1
|
Fan J, Zheng S, Wang M, Yuan X. The critical roles of caveolin-1 in lung diseases. Front Pharmacol 2024; 15:1417834. [PMID: 39380904 PMCID: PMC11458383 DOI: 10.3389/fphar.2024.1417834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Caveolin-1 (Cav-1), a structural and functional component in the caveolae, plays a critical role in transcytosis, endocytosis, and signal transduction. Cav-1 has been implicated in the mediation of cellular processes by interacting with a variety of signaling molecules. Cav-1 is widely expressed in the endothelial cells, smooth muscle cells, and fibroblasts in the various organs, including the lungs. The Cav-1-mediated internalization and regulation of signaling molecules participate in the physiological and pathological processes. Particularly, the MAPK, NF-κB, TGFβ/Smad, and eNOS/NO signaling pathways have been involved in the regulatory effects of Cav-1 in lung diseases. The important effects of Cav-1 on the lungs indicate that Cav-1 can be a potential target for the treatment of lung diseases. A Cav-1 scaffolding domain peptide CSP7 targeting Cav-1 has been developed. In this article, we mainly discuss the structure of Cav-1 and its critical roles in lung diseases, such as pneumonia, acute lung injury (ALI), asthma, chronic obstructive pulmonary disease (COPD), pulmonary hypertension, pulmonary fibrosis, and lung cancer.
Collapse
Affiliation(s)
| | | | | | - Xiaoliang Yuan
- Department of Respiratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
2
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
3
|
Ge S, Lian W, Bai Y, Wang L, Zhao F, Li H, Wang D, Pang Q. TMT-based quantitative proteomics reveals the targets of andrographolide on LPS-induced liver injury. BMC Vet Res 2023; 19:199. [PMID: 37817228 PMCID: PMC10563216 DOI: 10.1186/s12917-023-03758-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/27/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Andrographolide (Andro) is a diterpenoid derived from Andrographis paniculate, which has anti-inflammatory, antibacterial, antiviral and hepatoprotective activities. Gram-negative bacterial infections can cause varying degrees of liver injury in chickens, although Andro has been shown to have a protective effect on the liver, its underlying mechanism of action and effects on liver proteins are not known. METHODS The toxicity of Andro on the viability of leghorn male hepatoma (LMH) cells at different concentrations and times was analyzed by CCK-8 assays. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities in the culture supernatants were measured using an automatic biochemical analyzer to evaluate the protective effect of androscopolide on LPS-induced injury of LMH cells. Subsequently, TMT proteomics analysis were performed on the negative control group (NC group), LPS, and LPS-Andro groups, and bioinformatics analysis was performed on the differentially expressed proteins (DEPs). RESULTS It was found that Andro reduced ALT and AST levels in the cell supernatant and alleviated LPS-induced injury in LMH cells. Proteomic analysis identified 50 and 166 differentially expressed proteins in the LPS vs. NC group and LPS-Andro vs. LPS group, respectively. Andro may be involved in steroid metabolic processes, negative regulation of MAPK cascade, oxidative stress, and other processes to protect against LPS-induced liver injury. CONCLUSIONS Andro protects against LPS-induced liver injury, HMGCS1, HMGCR, FDPS, PBK, CAV1, PRDX1, PRDX4, and PRDX6, which were identified by differential proteomics, may be the targets of Andro. Our study may provide new theoretical support for Andro protection against liver injury.
Collapse
Affiliation(s)
- Shihao Ge
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
- College of Pharmacy, Heze University, Heze, 274000, Shangdong, China
| | - Wenqi Lian
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yongjiang Bai
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Linzheng Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250035, Shangdong, China
| | - Fuwei Zhao
- College of Pharmacy, Heze University, Heze, 274000, Shangdong, China
| | - Houmei Li
- Shuozhou grass and animal husbandry development center, ShuoZhou, 036000, Shanxi, China
| | - Dongliang Wang
- ShuoZhou Vocational Technology College, ShuoZhou, 036000, Shanxi, China
| | - Quanhai Pang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China.
| |
Collapse
|
4
|
Shu Y, Jin S. Caveolin-1 in endothelial cells: A potential therapeutic target for atherosclerosis. Heliyon 2023; 9:e18653. [PMID: 37554846 PMCID: PMC10405014 DOI: 10.1016/j.heliyon.2023.e18653] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/10/2023] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease characterized by lipid accumulation and the activation of the inflammatory response; it remains the leading nation-wide cause of death. Early in the progression of AS, stimulation by pro-inflammatory agonists (TNF-α, LPS, and others), oxidized lipoproteins (ox-LDL), and biomechanical stimuli (low shear stress) lead to endothelial cell activation and dysfunction. Consequently, it is crucial to investigate how endothelial cells respond to different stressors and ways to alter endothelial cell activation in AS development, as they are the earliest cells to respond. Caveolin-1 (Cav1) is a 21-24-kDa membrane protein located in caveolae and highly expressed in endothelial cells, which plays a vital role in regulating lipid transport, inflammatory responses, and various cellular signaling pathways and has atherogenic effects. This review summarizes recent studies on the structure and physiological functions of Cav1 and outlines the potential mechanisms it mediates in AS development. Included are the roles of Cav1 in the regulation of endothelial cell autophagy, response to shear stress, modulation of the eNOS/NO axis, and transduction of inflammatory signaling pathways. This review provides a rationale for proposing Cav1 as a novel target for the prevention of AS, as well as new ideas for therapeutic strategies for early AS.
