seen as both reduced developmental stage (p = .001) and reduced number of blastomeres (p = .04). Blastocysts showed an altered lipid distribution that was more pronounced after exposure to PFOS-53 (increased total lipid volume, p=.0003, lipid volume/cell p < .0001) than PFOS-02, where only decreased average lipid droplet size (p=.02) was observed. Gene expression analyses revealed pathways differently regulated in the PFOS-treated groups compared to the controls, which were related to cell death and survival through e.g., P38 mitogen-activated protein kinases and signal transducer and activator of transcription 3, which in turn activates tumour protein 53 (TP53). Transcriptomic changes were also associated with metabolic stress response, differentiation and proliferation, which could help to explain the phenotypic changes seen in the blastocysts. The gene expression changes were more pronounced after exposure to PFOS-53 compared to PFOS-02. DNA-methylation changes were associated with similar biological functions as the transcriptomic data, with the most significantly associated pathway being TP53. Collectively, these results reveal that brief PFOS exposure during oocyte maturation alters the early embryo development at concentrations relevant to humans. This study adds to the evidence that PFOS has the potential to affect female fertility.
Collapse
Affiliation(s)
- Ida Hallberg
- Department of Clinical Sciences, Division of Reproduction, The Centre for Reproductive Biology in Uppsala, Swedish University of Agricultural Sciences, SE-750 07, Uppsala, Sweden.
| | - Sara Persson
- Department of Clinical Sciences, Division of Reproduction, The Centre for Reproductive Biology in Uppsala, Swedish University of Agricultural Sciences, SE-750 07, Uppsala, Sweden
| | - Matts Olovsson
- Department of Women's and Children's Health, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Marc-André Sirard
- Department of Animal Sciences, Laval University, QC G1V 0A6, Quebec, Canada
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, SE-141 86, Stockholm, Sweden
| | - Joëlle Rüegg
- Department of Organismal Biology, Program of Environmental Toxicology, Uppsala University, SE-752 36, Uppsala, Sweden
| | - Ylva C B Sjunnesson
- Department of Clinical Sciences, Division of Reproduction, The Centre for Reproductive Biology in Uppsala, Swedish University of Agricultural Sciences, SE-750 07, Uppsala, Sweden
| |
Collapse
|
15
|
Dong Z, Wei Y, Tao M, Zhang L. Activation of the purinergic receptor P2X7 improves hepatosteatosis by promoting lipophagy. FEBS Lett 2021; 595:2768-2780. [PMID: 34652813 DOI: 10.1002/1873-3468.14207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/25/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health problem that develops through unclear molecular mechanisms. The P2X7 purinergic receptor (P2RX7) is an ATP-gated ion channel that belongs to the P2XR family. Thus far, studies on P2RX7 in NAFLD have been largely contradictory. Integrating experiments and modeling, we elucidate the dynamic processes of lipid droplet fusion and degradation following regulation of P2RX7. We show that activation of P2RX7 can activate the AMPK/ULK1 pathway to promote autophagosome generation and lysosomal degradation of autophagosomes. Inhibiting P2RX7 has the opposite effect. Notably, we find that lipid droplets become larger by the fusion of dysfunctional lysosomes but cannot be degraded by them following P2RX7 inhibition. Our study provides evidence that P2RX7 activation improves NAFLD by promoting lipophagy.
Collapse
Affiliation(s)
- Zizhi Dong
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Yujia Wei
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Min Tao
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Lili Zhang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, China
| |
Collapse
|
16
|
Khatibi N, Mirzababaei A, Shiraseb F, Abaj F, Koohdani F, Mirzaei K. Interactions between caveolin 1 polymorphism and the Mediterranean and Mediterranean-DASH Intervention for Neurodegenerative Delay diet (MIND) diet on metabolic dyslipidemia in overweight and obese adult women: a cross-sectional study. BMC Res Notes 2021; 14:364. [PMID: 34544501 PMCID: PMC8454002 DOI: 10.1186/s13104-021-05777-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/07/2021] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE The increased prevalence of metabolic dyslipidemia (MD) and its association with a variety of disorders raised a lot of attention to its management. Caveolin 1 (CAV1) the key protein in the caval structure of plasma membranes is many cell types that play an important role in its function. (CAV1) is a known gene associated with obesity. Today, a novel diet recognized as the Mediterranean and Mediterranean-DASH Intervention for Neurodegenerative Delay diet (MIND) is reported to have a positive effect on overall health. Hence, we aimed to investigate the interactions between CAV1 polymorphism and MIND diet on the MD in overweight and obese patients. RESULTS Remarkably, there was a significant interaction between the MIND diet and CAV1 rs3807992 for dyslipidemia (β = - 0.25 ± 132, P = 0.05) in the crude model. Whereby, subjects with dominant alleles had a lower risk of dyslipidemia and risk allele carriers with higher adherence to the MIND diet may exhibit the lower dyslipidemia. This study presented the CAV1 gene as a possible genetic marker in recognizing people at higher risks for metabolic diseases. It also indicated that using the MIND diet may help in improving dyslipidemia through providing a probable interaction with CAV1 rs3807992 polymorphism.
Collapse
Affiliation(s)
- Nasim Khatibi
- Shahid Sadoughi University of Medical Science, Yazd, Iran
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Atieh Mirzababaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Faezeh Abaj
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Fariba Koohdani
- Department of Cellular, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Molecular Nutrition, Tehran, Iran
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran.
| |
Collapse
|
17
|
Ramírez CM, Torrecilla-Parra M, Pardo-Marqués V, de-Frutos MF, Pérez-García A, Tabraue C, de la Rosa JV, Martín-Rodriguez P, Díaz-Sarmiento M, Nuñez U, Orizaola MC, Través PG, Camps M, Boscá L, Castrillo A. Crosstalk Between LXR and Caveolin-1 Signaling Supports Cholesterol Efflux and Anti-Inflammatory Pathways in Macrophages. Front Endocrinol (Lausanne) 2021; 12:635923. [PMID: 34122329 PMCID: PMC8190384 DOI: 10.3389/fendo.2021.635923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/24/2021] [Indexed: 02/05/2023] Open
Abstract
Macrophages are immune cells that play crucial roles in host defense against pathogens by triggering their exceptional phagocytic and inflammatory functions. Macrophages that reside in healthy tissues also accomplish important tasks to preserve organ homeostasis, including lipid uptake/efflux or apoptotic-cell clearance. Both homeostatic and inflammatory functions of macrophages require the precise stability of lipid-rich microdomains located at the cell membrane for the initiation of downstream signaling cascades. Caveolin-1 (Cav-1) is the main protein responsible for the biogenesis of caveolae and plays an important role in vascular inflammation and atherosclerosis. The Liver X receptors (LXRs) are key transcription factors for cholesterol efflux and inflammatory gene responses in macrophages. Although the role of Cav-1 in cellular cholesterol homeostasis and vascular inflammation has been reported, the connection between LXR transcriptional activity and Cav-1 expression and function in macrophages has not been investigated. Here, using gain and loss of function approaches, we demonstrate that LXR-dependent transcriptional pathways modulate Cav-1 expression and compartmentation within the membrane during macrophage activation. As a result, Cav-1 participates in LXR-dependent cholesterol efflux and the control of inflammatory responses. Together, our data show modulation of the LXR-Cav-1 axis could be exploited to control exacerbated inflammation and cholesterol overload in the macrophage during the pathogenesis of lipid and immune disorders, such as atherosclerosis.
Collapse
Affiliation(s)
- Cristina M. Ramírez
- Instituto Madrileño de Estudios Avanzados (IMDEA) Research Institute of Food and Health Sciences, Madrid, Spain
| | - Marta Torrecilla-Parra
- Instituto Madrileño de Estudios Avanzados (IMDEA) Research Institute of Food and Health Sciences, Madrid, Spain
| | - Virginia Pardo-Marqués
- Instituto Madrileño de Estudios Avanzados (IMDEA) Research Institute of Food and Health Sciences, Madrid, Spain
| | - Mario Fernández de-Frutos
- Instituto Madrileño de Estudios Avanzados (IMDEA) Research Institute of Food and Health Sciences, Madrid, Spain
| | - Ana Pérez-García
- Instituto Madrileño de Estudios Avanzados (IMDEA) Research Institute of Food and Health Sciences, Madrid, Spain
| | - Carlos Tabraue
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Departamento de Morfología, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Juan Vladimir de la Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Patricia Martín-Rodriguez
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Mercedes Díaz-Sarmiento
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Uxue Nuñez
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Marta C. Orizaola
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas CSIC-Universidad Autónoma de Madrid, Madrid, Spain
| | - Paqui G. Través
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas CSIC-Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Camps
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Lisardo Boscá
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas CSIC-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación en Red sobre Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Antonio Castrillo
- Unidad de Biomedicina (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas CSIC-Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
18
|
Matthaeus C, Taraska JW. Energy and Dynamics of Caveolae Trafficking. Front Cell Dev Biol 2021; 8:614472. [PMID: 33692993 PMCID: PMC7939723 DOI: 10.3389/fcell.2020.614472] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Caveolae are 70–100 nm diameter plasma membrane invaginations found in abundance in adipocytes, endothelial cells, myocytes, and fibroblasts. Their bulb-shaped membrane domain is characterized and formed by specific lipid binding proteins including Caveolins, Cavins, Pacsin2, and EHD2. Likewise, an enrichment of cholesterol and other lipids makes caveolae a distinct membrane environment that supports proteins involved in cell-type specific signaling pathways. Their ability to detach from the plasma membrane and move through the cytosol has been shown to be important for lipid trafficking and metabolism. Here, we review recent concepts in caveolae trafficking and dynamics. Second, we discuss how ATP and GTP-regulated proteins including dynamin and EHD2 control caveolae behavior. Throughout, we summarize the potential physiological and cell biological roles of caveolae internalization and trafficking and highlight open questions in the field and future directions for study.
Collapse
Affiliation(s)
- Claudia Matthaeus
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Justin W Taraska
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
19
|
Wang J, Chen MY, Chen JF, Ren QL, Zhang JQ, Cao H, Xing BS, Pan CY. LncRNA IMFlnc1 promotes porcine intramuscular adipocyte adipogenesis by sponging miR-199a-5p to up-regulate CAV-1. BMC Mol Cell Biol 2020; 21:77. [PMID: 33148167 PMCID: PMC7640402 DOI: 10.1186/s12860-020-00324-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023] Open
Abstract
Background Local Chinese local pig breeds have thinner muscle fiber and higher intramuscular-fat (IMF) content. But its regulation mechanism has not been discussed in-depth. Studies indicated that long non coding RNAs (lncRNAs) play important role in muscle and fat development. Results The lncRNAs expressional differences in the longissimus dorsi (LD) muscle were identified between Huainan pigs (local Chinese pigs, fat-type, HN) and Large White pigs (lean-type, LW) at 38, 58, and 78 days post conception (dpc). In total, 2131 novel lncRNAs were identified in 18 samples, and 291, 305, and 683 differentially expressed lncRNAs (DELs) were found between these two breeds at three stages, respectively. The mRNAs that co-expressed with these DELs were used for GO and KEGG analysis, and the results showed that muscle development and energy metabolism were more active at 58 dpc in HN, but at 78 dpc in LW pigs. Muscle cell differentiation and myofibril assembly might associated with earlier myogenesis and primary-muscle-fiber assembly in HN, and cell proliferation, insulin, and the MAPK pathway might be contribute to longer proliferation and elevated energy metabolism in LW pigs at 78 dpc. The PI3K/Akt and cAMP pathways were associated with higher IMF deposition in HN. Intramuscular fat deposition-associated long noncoding RNA 1 (IMFlnc1) was selected for functional verification, and results indicated that it regulated the expressional level of caveolin-1 (CAV-1) by acting as competing endogenous RNA (ceRNA) to sponge miR-199a-5p. Conclusions Our data contributed to understanding the role of lncRNAs in porcine-muscle development and IMF deposition, and provided valuable information for improving pig-meat quality. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-020-00324-8.
Collapse
Affiliation(s)
- Jing Wang
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Number 116, Hua Yuan Road, Jinshui District, Zhengzhou, 450002, China
| | - Ming-Yue Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Key Laboratory of Animal Biotechnology, College of Animal Science and Technology, Northwest A&F University, Ministry of Agriculture, Number 22, Xi Nong Road, Yangling, 712100, Shaanxi, China
| | - Jun-Feng Chen
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Number 116, Hua Yuan Road, Jinshui District, Zhengzhou, 450002, China
| | - Qiao-Ling Ren
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Number 116, Hua Yuan Road, Jinshui District, Zhengzhou, 450002, China
| | - Jia-Qing Zhang
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Number 116, Hua Yuan Road, Jinshui District, Zhengzhou, 450002, China
| | - Hai Cao
- Henan Xing Rui Agriculture and Animal Husbandry Technology Co., LTD, Number 59, Jie Fang Road, Xinxian, Xinyang, 465550, China
| | - Bao-Song Xing
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Number 116, Hua Yuan Road, Jinshui District, Zhengzhou, 450002, China.
| | - Chuan-Ying Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Key Laboratory of Animal Biotechnology, College of Animal Science and Technology, Northwest A&F University, Ministry of Agriculture, Number 22, Xi Nong Road, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
20
|
Buwa N, Mazumdar D, Balasubramanian N. Caveolin1 Tyrosine-14 Phosphorylation: Role in Cellular Responsiveness to Mechanical Cues. J Membr Biol 2020; 253:509-534. [PMID: 33089394 DOI: 10.1007/s00232-020-00143-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
The plasma membrane is a dynamic lipid bilayer that engages with the extracellular microenvironment and intracellular cytoskeleton. Caveolae are distinct plasma membrane invaginations lined by integral membrane proteins Caveolin1, 2, and 3. Caveolae formation and stability is further supported by additional proteins including Cavin1, EHD2, Pacsin2 and ROR1. The lipid composition of caveolar membranes, rich in cholesterol and phosphatidylserine, actively contributes to caveolae formation and function. Post-translational modifications of Cav1, including its phosphorylation of the tyrosine-14 residue (pY14Cav1) are vital to its function in and out of caveolae. Cells that experience significant mechanical stress are seen to have abundant caveolae. They play a vital role in regulating cellular signaling and endocytosis, which could further affect the abundance and distribution of caveolae at the PM, contributing to sensing and/or buffering mechanical stress. Changes in membrane tension in cells responding to multiple mechanical stimuli affects the organization and function of caveolae. These mechanical cues regulate pY14Cav1 levels and function in caveolae and focal adhesions. This review, along with looking at the mechanosensitive nature of caveolae, focuses on the role of pY14Cav1 in regulating cellular mechanotransduction.
Collapse
Affiliation(s)
- Natasha Buwa
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Debasmita Mazumdar
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Nagaraj Balasubramanian
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India.
| |
Collapse
|
21
|
Lipid Stores and Lipid Metabolism Associated Gene Expression in Porcine and Bovine Parthenogenetic Embryos Revealed by Fluorescent Staining and RNA-seq. Int J Mol Sci 2020; 21:ijms21186488. [PMID: 32899450 PMCID: PMC7555686 DOI: 10.3390/ijms21186488] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022] Open
Abstract
Compared to other mammalian species, porcine oocytes and embryos are characterized by large amounts of lipids stored mainly in the form of droplets in the cytoplasm. The amount and the morphology of lipid droplets (LD) change throughout the preimplantation development, however, relatively little is known about expression of genes involved in lipid metabolism of early embryos. We compared porcine and bovine blastocyst stage embryos as well as dissected inner cell mass (ICM) and trophoblast (TE) cell populations with regard to lipid droplet storage and expression of genes functionally annotated to selected lipid gene ontology terms using RNA-seq. Comparing the number and the volume occupied by LD between bovine and porcine blastocysts, we have found significant differences both at the level of single embryo and a single blastomere. Aside from different lipid content, we found that embryos regulate the lipid metabolism differentially at the gene expression level. Out of 125 genes, we found 73 to be differentially expressed between entire porcine and bovine blastocyst, and 36 and 51 to be divergent between ICM and TE cell lines. We noticed significant involvement of cholesterol and ganglioside metabolism in preimplantation embryos, as well as a possible shift towards glucose, rather than pyruvate dependence in bovine embryos. A number of genes like DGAT1, CD36 or NR1H3 may serve as lipid associated markers indicating distinct regulatory mechanisms, while upregulated PLIN2, APOA1, SOAT1 indicate significant function during blastocyst formation and cell differentiation in both models.