Collapse
Affiliation(s)
- Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, 430077, China
| |
Collapse
|
5
|
Puddu A, Montecucco F, Maggi D. Caveolin-1 and Atherosclerosis: Regulation of LDLs Fate in Endothelial Cells. Int J Mol Sci 2023; 24:ijms24108869. [PMID: 37240214 DOI: 10.3390/ijms24108869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Caveolae are 50-100 nm cell surface plasma membrane invaginations observed in terminally differentiated cells. They are characterized by the presence of the protein marker caveolin-1. Caveolae and caveolin-1 are involved in regulating several signal transduction pathways and processes. It is well recognized that they have a central role as regulators of atherosclerosis. Caveolin-1 and caveolae are present in most of the cells involved in the development of atherosclerosis, including endothelial cells, macrophages, and smooth muscle cells, with evidence of either pro- or anti-atherogenic functions depending on the cell type examined. Here, we focused on the role of caveolin-1 in the regulation of the LDLs' fate in endothelial cells.
Collapse
Affiliation(s)
- Alessandra Puddu
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Davide Maggi
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy
| |
Collapse
|
6
|
Qiu Y, Shi YN, Zhu N, Zhang S, Zhang CJ, Gu J, He P, Dai AG, Qin L. A Lipid Perspective on Regulated Pyroptosis. Int J Biol Sci 2023; 19:2333-2348. [PMID: 37215994 PMCID: PMC10197892 DOI: 10.7150/ijbs.81017] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/03/2023] [Indexed: 05/24/2023] Open
Abstract
Pyroptosis is a novel pro-inflammatory cell programmed death dependent on Gasdermin (GSMD) family-mediated membrane pore formation and subsequent cell lysis, accompanied by the release of inflammatory factors and expanding inflammation in multiple tissues. All of these processes have impacts on a variety of metabolic disorders. Dysregulation of lipid metabolism is one of the most prominent metabolic alterations in many diseases, including the liver, cardiovascular system, and autoimmune diseases. Lipid metabolism produces many bioactive lipid molecules, which are important triggers and endogenous regulators of pyroptosis. Bioactive lipid molecules promote pyroptosis through intrinsic pathways involving reactive oxygen species (ROS) production, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, lysosomal disruption, and the expression of related molecules. Pyroptosis can also be regulated during the processes of lipid metabolism, including lipid uptake and transport, de novo synthesis, lipid storage, and lipid peroxidation. Taken together, understanding the correlation between lipid molecules such as cholesterol and fatty acids and pyroptosis during metabolic processes can help to gain insight into the pathogenesis of many diseases and develop effective strategies from the perspective of pyroptosis.
Collapse
Affiliation(s)
- Yun Qiu
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ya-Ning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Science and Technology Innovation Center, Hunan University of Chinese Medicine, 410208, Changsha, Hunan, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Shuo Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chan-Juan Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jia Gu
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Peng He
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ai-Guo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
- Department of Respiratory Medicine, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| |
Collapse
|
7
|
Choi H, Yang SW, Joo JS, Park M, Jin Y, Kim JW, Lee SY, Lee SV, Yun TJ, Cho ML, Hwang HS, Kang YS. Sialylated IVIg binding to DC-SIGN + Hofbauer cells induces immune tolerance through the caveolin-1/NF-kB pathway and IL-10 secretion. Clin Immunol 2023; 246:109215. [PMID: 36581222 DOI: 10.1016/j.clim.2022.109215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/15/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Although the use of IVIg has increased in various immune-driven diseases and even in pregnancy, the exact action mechanisms of IVIg are not fully understood. Dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN) is a known receptor for α-2,6-sialylated IgG (sIVIg), which is responsible for the anti-inflammatory effect of IVIg. DC-SIGN is expressed on Hofbauer cells (HBCs) of the fetal villi of the placenta which act as an innate immune modulator at the maternal-fetal interface. Preeclampsia is a major complication in pregnancy and is related to IL-10, a cytokine with an important role in immune tolerance. DC-SIGN interaction with sIVIg in HBCs promoted IL-10 secretion through the activation of the caveolin-1/NF-κB pathway, especially in plasma lipid rafts. Consistent results were obtained for HBCs from patients with preeclampsia. Collectively, the stimulation of DC-SIGN+ HBCs with sIVIg enhanced immune tolerance in the feto-maternal environment, suggesting the therapeutic application of sIVIg to prevent preeclampsia.