Collapse
|
22
|
Affiliation(s)
- Yutong Zhao
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| | - Jing Zhao
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
23
|
Affiliation(s)
- Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom.
| | - Elina Ikonen
- Stem Cells and Metabolism Research Program and Dept. of Anatomy, Faculty of Medicine, University of Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
24
|
Coleman RA. The "discovery" of lipid droplets: A brief history of organelles hidden in plain sight. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158762. [PMID: 32622088 DOI: 10.1016/j.bbalip.2020.158762] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
Abstract
Mammalian lipid droplets (LDs), first described as early as the 1880s, were virtually ignored for more than 100 years. Between 1991 and the early 2000s, however, a series of discoveries and conceptual breakthroughs led to a resurgent interest in obesity as a disease, in the metabolism of intracellular triacylglycerol (TAG), and in the physical locations of LDs as cellular structures with their associated proteins. Insights included the recognition that obesity underlies major chronic diseases, that appetite is hormonally controlled, that hepatic steatosis is not a benign finding, and that diabetes might fundamentally be a disorder of lipid metabolism. In this brief review, I describe the metamorphosis of LDs from overlooked globs of stored fat to dynamic organelles that control insulin resistance, mitochondrial oxidation, and viral replication.
Collapse
Affiliation(s)
- Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America.
| |
Collapse
|
25
|
Wu Y, Fu A, Yossifon G. Active Particle Based Selective Transport and Release of Cell Organelles and Mechanical Probing of a Single Nucleus. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906682. [PMID: 32363783 DOI: 10.1002/smll.201906682] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Self-propelling micromotors are emerging as a promising microscale tool for single-cell analysis. The authors have recently shown that the field gradients necessary to manipulate matter via dielectrophoresis can be induced at the surface of a polarizable active ("self-propelling") metallo-dielectric Janus particle (JP) under an externally applied electric field, acting essentially as a mobile floating microelectrode. Here, the application of the mobile floating microelectrode to trap and transport cell organelles in a selective and releasable manner is successfully extended. This selectivity is driven by the different dielectrophoretic (DEP) potential wells on the JP surface that is controlled by the frequency of the electric field, along with the hydrodynamic shearing and size of the trapped organelles. Such selective and directed loading enables purification of targeted organelles of interest from a mixed biological sample while their dynamic release enables their harvesting for further analysis such as gene/RNA sequencing or proteomics. Moreover, the electro-deformation of the trapped nucleus is shown to be in correlation with the DEP force and hence, can act as a promising label-free biomechanical marker. Hence, the active carrier constitutes an important and novel ex vivo platform for manipulation and mechanical probing of subcellular components of potential for single cell analysis.
Collapse
Affiliation(s)
- Yue Wu
- Faculty of Mechanical Engineering, Micro- and Nano-Fluidics Laboratory, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Afu Fu
- Technion Integrated Cancer Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, 3525433, Israel
| | - Gilad Yossifon
- Faculty of Mechanical Engineering, Micro- and Nano-Fluidics Laboratory, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| |
Collapse
|
26
|
Pol A, Morales-Paytuví F, Bosch M, Parton RG. Non-caveolar caveolins – duties outside the caves. J Cell Sci 2020; 133:133/9/jcs241562. [DOI: 10.1242/jcs.241562] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT
Caveolae are invaginations of the plasma membrane that are remarkably abundant in adipocytes, endothelial cells and muscle. Caveolae provide cells with resources for mechanoprotection, can undergo fission from the plasma membrane and can regulate a variety of signaling pathways. Caveolins are fundamental components of caveolae, but many cells, such as hepatocytes and many neurons, express caveolins without forming distinguishable caveolae. Thus, the function of caveolins goes beyond their roles as caveolar components. The membrane-organizing and -sculpting capacities of caveolins, in combination with their complex intracellular trafficking, might contribute to these additional roles. Furthermore, non-caveolar caveolins can potentially interact with proteins normally excluded from caveolae. Here, we revisit the non-canonical roles of caveolins in a variety of cellular contexts including liver, brain, lymphocytes, cilia and cancer cells, as well as consider insights from invertebrate systems. Non-caveolar caveolins can determine the intracellular fluxes of active lipids, including cholesterol and sphingolipids. Accordingly, caveolins directly or remotely control a plethora of lipid-dependent processes such as the endocytosis of specific cargoes, sorting and transport in endocytic compartments, or different signaling pathways. Indeed, loss-of-function of non-caveolar caveolins might contribute to the common phenotypes and pathologies of caveolin-deficient cells and animals.
Collapse
Affiliation(s)
- Albert Pol
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Frederic Morales-Paytuví
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Marta Bosch
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
| | - Robert G. Parton
- Institute for Molecular Bioscience (IMB), The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
- Centre for Microscopy and Microanalysis (CMM) IMB, The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
| |
Collapse
|
27
|
Bhattachan P, Rae J, Yu H, Jung W, Wei J, Parton RG, Dong B. Ascidian caveolin induces membrane curvature and protects tissue integrity and morphology during embryogenesis. FASEB J 2019; 34:1345-1361. [PMID: 31914618 DOI: 10.1096/fj.201901281r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/30/2019] [Accepted: 11/14/2019] [Indexed: 01/20/2023]
Abstract
Cell morphology and tissue integrity are essential for embryogenesis. Caveolins are membrane proteins that induce the formation of surface pits called caveolae that serve as membrane reservoirs for cell and tissue protection during development. In vertebrates, caveolin 1 (Cav1) and caveolin 3 (Cav3) are required for caveola formation. However, the formation of caveola and the function of caveolins in invertebrates are largely unknown. In this study, three caveolins, Cav-a, Cav-b, and CavY, are identified in the genome of the invertebrate chordate Ciona spp. Based on phylogenetic analysis, Cav-a is found to be closely related to the vertebrate Cav1 and Cav3. In situ hybridization shows that Cav-a is expressed in Ciona embryonic notochord and muscle. Cell-free experiments, model cell culture systems, and in vivo experiments demonstrate that Ciona Cav-a has the ability to induce membrane curvature at the plasma membrane. Knockdown of Cav-a in Ciona embryos causes loss of invaginations in the plasma membrane and results in the failure of notochord elongation and lumenogenesis. Expression of a dominant-negative Cav-a point mutation causes cells to change shape and become displaced from the muscle and notochord to disrupt tissue integrity. Furthermore, we demonstrate that Cav-a vesicles show polarized trafficking and localize at the luminal membrane during notochord lumenogenesis. Taken together, these results show that the invertebrate chordate caveolin from Ciona plays crucial roles in tissue integrity and morphology by inducing membrane curvature and intracellular vesicle trafficking during embryogenesis.