Collapse
Affiliation(s)
- Hyeongjwa Choi
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seung-Woo Yang
- Department of Obstetrics and Gynecology, Sang-Gye Paik Hospital, Inje University School of Medicine; Seoul 01757, Republic of Korea
| | - Jin-Soo Joo
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Min Park
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Yihua Jin
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Ji-Woon Kim
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seon-Yeong Lee
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Vin Lee
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Tae-Jin Yun
- Department of Pathology, New York University Grossman School of Medicine; New York, NY 10016, USA
| | - Mi-La Cho
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, South Korea
| | - Han-Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine; Seoul, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Young-Sun Kang
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; KU Research Center for Zoonosis, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea.
| |
Collapse
|
8
|
5,2′-Dibromo-2,4′,5′-trihydroxydiphenylmethanone Inhibits LPS-Induced Vascular Inflammation by Targeting the Cav1 Protein. Molecules 2022; 27:molecules27092884. [PMID: 35566232 PMCID: PMC9101869 DOI: 10.3390/molecules27092884] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular inflammation is directly responsible for atherosclerosis. 5,2′-Dibromo-2,4′,5′-trihydroxydiphenylmethanone (TDD), a synthetic bromophenol derivative, exhibits anti-atherosclerosis and anti-inflammatory effects. However, the underlying pathways are not yet clear. In this study, we first examined the effects of TDD on toll-like receptor-4 (TLR4) activity, the signaling receptor for lipopolysaccharide (LPS), and found that TDD does not inhibit LPS-induced TLR4 expression in EA.hy926 cells and the vascular wall in vivo. Next, we investigated the global protein alterations and the mechanisms underlying the action of TDD in LPS-treated EA.hy926 cells using an isobaric tag for the relative and absolute quantification technique. Western blot analysis revealed that TDD inhibited NF-κB activation by regulating the phosphorylation and subsequent degradation IκBα. Among the differentially expressed proteins, TDD concentration-dependently inhibited Caveolin 1(Cav1) expression. The interaction between Cav1 and TDD was determined by using biolayer interference assay, UV-vis absorption spectra, fluorescence spectrum, and molecular docking. We found that TDD can directly bind to Cav1 through hydrogen bonds and van der Waals forces. In conclusion, our results showed that TDD inhibited LPS-induced vascular inflammation and the NF-κB signaling pathway by specifically targeting the Cav1 protein. TDD may be a novel anti-inflammatory compound, especially for the treatment of atherosclerosis.
Collapse
|
9
|
Paukner K, Králová Lesná I, Poledne R. Cholesterol in the Cell Membrane-An Emerging Player in Atherogenesis. Int J Mol Sci 2022; 23:533. [PMID: 35008955 PMCID: PMC8745363 DOI: 10.3390/ijms23010533] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Membrane cholesterol is essential for cell membrane properties, just as serum cholesterol is important for the transport of molecules between organs. This review focuses on cholesterol transport between lipoproteins and lipid rafts on the surface of macrophages. Recent studies exploring this mechanism and recognition of the central dogma-the key role of macrophages in cardiovascular disease-have led to the notion that this transport mechanism plays a major role in the pathogenesis of atherosclerosis. The exact molecular mechanism of this transport remains unclear. Future research will improve our understanding of the molecular and cellular bases of lipid raft-associated cholesterol transport.
Collapse
Affiliation(s)
- Karel Paukner
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.K.L.); (R.P.)
- Department of Physiology, Faculty of Science, Charles University, 128 44 Prague, Czech Republic
- Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Small Animal Clinic, 612 00 Brno, Czech Republic
| | - Ivana Králová Lesná
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.K.L.); (R.P.)
- Department of Anesthesia and Intensive Medicine, First Faculty of Medicine, Charles University and University Military Hospital, 128 08 Prague, Czech Republic
| | - Rudolf Poledne
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (I.K.L.); (R.P.)
| |
Collapse
|
10
|
Kotlyarov S. Diversity of Lipid Function in Atherogenesis: A Focus on Endothelial Mechanobiology. Int J Mol Sci 2021; 22:11545. [PMID: 34768974 PMCID: PMC8584259 DOI: 10.3390/ijms222111545] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is one of the most important problems in modern medicine. Its high prevalence and social significance determine the need for a better understanding of the mechanisms of the disease's development and progression. Lipid metabolism and its disorders are one of the key links in the pathogenesis of atherosclerosis. Lipids are involved in many processes, including those related to the mechanoreception of endothelial cells. The multifaceted role of lipids in endothelial mechanobiology and mechanisms of atherogenesis are discussed in this review. Endothelium is involved in ensuring adequate vascular hemodynamics, and changes in blood flow characteristics are detected by endothelial cells and affect their structure and function.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
11
|
He J, Cui Z, Zhu Y. The role of caveolae in endothelial dysfunction. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:78-91. [PMID: 37724072 PMCID: PMC10388784 DOI: 10.1515/mr-2021-0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/03/2021] [Indexed: 09/20/2023]
Abstract
Caveolae, the specialized cell-surface plasma membrane invaginations which are abundant in endothelial cells, play critical roles in regulating various cellular processes, including cholesterol homeostasis, nitric oxide production, and signal transduction. Endothelial caveolae serve as a membrane platform for compartmentalization, modulation, and integration of signal events associated with endothelial nitric oxide synthase, ATP synthase β, and integrins, which are involved in the regulation of endothelial dysfunction and related cardiovascular diseases, such as atherosclerosis and hypertension. Furthermore, these dynamic microdomains on cell membrane are modulated by various extracellular stimuli, including cholesterol and flow shear stress. In this brief review, we summarize the critical roles of caveolae in the orchestration of endothelial function based on recent findings as well as our work over the past two decades.
Collapse
Affiliation(s)
- Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin300070, China
| | - Zhen Cui
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin300070, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin300070, China
| |
Collapse
|
12
|
Tan M, Ye J, Zhao M, Ke X, Huang K, Liu H. Recent developments in the regulation of cholesterol transport by natural molecules. Phytother Res 2021; 35:5623-5633. [PMID: 34327759 DOI: 10.1002/ptr.7198] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 11/10/2022]
Abstract
The dysregulation of cholesterol metabolism is a high-risk factor for non-alcoholic fatty liver disease (NAFLD), dyslipidemia, and atherosclerosis (AS). Cholesterol transport maintains whole-body cholesterol homeostasis. Low-density apolipoprotein receptor (LDLR) mediates cholesterol uptake in cells and plays an important role in the primary route of circulatory cholesterol clearance in liver cells. Caveolins 1 is an integral membrane protein and shuttle between the cytoplasm and cell membrane. Caveolins 1 not only plays a role in promoting cholesterol absorption in cells but also in the transport of cellular cholesterol efflux by interacting with the ATP-binding cassette transporter A1 (ABCA1) and scavenger receptor class B type I (SR-BI). These proteins, which are associated with reverse cholesterol transport (RCT), are potential therapeutic targets for NAFLD and AS. Many studies have indicated that natural products have lipid-lowering effects. Moreover, natural molecules, derived from natural products, have the potential to be developed into novel drugs. However, the mechanisms underlying the regulation of cholesterol transport by natural molecules have not yet been adequately investigated. In this review, we briefly describe the process of cholesterol transport and summarize the mechanisms by which molecules regulate cholesterol transport. This article provides an overview of recent studies and focuses on the potential therapeutic effects of natural molecules; however, further high-quality studies are needed to firmly establish the clinical efficacies of natural molecules.
Collapse
Affiliation(s)
- Meiao Tan
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, China.,First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jintong Ye
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Zhao
- Guangzhou Liwan District Traditional Chinese Medicine Hospital, Guangzhou, China
| | - Xuehong Ke
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Keer Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huabao Liu
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
13
|
Mattoso Miskulin Cardoso AP, Tavares Pereira M, Dos Santos Silva R, Medeiros de Carvalho Sousa LM, Giometti IC, Kowalewski MP, de Carvalho Papa P. Global transcriptome analysis implicates cholesterol availability in the regulation of canine cyclic luteal function. Gen Comp Endocrinol 2021; 307:113759. [PMID: 33771531 DOI: 10.1016/j.ygcen.2021.113759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/06/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022]
Abstract
Considering the key role of the corpus luteum in the regulation of the canine diestrus, the present study aimed to investigate changes in the luteal transcriptome of pseudopregnant dogs (n = 18) from days (D) 10, 20, 30, 40, 50 and 60 post-ovulation. After RNAsequencing was performed, data was analyzed by resorting to several informatic tools. A total of 3300 genes were differently expressed among all samples (FDR < 0.01). By comparing different time points, enriched biological processes as response to estradiol and lipids (D20 vs D10) and intracellular cholesterol transport (D40 vs D60) were observed. Moreover, LXR/RXR (liver X receptor- retinoid X receptor) signaling appeared as an overrepresented pathway in all comparisons. Thus, the expression of 19 genes involved in intracellular cholesterol availability was further evaluated; most were affected by time (P < 0.05). Adding to the deep transcriptomic analysis, presented data implies the importance of cholesterol regulation in luteal physiology of pseudopregnant dogs.
Collapse
Affiliation(s)
| | - Miguel Tavares Pereira
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Renata Dos Santos Silva
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Ines Cristina Giometti
- Faculty of Veterinary Medicine, University of Western São Paulo, Presidente Prudente, Brazil
| | | | - Paula de Carvalho Papa
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil; Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
14
|
Buwaneka P, Ralko A, Liu SL, Cho W. Evaluation of the available cholesterol concentration in the inner leaflet of the plasma membrane of mammalian cells. J Lipid Res 2021; 62:100084. [PMID: 33964305 PMCID: PMC8178126 DOI: 10.1016/j.jlr.2021.100084] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Cholesterol is an essential component of the mammalian plasma membrane involved in diverse cellular processes. Our recent quantitative imaging analysis using ratiometric cholesterol sensors showed that the available cholesterol concentration in the inner leaflet of the plasma membrane (IPM) is low in unstimulated cells and increased in a stimulus-specific manner to trigger cell signaling events. However, the transbilayer distribution of cholesterol in the plasma membrane of mammalian cells remains controversial. Here we report a systematic and rigorous evaluation of basal IPM cholesterol levels in a wide range of mammalian cells with different properties employing cholesterol sensors derived from the D4 domain of the Perfringolysin O toxin and a sterol-transfer protein, Osh4. Results consistently showed that, although basal IPM cholesterol levels vary significantly among cells, they remain significantly lower than cholesterol levels in the outer leaflets. We found that IPM cholesterol levels were particularly low in all tested primary cells. These results support the universality of the low basal IPM cholesterol concentration under physiological conditions. We also report here the presence of sequestered IPM cholesterol pools, which may become available to cytosolic proteins under certain physiological conditions. We hypothesize that these pools may partly account for the low basal level of available IPM cholesterol. In conclusion, we provide new experimental data that confirm the asymmetric transbilayer distribution of the plasma membrane cholesterol, which may contribute to regulation of various cellular signaling processes at the plasma membrane.