Collapse
Affiliation(s)
- Punit Bhattachan
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - James Rae
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Haiyan Yu
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - WooRam Jung
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Jiankai Wei
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, St. Lucia, QLD, Australia
| | - Bo Dong
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| |
Collapse
|
28
|
Yokomori H, Ando W, Oda M. Cavin-1 is linked to lipid droplet formation in human hepatic stellate cells. Med Mol Morphol 2019; 53:56-59. [PMID: 30877476 DOI: 10.1007/s00795-019-00219-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Hiroaki Yokomori
- Department of Internal Medicine, Kitasato University Medical Center, Saitama, 364-8501, Japan.
| | - Wataru Ando
- Department of Clinical Pharmacy, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Masaya Oda
- Organized Center of Clinical Medicine, International University of Health and Welfare, Tokyo, Japan
| |
Collapse
|
29
|
Clarke D, Pearse Y, Kan SH, Le SQ, Sanghez V, Cooper JD, Dickson PI, Iacovino M. Genetically Corrected iPSC-Derived Neural Stem Cell Grafts Deliver Enzyme Replacement to Affect CNS Disease in Sanfilippo B Mice. Mol Ther Methods Clin Dev 2018; 10:113-127. [PMID: 30101150 PMCID: PMC6076361 DOI: 10.1016/j.omtm.2018.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/07/2018] [Indexed: 12/22/2022]
Abstract
Sanfilippo syndrome type B (mucopolysaccharidosis type IIIB [MPS IIIB]) is a lysosomal storage disorder primarily affecting the brain that is caused by a deficiency in the enzyme α-N-acetylglucosaminidase (NAGLU), leading to intralysosomal accumulation of heparan sulfate. There are currently no treatments for this disorder. Here we report that, ex vivo, lentiviral correction of Naglu-/- neural stem cells derived from Naglu-/- mice (iNSCs) corrected their lysosomal pathology and allowed them to secrete a functional NAGLU enzyme that could be taken up by deficient cells. Following long-term transplantation of these corrected iNSCs into Naglu-/- mice, we detected NAGLU activity in the majority of engrafted animals. Successfully transplanted Naglu-/- mice showed a significant decrease in storage material, a reduction in astrocyte activation, and complete prevention of microglial activation within the area of engrafted cells and neighboring regions, with beneficial effects extending partway along the rostrocaudal axis of the brain. Our results demonstrate long-term engraftment of iNSCs in the brain that are capable of cross-correcting pathology in Naglu-/- mice. Our findings suggest that genetically engineered iNSCs could potentially be used to deliver enzymes and treat MPS IIIB.
Collapse
Affiliation(s)
- Don Clarke
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Phoenix Nest Inc., P.O. Box 150057, Brooklyn, NY 11215, USA
| | - Yewande Pearse
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Shih-hsin Kan
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Steven Q. Le
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Valentina Sanghez
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jonathan D. Cooper
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Patricia I. Dickson
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Michelina Iacovino
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| |
Collapse
|
30
|
Intracellular and Plasma Membrane Events in Cholesterol Transport and Homeostasis. J Lipids 2018; 2018:3965054. [PMID: 30174957 PMCID: PMC6106919 DOI: 10.1155/2018/3965054] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Cholesterol transport between intracellular compartments proceeds by both energy- and non-energy-dependent processes. Energy-dependent vesicular traffic partly contributes to cholesterol flux between endoplasmic reticulum, plasma membrane, and endocytic vesicles. Membrane contact sites and lipid transfer proteins are involved in nonvesicular lipid traffic. Only “active" cholesterol molecules outside of cholesterol-rich regions and partially exposed in water phase are able to fast transfer. The dissociation of partially exposed cholesterol molecules in water determines the rate of passive aqueous diffusion of cholesterol out of plasma membrane. ATP hydrolysis with concomitant conformational transition is required to cholesterol efflux by ABCA1 and ABCG1 transporters. Besides, scavenger receptor SR-B1 is involved also in cholesterol efflux by facilitated diffusion via hydrophobic tunnel within the molecule. Direct interaction of ABCA1 with apolipoprotein A-I (apoA-I) or apoA-I binding to high capacity binding sites in plasma membrane is important in cholesterol escape to free apoA-I. ABCG1-mediated efflux to fully lipidated apoA-I within high density lipoprotein particle proceeds more likely through the increase of “active” cholesterol level. Putative cholesterol-binding linear motifs within the structure of all three proteins ABCA1, ABCG1, and SR-B1 are suggested to contribute to the binding and transfer of cholesterol molecules from cytoplasmic to outer leaflets of lipid bilayer. Together, plasma membrane events and intracellular cholesterol metabolism and traffic determine the capacity of the cell for cholesterol efflux.
Collapse
|
31
|
Fonseca MDC, França A, Florentino RM, Fonseca RC, Lima Filho ACM, Vidigal PTV, Oliveira AG, Dubuquoy L, Nathanson MH, Leite MF. Cholesterol-enriched membrane microdomains are needed for insulin signaling and proliferation in hepatic cells. Am J Physiol Gastrointest Liver Physiol 2018; 315:G80-G94. [PMID: 29471671 PMCID: PMC6109708 DOI: 10.1152/ajpgi.00008.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatocyte proliferation during liver regeneration is a well-coordinated process regulated by the activation of several growth factor receptors, including the insulin receptor (IR). The IR can be localized in part to cholesterol-enriched membrane microdomains, but the role of such domains in insulin-mediated events in hepatocytes is not known. We investigated whether partitioning of IRs into cholesterol-enriched membrane rafts is important for the mitogenic effects of insulin in the hepatic cells. IR and lipid rafts were labeled in HepG2 cells and primary rat hepatocytes. Membrane cholesterol was depleted in vitro with metyl-β-cyclodextrin (MβCD) and in vivo with lovastatin. Insulin-induced calcium (Ca2+) signals studies were examined in HepG2 cells and in freshly isolated rat hepatocytes as well as in whole liver in vivo by intravital confocal imaging. Liver regeneration was studied by 70% partial hepatectomy (PH), and hepatocyte proliferation was assessed by PCNA staining. A subpopulation of IR was found in membrane microdomains enriched in cholesterol. Depletion of cholesterol from plasma membrane resulted in redistribution of the IR along the cells, which was associated with impaired insulin-induced nuclear Ca2+ signals, a signaling event that regulates hepatocyte proliferation. Cholesterol depletion also led to ERK1/2 hyper-phosphorylation. Lovastatin administration to rats decreased hepatic cholesterol content, disrupted lipid rafts and decreased insulin-induced Ca2+ signaling in hepatocytes, and delayed liver regeneration after PH. Therefore, membrane cholesterol content and lipid rafts integrity showed to be important for the proliferative effects of insulin in hepatic cells. NEW & NOTEWORTHY One of insulin's actions is to stimulate liver regeneration. Here we show that a subpopulation of insulin receptors is in a specialized cholesterol-enriched region of the cell membrane and this subfraction is important for insulin's proliferative effects.
Collapse
Affiliation(s)
- Matheus de Castro Fonseca
- 1Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Sao Paulo, Brazil,2Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andressa França
- 2Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil,3Department of Molecular Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo Machado Florentino
- 2Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Roberta Cristelli Fonseca
- 2Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil,4Center for Gastrointestinal Biology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Paula Teixeira Vieira Vidigal
- 5Department of Pathological Anatomy and Forensic Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - André Gustavo Oliveira
- 2Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil,4Center for Gastrointestinal Biology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laurent Dubuquoy
- 6Lille Inflammation Research International Center–UMR995, INSERM, University of Lille, Lille, France
| | - Michael H. Nathanson
- 7Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - M. Fátima Leite
- 2Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
32
|
Lian X, Huang Y, Zhu Y, Fang Y, Zhao R, Joseph E, Li J, Pellois JP, Zhou HC. Enzyme-MOF Nanoreactor Activates Nontoxic Paracetamol for Cancer Therapy. Angew Chem Int Ed Engl 2018; 57:5725-5730. [PMID: 29536600 PMCID: PMC6621563 DOI: 10.1002/anie.201801378] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/09/2018] [Indexed: 11/08/2022]
Abstract
Prodrug activation, by exogenously administered enzymes, for cancer therapy is an approach to achieve better selectivity and less systemic toxicity than conventional chemotherapy. However, the short half-lives of the activating enzymes in the bloodstream has limited its success. Demonstrated here is that a tyrosinase-MOF nanoreactor activates the prodrug paracetamol in cancer cells in a long-lasting manner. By generating reactive oxygen species (ROS) and depleting glutathione (GSH), the product of the enzymatic conversion of paracetamol is toxic to drug-resistant cancer cells. Tyrosinase-MOF nanoreactors cause significant cell death in the presence of paracetamol for up to three days after being internalized by cells, while free enzymes totally lose activity in a few hours. Thus, enzyme-MOF nanocomposites are envisioned to be novel persistent platforms for various biomedical applications.
Collapse
Affiliation(s)
- Xizhen Lian
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255 (USA)
| | - Yanyan Huang
- Beijing National Laboratory for MolecularSciences; CAS Key, Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy Of Sciences, Beijing, 100190(China)
| | - Yuanyuan Zhu
- Beijing National Laboratory for MolecularSciences; CAS Key, Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy Of Sciences, Beijing, 100190(China)
| | - Yu Fang
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255 (USA)
| | - Rui Zhao
- Beijing National Laboratory for MolecularSciences; CAS Key, Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy Of Sciences, Beijing, 100190(China)
| | - Elizabeth Joseph
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255 (USA)
| | - Jialuo Li
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255 (USA)
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University College Station, TX 77843-2128 (USA); Department of Chemistry, Texas A&M University, College Station, TX 77843-3255 (USA)
| | - Hong-Cai Zhou
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255 (USA)
| |
Collapse
|
33
|
Lian X, Huang Y, Zhu Y, Fang Y, Zhao R, Joseph E, Li J, Pellois JP, Zhou HC. Enzyme-MOF Nanoreactor Activates Nontoxic Paracetamol for Cancer Therapy. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201801378] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Xizhen Lian
- Department of Chemistry; Texas A&M University; College Station TX 77843-3255 USA
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 China
| | - Yuanyuan Zhu
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 China
| | - Yu Fang
- Department of Chemistry; Texas A&M University; College Station TX 77843-3255 USA
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences; CAS Key Laboratory of Analytical Chemistry for Living Biosystems; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 China
| | - Elizabeth Joseph
- Department of Chemistry; Texas A&M University; College Station TX 77843-3255 USA
| | - Jialuo Li
- Department of Chemistry; Texas A&M University; College Station TX 77843-3255 USA
| | - Jean-Philippe Pellois
- Department of Chemistry; Texas A&M University; College Station TX 77843-3255 USA
- Department of Biochemistry and Biophysics; Texas A&M University; College Station TX 77843-2128 USA
| | - Hong-Cai Zhou
- Department of Chemistry; Texas A&M University; College Station TX 77843-3255 USA
| |
Collapse
|
34
|
McMillin M, Grant S, Frampton G, Petrescu AD, Kain J, Williams E, Haines R, Canady L, DeMorrow S. FXR-Mediated Cortical Cholesterol Accumulation Contributes to the Pathogenesis of Type A Hepatic Encephalopathy. Cell Mol Gastroenterol Hepatol 2018; 6:47-63. [PMID: 29928671 PMCID: PMC6008252 DOI: 10.1016/j.jcmgh.2018.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 02/26/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Hepatic encephalopathy is a serious neurologic complication of acute and chronic liver diseases. We previously showed that aberrant bile acid signaling contributes to the development of hepatic encephalopathy via farnesoid X receptor (FXR)-mediated mechanisms in neurons. In the brain, a novel alternative bile acid synthesis pathway, catalyzed by cytochrome p450 46A1 (Cyp46A1), is the primary mechanism by which the brain regulates cholesterol homeostasis. The aim of this study was to determine if FXR activation in the brain altered cholesterol homeostasis during hepatic encephalopathy. METHODS Cyp7A1-/- mice or C57Bl/6 mice pretreated with central infusion of FXR vivo morpholino, 2-hydroxypropyl-β-cyclodextrin, or fed a cholestyramine-supplemented diet were injected with azoxymethane (AOM). Cognitive and neuromuscular impairment as well as liver damage and expression of Cyp46A1 were assessed using standard techniques. The subsequent cholesterol content in the frontal cortex was measured using commercially available kits and by Filipin III and Nile Red staining. RESULTS There was an increase in membrane-bound and intracellular cholesterol in the cortex of mice treated with AOM that was associated with decreased Cyp46A1 expression. Strategies to inhibit FXR signaling prevented the down-regulation of Cyp46A1 and the accumulation of cholesterol. Treatment of mice with 2-hydroxypropyl-β-cyclodextrin attenuated the AOM-induced cholesterol accumulation in the brain and the cognitive and neuromuscular deficits without altering the underlying liver pathology. CONCLUSIONS During hepatic encephalopathy, FXR signaling increases brain cholesterol and contributes to neurologic decline. Targeting cholesterol accumulation in the brain may be a possible therapeutic target for the management of hepatic encephalopathy.
Collapse
Key Words
- 2-HβC, 2-hypdroxypropyl-β-cyclodextrin
- AOM, azoxymethane
- Acute Liver Failure
- Azoxymethane
- CYP46A1, cytochrome p450 46A1
- CYP7A1, cytochrome p450 7A1
- Cytochrome p450 46A1
- FXR, farnesoid X receptor
- Farnesoid X Receptor
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- PBS, phosphate-buffered saline
- PFA, paraformaldehyde
- RT-PCR, reverse-transcription polymerase chain reaction
- WT, wild-type
Collapse
Affiliation(s)
- Matthew McMillin
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Stephanie Grant
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Gabriel Frampton
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Anca D. Petrescu
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Jessica Kain
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Elaina Williams
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Rebecca Haines
- Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Lauren Canady
- Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas
| | - Sharon DeMorrow
- Central Texas Veterans Healthcare System, Temple, Texas,Department of Medical Physiology, Texas A&M College of Medicine, Temple, Texas,Correspondence Address correspondence to: Sharon DeMorrow, PhD, Department of Medical Physiology, Texas A&M Health Science Center, Central Texas Veterans Health Care System, Building 205, 1901 South 1st Street, Temple, Texas 76504. fax: (254) 743-0378.
| |
Collapse
|
35
|
Pezeshkian W, Chevrot G, Khandelia H. The role of caveolin-1 in lipid droplets and their biogenesis. Chem Phys Lipids 2018; 211:93-99. [DOI: 10.1016/j.chemphyslip.2017.11.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/03/2017] [Accepted: 11/09/2017] [Indexed: 12/21/2022]
|
36
|
Monson EA, Crosse KM, Das M, Helbig KJ. Lipid droplet density alters the early innate immune response to viral infection. PLoS One 2018; 13:e0190597. [PMID: 29293661 PMCID: PMC5749834 DOI: 10.1371/journal.pone.0190597] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/17/2017] [Indexed: 12/30/2022] Open
Abstract
The cellular localisation of many innate signalling events following viral infection has yet to be elucidated, however there has been a few cases in which membranes of certain cellular organelles have acted as platforms to these events. Of these, lipid droplets (LDs) have recently been identified as signalling platforms for innate TLR7 and 9 signalling. Despite their wide range of similar roles in various metabolic pathways, LDs have been overlooked as potential platforms for antiviral innate signalling events. This study established an in vitro model to evaluate the efficiency of the early innate immune response in cells with reduced LD content to the viral mimics, dsDNA and dsRNA, and Sendai viral infection. Using RT-qPCR, the expression of IFN-β and IFN-λ was quantified following stimulation along with the expression of specific ISGs. Luciferase based assays evaluated the combined expression of ISRE-promoter driven ISGs under IFN-β stimulation. Cellular LD content did not alter the entry of fluorescently labelled viral mimics into cells, but significantly decreased the ability of both Huh-7 and HeLa cells to produce type I and III IFN, as well as downstream ISG expression, indicative of an impeded innate immune response. This observation was also seen during Sendai virus infection of HeLa cells, where both control and LD reduced cells replicated the virus to the same level, but a significantly impaired type I and III IFN response was observed in the LD reduced cells. In addition to altered IFN production, cells with reduced LD content exhibited decreased expression of specific antiviral ISGs: Viperin, IFIT-1 and OAS-1 under IFN-β stimulation; However the overall induction of the ISRE-promoter was not effected. This study implicates a role for LDs in an efficient early innate host response to viral infection and future work will endeavour to determine the precise role these important organelles play in induction of an antiviral response.