Collapse
Affiliation(s)
- Pawanthi Buwaneka
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Arthur Ralko
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Shu-Lin Liu
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Wonhwa Cho
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
15
|
Wang XL, Zhang W, Li Z, Han WQ, Wu HY, Wang QR, Liu XH, Xing K, Cheng G, Chang FJ. Vascular damage effect of circulating microparticles in patients with ACS is aggravated by type 2 diabetes. Mol Med Rep 2021; 23:474. [PMID: 33899122 PMCID: PMC8097757 DOI: 10.3892/mmr.2021.12113] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 11/03/2020] [Indexed: 01/18/2023] Open
Abstract
As a common factor of both type 2 diabetes mellitus (T2DM) and acute coronary syndrome (ACS), circulating microparticles (MPs) may provide a link between these two diseases. The present study compared the content and function of MPs from patients with ACS with or without T2DM. MPs from healthy subjects (n=20), patients with ACS (n=24), patients with T2DM (n=20) and patients with combined ACS and T2DM (n=24) were obtained. After incubating rat thoracic tissue with MPs, the effect of MPs on endothelial‑dependent vasodilatation, expression of caveolin‑1 and endothelial nitric oxide synthase (eNOS), phosphorylation of eNOS at the S1177 and T495 sites and its association with heat shock protein 90 (Hsp90), and the generation of NO and superoxide anion (O2˙‑) were determined. MP concentrations were higher in patients with T2DM and patients with ACS with or without T2DM than in healthy subjects. Moreover, MPs from patients with T2DM or ACS led to impairment in endothelial‑dependent vasodilatation, decreased expression of NO, as well as eNOS and its phosphorylation at Ser1177 and association with Hsp90, but increased eNOS phosphorylation at T495, caveolin‑1 expression and O2˙‑ generation. These effects were strengthened by MPs from patients with ACS combined with T2DM. T2DM not only increased MP content but also resulted in greater vascular impairment effects in ACS. These results may provide novel insight into the treatment of patients with ACS and T2DM.
Collapse
Affiliation(s)
- Xu-Lan Wang
- Department of Nursing, Xian'yang Vocational and Technical College, Fengxi New Town United Avenue, Xi'an, Shaanxi 712000, P.R. China
| | - Wei Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Zhe Li
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Wen-Qi Han
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Hao-Yu Wu
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Qun-Rang Wang
- Department of Cardiology, Affiliated Hospital of Shaanxi Traditional Chinese Medicine University, Xianyang, Shaanxi 712000, P.R. China
| | - Xin-Hong Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Kun Xing
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Gong Cheng
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| | - Feng-Jun Chang
- Department of Cardiology, Shaanxi Provincial People's Hospital and The Third Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
16
|
Yang Y, He X, Xia S, Liu F, Luo L. Porphyromonas gingivalis facilitated the foam cell formation via lysosomal integral membrane protein 2 (LIMP2). J Periodontal Res 2021; 56:265-274. [PMID: 33372271 DOI: 10.1111/jre.12812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The involvement of lysosomal integral membrane protein 2 (LIMP2) in cholesterol transport and formation of foam cells under the infection of Porphyromonas gingivalis (P. gingivalis) is yet to be elucidated. The current study verified the role and explored the mechanism of LIMP2 in promoting foam cell formation by P. gingivalis. BACKGROUND An association between periodontitis and atherosclerosis (AS) has been established. P. gingivalis is a key pathogen of periodontitis that promotes foam cell formation by regulating activities of CD36 scavenger receptors expressed on the macrophages. LIMP2, a member of CD36 superfamily, is involved in cholesterol efflux. However, whether LIMP2 is involved in the formation of foam cells promoted by P. gingivalis remains unclear. METHODS The formation of foam cells was examined by Oil Red O staining. The knockdown of limp2 was identified by qRT-PCR. The accumulation of cholesterol was monitored by Cholesterol Assay Kit. The location of P. gingivalis was visualized by confocal microscopy. Cathepsin L activity was monitored with Magic Red Cathepsin L Assay Kit. The key genes and pathways in P. gingivalis-infected macrophages were explored by RNA sequencing. The protein level was investigated by Western blotting. RESULTS Porphyromonas gingivalis increases foam cells formation and upregulates the expression of LIMP2 in foam cells. The knockdown of limp2 decreases the number of foam cells and increases cholesterol export, which is related to lysosomal functions. In addition, the interaction between LIMP2 and caveolin-1(CAV1) might contribute to this process, and NF-κB and JNK activity is required for increased expression of P. gingivalis-induced LIMP2. CONCLUSIONS This study suggested that LIMP2 is involved in the foam cells formation facilitated by P. gingivalis, which favors a close connection between periodontitis and atherosclerosis (AS).