Collapse
Affiliation(s)
- Ebony A Monson
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria
| | - Keaton M Crosse
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria
| | - Mithun Das
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria
| | - Karla J Helbig
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria
| |
Collapse
|
37
|
Abstract
Triglyceride molecules represent the major form of storage and transport of fatty acids within cells and in the plasma. The liver is the central organ for fatty acid metabolism. Fatty acids accrue in liver by hepatocellular uptake from the plasma and by de novo biosynthesis. Fatty acids are eliminated by oxidation within the cell or by secretion into the plasma within triglyceride-rich very low-density lipoproteins. Notwithstanding high fluxes through these pathways, under normal circumstances the liver stores only small amounts of fatty acids as triglycerides. In the setting of overnutrition and obesity, hepatic fatty acid metabolism is altered, commonly leading to the accumulation of triglycerides within hepatocytes, and to a clinical condition known as nonalcoholic fatty liver disease (NAFLD). In this review, we describe the current understanding of fatty acid and triglyceride metabolism in the liver and its regulation in health and disease, identifying potential directions for future research. Advances in understanding the molecular mechanisms underlying the hepatic fat accumulation are critical to the development of targeted therapies for NAFLD. © 2018 American Physiological Society. Compr Physiol 8:1-22, 2018.
Collapse
Affiliation(s)
- Michele Alves-Bezerra
- Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, USA
| | - David E Cohen
- Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, USA
| |
Collapse
|
38
|
Desai AJ, Miller LJ. Changes in the plasma membrane in metabolic disease: impact of the membrane environment on G protein-coupled receptor structure and function. Br J Pharmacol 2017; 175:4009-4025. [PMID: 28691227 DOI: 10.1111/bph.13943] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/08/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022] Open
Abstract
Drug development targeting GPCRs often utilizes model heterologous cell expression systems, reflecting an implicit assumption that the membrane environment has little functional impact on these receptors or on their responsiveness to drugs. However, much recent data have illustrated that membrane components can have an important functional impact on intrinsic membrane proteins. This review is directed toward gaining a better understanding of the structure of the plasma membrane in health and disease, and how this organelle can influence GPCR structure, function and regulation. It is important to recognize that the membrane provides a potential mode of lateral allosteric regulation of GPCRs and can affect the effectiveness of drugs and their biological responses in various disease states, which can even vary among individuals across the population. The type 1 cholecystokinin receptor is reviewed as an exemplar of a class A GPCR that is affected in this way by changes in the plasma membrane. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
39
|
The Role of Caveolin 1 in HIV Infection and Pathogenesis. Viruses 2017; 9:v9060129. [PMID: 28587148 PMCID: PMC5490806 DOI: 10.3390/v9060129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/02/2017] [Accepted: 05/22/2017] [Indexed: 12/29/2022] Open
Abstract
Caveolin 1 (Cav-1) is a major component of the caveolae structure and is expressed in a variety of cell types including macrophages, which are susceptible to human immunodeficiency virus (HIV) infection. Caveolae structures are present in abundance in mechanically stressed cells such as endothelial cells and adipocytes. HIV infection induces dysfunction of these cells and promotes pathogenesis. Cav-1 and the caveolae structure are believed to be involved in multiple cellular processes that include signal transduction, lipid regulation, endocytosis, transcytosis, and mechanoprotection. Such a broad biological role of Cav-1/caveolae is bound to have functional cross relationships with several molecular pathways including HIV replication and viral-induced pathogenesis. The current review covers the relationship of Cav-1 and HIV in respect to viral replication, persistence, and the potential role in pathogenesis.
Collapse
|
40
|
Caveolin-1: An Oxidative Stress-Related Target for Cancer Prevention. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7454031. [PMID: 28546853 PMCID: PMC5436035 DOI: 10.1155/2017/7454031] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/23/2017] [Accepted: 03/07/2017] [Indexed: 01/19/2023]
Abstract
Aberrant oxidative metabolism is one of the hallmarks of cancer. Reactive species overproduction could promote carcinogenesis via inducing genetic mutations and activating oncogenic pathways, and thus, antioxidant therapy was considered as an important strategy for cancer prevention and treatment. Caveolin-1 (Cav-1), a constituent protein of caveolae, has been shown to mediate tumorigenesis and progression through oxidative stress modulation recently. Reactive species could modulate the expression, degradation, posttranslational modifications, and membrane trafficking of Cav-1, while Cav-1-targeted treatments could scavenge the reactive species. More importantly, emerging evidences have indicated that multiple antioxidants could exert antitumor activities in cancer cells and protective activities in normal cells by modulating the Cav-1 pathway. Altogether, these findings indicate that Cav-1 may be a promising oxidative stress-related target for cancer antioxidant prevention. Elucidating the underlying interaction mechanisms between oxidative stress and Cav-1 is helpful for enhancing the preventive effects of antioxidants on cancer, for improving clinical outcomes of antioxidant-related therapeutics in cancer patients, and for developing Cav-1 targeted drugs. Herein, we summarize the available evidence of the roles of Cav-1 and oxidative stress in tumorigenesis and development and shed novel light on designing strategies for cancer prevention or treatment by utilizing the interaction mode between Cav-1 and oxidative stress.
Collapse
|
41
|
Ludwig A, Nguyen TH, Leong D, Ravi LI, Tan BH, Sandin S, Sugrue RJ. Caveolae provide a specialized membrane environment for respiratory syncytial virus assembly. J Cell Sci 2017; 130:1037-1050. [PMID: 28154158 PMCID: PMC5358342 DOI: 10.1242/jcs.198853] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/26/2017] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is an enveloped virus that assembles into filamentous virus particles on the surface of infected cells. Morphogenesis of RSV is dependent upon cholesterol-rich (lipid raft) membrane microdomains, but the specific role of individual raft molecules in RSV assembly is not well defined. Here, we show that RSV morphogenesis occurs within caveolar membranes and that both caveolin-1 and cavin-1 (also known as PTRF), the two major structural and functional components of caveolae, are actively recruited to and incorporated into the RSV envelope. The recruitment of caveolae occurred just prior to the initiation of RSV filament assembly, and was dependent upon an intact actin network as well as a direct physical interaction between caveolin-1 and the viral G protein. Moreover, cavin-1 protein levels were significantly increased in RSV-infected cells, leading to a virus-induced change in the stoichiometry and biophysical properties of the caveolar coat complex. Our data indicate that RSV exploits caveolae for its assembly, and we propose that the incorporation of caveolae into the virus contributes to defining the biological properties of the RSV envelope.
Collapse
Affiliation(s)
- Alexander Ludwig
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Tra Huong Nguyen
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Daniel Leong
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510
| | - Laxmi Iyer Ravi
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Boon Huan Tan
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510
| | - Sara Sandin
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| |
Collapse
|
42
|
García-Ruiz C, Ribas V, Baulies A, Fernández-Checa JC. Mitochondrial Cholesterol and the Paradox in Cell Death. Handb Exp Pharmacol 2017; 240:189-210. [PMID: 28035533 DOI: 10.1007/164_2016_110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondria are considered cholesterol-poor organelles, and obtain their cholesterol load by the action of specialized proteins involved in its delivery from extramitochondrial sources and trafficking within mitochondrial membranes. Although mitochondrial cholesterol fulfills vital physiological functions, such as the synthesis of bile acids in the liver or the formation of steroid hormones in specialized tissues, recent evidence indicates that the accumulation of cholesterol in mitochondria may be a key event in prevalent human diseases, in particular in the development of steatohepatitis (SH) and its progression to hepatocellular carcinoma (HCC). Mitochondrial cholesterol accumulation promotes the transition from simple steatosis to SH due to the sensitization to oxidative stress and cell death. However, mitochondrial cholesterol loading in HCC determines apoptosis resistance and insensitivity to chemotherapy. These opposing functions of mitochondrial cholesterol in SH and HCC define its paradoxical role in cell death as a pro- and anti-apoptotic factor. Further understanding of this conundrum may be useful to modulate the progression from SH to HCC by targeting mitochondrial cholesterol trafficking.