Collapse
Affiliation(s)
- Yanan Yang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Department of Periodontics, School of Stomatology, Tongji University, Shanghai, China
| | - Xiaoli He
- Dental Diseases Prevention & Treatment Center of Jiading District, Shanghai, China
| | - Siying Xia
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Department of Periodontics, School of Stomatology, Tongji University, Shanghai, China
| | - Feng Liu
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology, Therapy Center for Obstructive Sleep Apnea, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lijun Luo
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Department of Periodontics, School of Stomatology, Tongji University, Shanghai, China
| |
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: 2.3] [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
- *Correspondence: Antonio Castrillo, ; Cristina M. Ramírez,
| | - 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
- *Correspondence: Antonio Castrillo, ; Cristina M. Ramírez,
| |
Collapse
|
18
|
Zhang H, Men K, Pan C, Gao Y, Li J, Lei S, Zhu G, Li R, Wei Y, Duan X. Treatment of Colon Cancer by Degradable rrPPC Nano-Conjugates Delivered STAT3 siRNA. Int J Nanomedicine 2020; 15:9875-9890. [PMID: 33324056 PMCID: PMC7732178 DOI: 10.2147/ijn.s277845] [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: 08/19/2020] [Accepted: 11/10/2020] [Indexed: 02/05/2023] Open
Abstract
Background Drugs that work based on the mechanism of RNA interference have shown strong potential in cancer gene therapy. Although significant progress has been made in small interfering RNA (siRNA) design and manufacturing, ideal delivery system remains a limitation for the development of siRNA-based drugs. Particularly, it is necessary to focus on parameters including delivery efficiency, stability, and safety when developing siRNA formulations for cancer therapy. Methods In this work, a novel degradable siRNA delivery system cRGD-R9-PEG-PEI-Cholesterol (rrPPC) was synthesized based on low molecular weight polyethyleneimine (PEI). Functional groups including cholesterol, cell penetrating peptides (CPPs), and poly(ethylene oxide) were introduced to PEI backbone to attain enhanced transfection efficiency and biocompatibility. Results The synthesized rrPPC was dispersed as nanoparticles in water with an average size of 195 nm and 41.9 mV in potential. rrPPC nanoparticles could efficiently deliver siRNA into C26 clone cancer cells and trigger caveolae-mediated pathway during transmembrane transportation. By loading the signal transducer and activator of transcription 3 (STAT3) targeting siRNA, rrPPC/STAT3 siRNA (rrPPC/siSTAT3) complex demonstrated strong anti-cancer effects in multiple colon cancer models following local delivery. In addition, intravenous (IV) injection of rrPPC/siSTAT3 complex efficiently suppressed lung metastasis tumor progression with ideal in vivo safety. Conclusion Our results provide evidence that rrPPC nanoparticles constitute a potential candidate vector for siRNA-based colon cancer gene therapy.
Collapse
Affiliation(s)
- Hongjia Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Congbin Pan
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Yan Gao
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Jingmei Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Guonian Zhu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Rui Li
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, People's Republic of China
| |
Collapse
|
19
|
Kurakin A, Bredesen DE. Alzheimer's disease as a systems network disorder: chronic stress/dyshomeostasis, innate immunity, and genetics. Aging (Albany NY) 2020; 12:17815-17844. [PMID: 32957083 PMCID: PMC7585078 DOI: 10.18632/aging.103883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/25/2020] [Indexed: 01/24/2023]
Abstract
Ineffective results of clinical trials of over 200 anti-Alzheimer's drug candidates, with a 99.6% attrition rate, suggest that the current paradigm of Alzheimer's disease (AD) may be incomplete, necessitating exploration of alternative and complementary frameworks.Using algorithms for hypothesis independent search and expert-assisted synthesis of heterogeneous data, we attempted to reconcile multimodal clinical profiles of early-stage AD patients and accumulated research data within a parsimonious framework. Results of our analysis suggest that Alzheimer's may not be a brain disease but a progressive system-level network disorder, which is driven by chronic network stress and dyshomeostasis. The latter can be caused by various endogenous and exogenous factors, such as chronic inflammatory conditions, infections, vascular dysfunction, head trauma, environmental toxicity, and immune disorders. Whether originating in the brain or on the periphery, chronic stress, toxicity, and inflammation are communicated to the central nervous system (CNS) via humoral and neural routes, preferentially targeting high-centrality regulatory nodes and circuits of the nervous system, and eventually manifesting as a neurodegenerative CNS disease.In this report, we outline an alternative perspective on AD as a systems network disorder and discuss biochemical and genetic evidence suggesting the central role of chronic tissue injury/dyshomeostasis, innate immune reactivity, and inflammation in the etiopathobiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Alexei Kurakin
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Dale E. Bredesen
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA,Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
20
|