Collapse
Affiliation(s)
- Carmen García-Ruiz
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Keck School of Medicine, USC, University of Southern California Research Center for Alcohol Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, CA, USA
| | - Vicente Ribas
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Anna Baulies
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Jose C Fernández-Checa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomedicas de Barcelona, CSIC, C/Rosello 161, 08036, Barcelona, Spain.
- Liver Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain.
- Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain.
- Keck School of Medicine, USC, University of Southern California Research Center for Alcohol Liver and Pancreatic Diseases and Cirrhosis, Los Angeles, CA, USA.
| |
Collapse
|
43
|
Modzel M, Lund FW, Wüstner D. Synthesis and Live-Cell Imaging of Fluorescent Sterols for Analysis of Intracellular Cholesterol Transport. Methods Mol Biol 2017; 1583:111-140. [PMID: 28205171 DOI: 10.1007/978-1-4939-6875-6_10] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Cellular cholesterol homeostasis relies on precise control of the sterol content of organelle membranes. Obtaining insight into cholesterol trafficking pathways and kinetics by live-cell imaging relies on two conditions. First, one needs to develop suitable analogs that resemble cholesterol as closely as possible with respect to their biophysical and biochemical properties. Second, the cholesterol analogs should have good fluorescence properties. This interferes, however, often with the first requirement, such that the imaging instrumentation must be optimized to collect photons from suboptimal fluorophores, but good cholesterol mimics, such as the intrinsically fluorescent sterols, cholestatrienol (CTL) or dehydroergosterol (DHE). CTL differs from cholesterol only in having two additional double bonds in the ring system, which is why it is slightly fluorescent in the ultraviolet (UV). In the first part of this protocol, we describe how to synthesize and image CTL in living cells relative to caveolin, a structural component of caveolae. In the second part, we explain in detail how to perform time-lapse experiments of commercially available BODIPY-tagged cholesterol (TopFluor-cholesterol®; TF-Chol) in comparison to DHE. Finally, using two-photon time-lapse imaging data of TF-Chol, we demonstrate how to use our imaging toolbox SpatTrack for tracking sterol rich vesicles in living cells over time.
Collapse
Affiliation(s)
- Maciej Modzel
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, 5230, Denmark
| | - Frederik W Lund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, 5230, Denmark.,Department of Biochemistry, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, 5230, Denmark.
| |
Collapse
|
44
|
Natarajan SK, Rasineni K, Ganesan M, Feng D, McVicker BL, McNiven MA, Osna NA, Mott JL, Casey CA, Kharbanda KK. Structure, Function and Metabolism of Hepatic and Adipose Tissue Lipid Droplets: Implications in Alcoholic Liver Disease. Curr Mol Pharmacol 2017; 10:237-248. [PMID: 26278390 PMCID: PMC4820363 DOI: 10.2174/1874467208666150817111727] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 02/08/2023]
Abstract
For more than 30 years, lipid droplets (LDs) were considered as an inert bag of lipid for storage of energy-rich fat molecules. Following a paradigm shift almost a decade ago, LDs are presently considered an active subcellular organelle especially designed for assembling, storing and subsequently supplying lipids for generating energy and membrane synthesis (and in the case of hepatocytes for VLDL secretion). LDs also play a central role in many other cellular functions such as viral assembly and protein degradation. Here, we have explored the structural and functional changes that occur in hepatic and adipose tissue LDs following chronic ethanol consumption in relation to their role in the pathogenesis of alcoholic liver injury.
Collapse
Affiliation(s)
- Sathish Kumar Natarajan
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| | - Karuna Rasineni
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Murali Ganesan
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dan Feng
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Benita L. McVicker
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Natalia A. Osna
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Justin L. Mott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| | - Carol A. Casey
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| | - Kusum K. Kharbanda
- Research Service, VA Nebraska-Western Iowa Health Care System (VA NWIHCS), and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center
| |
Collapse
|
45
|
Janković J, Tatić S, Božić V, Živaljević V, Cvejić D, Paskaš S. Inverse expression of caveolin-1 and EGFR in thyroid cancer patients. Hum Pathol 2016; 61:164-172. [PMID: 27818286 DOI: 10.1016/j.humpath.2016.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/23/2016] [Accepted: 10/14/2016] [Indexed: 01/21/2023]
Abstract
Cytological analysis of fine-needle aspiration (FNA) is the first step in evaluation of patients with nodular thyroid disease with the primary goal to exclude thyroid malignancy. Its improvement by combining cytology with molecular markers is still a matter of investigation. In this study, 2 molecular markers were used: caveolin-1 and epidermal growth factor receptor (EGFR), along with the well-established genetic marker BRAF V600E mutation. We set out to determine the expression signatures of EGFR and caveolin-1 in patients with thyroid malignancy as well as to determine their possible association with disease severity. In FNA biopsy samples (n=186), immunocytochemical expression of caveolin-1 and BRAF V600E mutation coincided with malignancy. The patients were sorted according to 3 parameters: final histopathological diagnosis, caveolin-1 expression, and BRAF V600E mutation status before measurement of EGFR mRNA expression. EGFR upregulation was detected in the group of patients with malignant diagnosis, no caveolin-1 expression, and wild-type BRAF. Spearman rank correlation yielded a statistically significant negative correlation of EGFR and caveolin-1. Double immunofluorescence confirmed colocalization and inverse expression of EGFR and caveolin-1. Our data demonstrated that EGFR overexpression is associated with malignancy but not with tumor aggressiveness. Furthermore, high-caveolin-1/low-EGFR cases were associated with an advanced pT status and had a greater degree of neoplastic infiltration than low-caveolin-1/high-EGFR cases. Combining caveolin-1 and BRAF V600E with EGFR might help in recognizing more aggressive thyroid lesions in a pool of relatively indolent tumors in FNA biopsies and thus be useful for early risk stratification of thyroid cancer patients.
Collapse
Affiliation(s)
- Jelena Janković
- Institute for the Application of Nuclear Energy-INEP, Department for Endocrinology and Radioimmunology, University of Belgrade, 11080 Belgrade, Serbia
| | - Svetislav Tatić
- Institute of Pathology, Medical Faculty, University of Belgrade, 11000 Belgrade, Serbia
| | - Vesna Božić
- Clinical Center of Serbia, Department of Endocrine and Cardiovascular Pathology, 11000 Belgrade, Serbia
| | - Vladan Živaljević
- Center for Endocrine Surgery, Institute of Endocrinology, Diabetes and Diseases of Metabolism, University of Belgrade, 11000 Belgrade, Serbia
| | - Dubravka Cvejić
- Institute for the Application of Nuclear Energy-INEP, Department for Endocrinology and Radioimmunology, University of Belgrade, 11080 Belgrade, Serbia
| | - Svetlana Paskaš
- Institute for the Application of Nuclear Energy-INEP, Department for Endocrinology and Radioimmunology, University of Belgrade, 11080 Belgrade, Serbia.
| |
Collapse
|
46
|
Atwal S, Giengkam S, VanNieuwenhze M, Salje J. Live imaging of the genetically intractable obligate intracellular bacteria Orientia tsutsugamushi using a panel of fluorescent dyes. J Microbiol Methods 2016; 130:169-176. [PMID: 27582280 PMCID: PMC5073074 DOI: 10.1016/j.mimet.2016.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/23/2016] [Accepted: 08/23/2016] [Indexed: 12/20/2022]
Abstract
Our understanding of the molecular mechanisms of bacterial infection and pathogenesis are disproportionally derived from a small number of well-characterised species and strains. One reason for this is the enormous time and resources required to develop a new organism into experimental system that can be interrogated at the molecular level, in particular with regards to the development of genetic tools. Live cell imaging by fluorescence microscopy is a powerful technique to study biological processes such as bacterial motility, host cell invasion, and bacterial growth and division. In the absence of genetic tools that enable exogenous expression of fluorescent proteins, fluorescent chemical probes can be used to label and track living cells. A large number of fluorescent chemical probes are commercially available, but these have overwhelmingly been applied to the study of eukaryotic cell systems. Here, we present a methodical analysis of four different classes of probes, which can be used to delineate the cytoplasm, nucleic acids, cell membrane or peptidoglycan of living bacterial cells. We have tested these in the context of the important but neglected human pathogen Orientia tsutsugamushi but expect that the methodology would be broadly applicable to other bacterial species. A panel of fluorescent dyes are used to label living cells of the obligate intracellular bacterium Orientia tsutsugamushi. These dyes include CFSE, SYTO9, DiI/DiO/DiD and HADA. These dyes label the bacterial cytoplasm, nucleic acids, lipid membrane and cell wall respectively.