Haas AV, Baudrand R, Easly RM, Murray GR, Touyz RM, Pojoga LH, Jeunemaitre X, Hopkins PN, Rosner B, Williams JS, Williams GH, Adler GK. Interplay Between Statins, Cav1 (Caveolin-1), and Aldosterone. Hypertension 2020; 76:962-967. [PMID: 32755411 PMCID: PMC7418929 DOI: 10.1161/hypertensionaha.120.14777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Statin use is associated with lower aldosterone levels. We hypothesized that caveolin-1 may be important for the uptake of statins into the adrenal gland and would affect statin’s aldosterone-lowering effects. The aim of this study was to test whether the caveolin-1 risk allele (rs926198) would affect aldosterone levels associated with statin use. The Hypertensive Pathotype database includes healthy and hypertensive individuals who have undergone assessment of adrenal hormones. Individuals were studied off antihypertensive medications but were maintained on statins if prescribed by their personal physician. Adrenal hormones were measured at baseline and after 1 hour of angiotensin II stimulation on both high- and low-sodium diets. A mixed-model repeated-measures analysis was employed with a priori selected covariates of age, sex, body mass index, and protocol (low versus high sodium, baseline versus angiotensin II stimulated aldosterone). A total of 250 individuals were included in the study; 31 individuals were taking statins (12.4%) and 219 were not. Among statin users, carrying a caveolin-1 risk allele resulted in a 25% (95% CI, 1–43.2) lower aldosterone level (P=0.04). However, among nonstatin users, carrying a caveolin-1 risk allele resulted in no significant effect on aldosterone levels (P=0.38). Additionally, the interaction between caveolin-1 risk allele and statin use on aldosterone levels was significant (P=0.03). These findings suggest caveolin-1 risk allele carrying individuals are likely to receive the most benefit from statin’s aldosterone-lowering properties; however, due to the observational nature of this study, these findings need further investigation.
Collapse
Affiliation(s)
- Andrea V Haas
- From the Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital (A.V.H., R.M.E., G.RM., L.H.P., J.S.W., G.H.W., G.K.A.), Harvard Medical School, Boston, MA
| | - Rene Baudrand
- Program for Adrenal Disorders and Endocrine Hypertension, Department of Endocrinology, CETREN, School of Medicine, Pontificia Universidad Catolica De Chile, Santiago, Chile (R.B.)
| | - Rebecca M Easly
- From the Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital (A.V.H., R.M.E., G.RM., L.H.P., J.S.W., G.H.W., G.K.A.), Harvard Medical School, Boston, MA
| | - Gillian R Murray
- From the Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital (A.V.H., R.M.E., G.RM., L.H.P., J.S.W., G.H.W., G.K.A.), Harvard Medical School, Boston, MA
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (R.M.T.)
| | - Luminita H Pojoga
- From the Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital (A.V.H., R.M.E., G.RM., L.H.P., J.S.W., G.H.W., G.K.A.), Harvard Medical School, Boston, MA
| | - Xavier Jeunemaitre
- University of Paris, Faculty of Health; INSERM, UMRS-970, F-75015 France (X.J.).,APHP, Department of Genetics, Hôpital Européen Georges Pompidou, F-75015 Paris, France (X.J.)
| | - Paul N Hopkins
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City (P.N.H.)
| | - Bernard Rosner
- Division of Network Medicine, Department of Medicine, Channing (B.R.), Harvard Medical School, Boston, MA
| | - Jonathan S Williams
- From the Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital (A.V.H., R.M.E., G.RM., L.H.P., J.S.W., G.H.W., G.K.A.), Harvard Medical School, Boston, MA
| | - Gordon H Williams
- From the Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital (A.V.H., R.M.E., G.RM., L.H.P., J.S.W., G.H.W., G.K.A.), Harvard Medical School, Boston, MA
| | - Gail K Adler
- From the Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital (A.V.H., R.M.E., G.RM., L.H.P., J.S.W., G.H.W., G.K.A.), Harvard Medical School, Boston, MA
| |
Collapse
|
21
|
Loss of Caveolin-1 Is Associated with a Decrease in Beta Cell Death in Mice on a High Fat Diet. Int J Mol Sci 2020; 21:ijms21155225. [PMID: 32718046 PMCID: PMC7432291 DOI: 10.3390/ijms21155225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022] Open
Abstract
Elevated free fatty acids (FFAs) impair beta cell function and reduce beta cell mass as a consequence of the lipotoxicity that occurs in type 2 diabetes (T2D). We previously reported that the membrane protein caveolin-1 (CAV1) sensitizes to palmitate-induced apoptosis in the beta pancreatic cell line MIN6. Thus, our hypothesis was that CAV1 knock-out (CAV1 KO) mice subjected to a high fat diet (HFD) should suffer less damage to beta cells than wild type (WT) mice. Here, we evaluated the in vivo response of beta cells in the pancreatic islets of 8-week-old C57Bl/6J CAV1 KO mice subjected to a control diet (CD, 14% kcal fat) or a HFD (60% kcal fat) for 12 weeks. We observed that CAV1 KO mice were resistant to weight gain when on HFD, although they had high serum cholesterol and FFA levels, impaired glucose tolerance and were insulin resistant. Some of these alterations were also observed in mice on CD. Interestingly, KO mice fed with HFD showed an adaptive response of the pancreatic beta cells and exhibited a significant decrease in beta cell apoptosis in their islets compared to WT mice. These in vivo results suggest that although the CAV1 KO mice are metabolically unhealthy, they adapt better to a HFD than WT mice. To shed light on the possible signaling pathway(s) involved, MIN6 murine beta cells expressing (MIN6 CAV) or not expressing (MIN6 Mock) CAV1 were incubated with the saturated fatty acid palmitate in the presence of mitogen-activated protein kinase inhibitors. Western blot analysis revealed that CAV1 enhanced palmitate-induced JNK, p38 and ERK phosphorylation in MIN6 CAV1 cells. Moreover, all the MAPK inhibitors partially restored MIN6 viability, but the effect was most notable with the ERK inhibitor. In conclusion, our results suggest that CAV1 KO mice adapted better to a HFD despite their altered metabolic state and that this may at least in part be due to reduced beta cell damage. Moreover, they indicate that the ability of CAV1 to increase sensitivity to FFAs may be mediated by MAPK and particularly ERK activation.