Collapse
Affiliation(s)
- Sharanjeet Atwal
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Suparat Giengkam
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Jeanne Salje
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
47
|
Gao L, Chen X, Peng T, Yang D, Wang Q, Lv Z, Shen J. Caveolin-1 protects against hepatic ischemia/reperfusion injury through ameliorating peroxynitrite-mediated cell death. Free Radic Biol Med 2016; 95:209-15. [PMID: 27021966 DOI: 10.1016/j.freeradbiomed.2016.03.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/23/2016] [Accepted: 03/23/2016] [Indexed: 12/24/2022]
Abstract
Nitrative stress is considered as an important pathological process of hepatic ischemia and reperfusion injury but its regulating mechanisms are largely unknown. In this study, we tested the hypothesis that caveolin-1 (Cav-1), a plasma membrane scaffolding protein, could be an important cellular signaling against hepatic I/R injury through inhibiting peroxynitrite (ONOO(-))-induced cellular damage. Male wild-type mice and Cav-1 knockout (Cav-1(-/-)) were subjected to 1h hepatic ischemia following 1, 6 and 12h of reperfusion by clipping and releasing portal vessels respectively. Immortalized human hepatocyte cell line (L02) was subjected to 1h hypoxia and 6h reoxygenation and treated with Cav-1 scaffolding domain peptide. The major discoveries included: (1) the expression of Cav-1 in serum and liver tissues of wild-type mice was time-dependently elevated during hepatic ischemia-reperfusion injury. (2) Cav-1 scaffolding domain peptide treatment inhibited cleaved caspase-3 expression in the hypoxia-reoxygenated L02 cells; (3) Cav-1 knockout (Cav-1(-/-)) mice had significantly higher levels of serum transaminases (ALT&AST) and TNF-α, and higher rates of apoptotic cell death in liver tissues than wild-type mice after subjected to 1h hepatic ischemia and 6hour reperfusion; (4) Cav-1(-/-) mice revealed higher expression levels of iNOS, ONOO(-) and 3-nitrotyrosine (3-NT) in the liver than wild-type mice, and Fe-TMPyP, a representative peroxynitrite decomposition catalyst (PDC), remarkably reduced level of ONOO(-) and 3-NT and ameliorated the serum ALT, AST and TNF-α levels in both wild-type and Cav-1(-/-) mice. Taken together, we conclude that Cav-1 could play a critical role in preventing nitrative stress-induced liver damage during hepatic ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Lei Gao
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xingmiao Chen
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; Research Centre of Heart, Brain, Hormone & Healthy Aging, the University of Hong Kong, Hong Kong, China
| | - Tao Peng
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Dan Yang
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; Research Centre of Heart, Brain, Hormone & Healthy Aging, the University of Hong Kong, Hong Kong, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
48
|
Lange Y, Steck TL. Active membrane cholesterol as a physiological effector. Chem Phys Lipids 2016; 199:74-93. [PMID: 26874289 DOI: 10.1016/j.chemphyslip.2016.02.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 02/05/2023]
Abstract
Sterols associate preferentially with plasma membrane sphingolipids and saturated phospholipids to form stoichiometric complexes. Cholesterol in molar excess of the capacity of these polar bilayer lipids has a high accessibility and fugacity; we call this fraction active cholesterol. This review first considers how active cholesterol serves as an upstream regulator of cellular sterol homeostasis. The mechanism appears to utilize the redistribution of active cholesterol down its diffusional gradient to the endoplasmic reticulum and mitochondria, where it binds multiple effectors and directs their feedback activity. We have also reviewed a broad literature in search of a role for active cholesterol (as opposed to bulk cholesterol or lipid domains such as rafts) in the activity of diverse membrane proteins. Several systems provide such evidence, implicating, in particular, caveolin-1, various kinds of ABC-type cholesterol transporters, solute transporters, receptors and ion channels. We suggest that this larger role for active cholesterol warrants close attention and can be tested easily.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, 1653 W. Congress Parkway, Chicago, IL 60612, USA.
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
49
|
Exercise-Induced Changes in Caveolin-1, Depletion of Mitochondrial Cholesterol, and the Inhibition of Mitochondrial Swelling in Rat Skeletal Muscle but Not in the Liver. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3620929. [PMID: 26839631 PMCID: PMC4709766 DOI: 10.1155/2016/3620929] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/22/2015] [Accepted: 10/11/2015] [Indexed: 01/09/2023]
Abstract
The reduction in cholesterol in mitochondria, observed after exercise, is related to the inhibition of mitochondrial swelling. Caveolin-1 (Cav-1) plays an essential role in the regulation of cellular cholesterol metabolism and is required by various signalling pathways. Therefore, the aim of this study was to investigate the effect of prolonged swimming on the mitochondrial Cav-1 concentration; additionally, we identified the results of these changes as they relate to the induction of changes in the mitochondrial swelling and cholesterol in rat skeletal muscle and liver. Male Wistar rats were divided into a sedentary control group and an exercise group. The exercised rats swam for 3 hours and were burdened with an additional 3% of their body weight. After the cessation of exercise, their quadriceps femoris muscles and livers were immediately removed for experimentation. The exercise protocol caused an increase in the Cav-1 concentration in crude muscle mitochondria; this was related to a reduction in the cholesterol level and an inhibition of mitochondrial swelling. There were no changes in rat livers, with the exception of increased markers of oxidative stress in mitochondria. These data indicate the possible role of Cav-1 in the adaptive change in the rat muscle mitochondria following exercise.
Collapse
|
50
|
Ariotti N, Hall TE, Rae J, Ferguson C, McMahon KA, Martel N, Webb RE, Webb RI, Teasdale RD, Parton RG. Modular Detection of GFP-Labeled Proteins for Rapid Screening by Electron Microscopy in Cells and Organisms. Dev Cell 2015; 35:513-25. [PMID: 26585296 DOI: 10.1016/j.devcel.2015.10.016] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/16/2015] [Accepted: 10/19/2015] [Indexed: 10/22/2022]
Abstract
Reliable and quantifiable high-resolution protein localization is critical for understanding protein function. However, the time required to clone and characterize any protein of interest is a significant bottleneck, especially for electron microscopy (EM). We present a modular system for enzyme-based protein tagging that allows for improved speed and sampling for analysis of subcellular protein distributions using existing clone libraries to EM-resolution. We demonstrate that we can target a modified soybean ascorbate peroxidase (APEX) to any GFP-tagged protein of interest by engineering a GFP-binding peptide (GBP) directly to the APEX-tag. We demonstrate that APEX-GBP (1) significantly reduces the time required to characterize subcellular protein distributions of whole libraries to less than 3 days, (2) provides remarkable high-resolution localization of proteins to organelle subdomains, and (3) allows EM localization of GFP-tagged proteins, including proteins expressed at endogenous levels, in vivo by crossing existing GFP-tagged transgenic zebrafish lines with APEX-GBP transgenic lines.
Collapse
Affiliation(s)
- Nicholas Ariotti
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia
| | - Thomas E Hall
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia
| | - James Rae
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia
| | - Charles Ferguson
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia
| | - Kerrie-Ann McMahon
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia
| | - Nick Martel
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia
| | - Robyn E Webb
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD 4072, Australia
| | - Richard I Webb
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD 4072, Australia
| | - Rohan D Teasdale
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, University of Queensland, QLD 4072, Australia; Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|