Collapse
|
22
|
Raz BD, Dimitry C, Andrea SS. The uptake mechanism and intracellular fate of Paraoxonase-1 in endothelial cells. Free Radic Biol Med 2020; 153:26-33. [PMID: 32244050 DOI: 10.1016/j.freeradbiomed.2020.03.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
Abstract
Paraoxonase-1 (PON1) is a high-density lipoprotein (HDL)-associated lactonase that plays a significant role in the anti-atherosclerotic activity of HDL. However, several studies have shown that PON1 localizes in cells, where it operates independently of HDL. Previously, we showed that PON1 localizes in endothelial cells (ECs), and impairs vasodilation mediated by the endothelium-derived hyperpolarizing factor (EDHF) 5,6-δ-DHTL. However, the internalization pathway of PON1 into ECs, and the intracellular fate of PON1 are unknown. Therefore, the present study aimed to elucidate the uptake mechanism, intracellular trafficking and the function of PON1 in ECs. We conducted a series of inhibition experiments of fluorescently labeled recombinant PON1 (rePON1) in ECs, followed by FACS analyses. We found that rePON1 binds the EC membrane via specific binding sites located in lipid-rafts/caveolae microdomains that are shared with HDL, and internalized through dynamin-dependent endocytosis. Qualitative assessments of the intracellular trafficking of rePON1, using confocal z-stack images, showed colocalization of the labeled rePON1 with early and late endosome/lysosome markers. Accordingly, a "pulse-chase" incubation of rePON1, followed by lactonase activity measurement in EC lysate, revealed that rePON1 retains its lactonase activity after binding to the cells. However, this activity decreases over time. Finally, induction of endothelial dysfunction with high glucose, angiotensin II, or palmitic acid increased rePON1 uptake by ECs. In conclusion, these results indicate that free PON1 interacts with ECs via binding sites located in lipid-rafts/caveolae, where it is enzymatically active and regulates endothelial functions. However, once internalized, PON1 is degraded. Additionally, alteration in endothelial function affects PON1 uptake by ECs.
Collapse
Affiliation(s)
- Ben-David Raz
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel
| | | | - Szuchman-Sapir Andrea
- Laboratory of Vascular Signaling Research, MIGAL-Galilee Research Institute, Ltd., Kiryat Shmona, Israel; Tel-Hai College, Upper Galilee, Israel.
| |
Collapse
|
23
|
Tian J, Popal MS, Huang R, Zhang M, Zhao X, Zhang M, Song X. Caveolin as a Novel Potential Therapeutic Target in Cardiac and Vascular Diseases: A Mini Review. Aging Dis 2020; 11:378-389. [PMID: 32257548 PMCID: PMC7069461 DOI: 10.14336/ad.2019.09603] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/03/2019] [Indexed: 12/27/2022] Open
Abstract
Caveolin, a structural protein of caveolae, play roles in the regulation of endothelial function, cellular lipid homeostasis, and cardiac function by affecting the activity and biogenesis of nitric oxide, and by modulating signal transduction pathways that mediate inflammatory responses and oxidative stress. In this review, we present the role of caveolin in cardiac and vascular diseases and the relevant signaling pathways involved. Furthermore, we discuss a novel therapeutic perspective comprising crosstalk between caveolin and autophagy.
Collapse
Affiliation(s)
- Jinfan Tian
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mohammad Sharif Popal
- 2 Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - RongChong Huang
- 3 Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100010, China
| | - Min Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xin Zhao
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingduo Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiantao Song
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
24
|
Zhu F, Huang J, Wang X, Li P, Yan Y, Zheng Y, He Y, Wu T, Ren Y, Wu R. The expression and significance of serum caveolin-1 in patients with Kawasaki disease. CHINESE J PHYSIOL 2020; 63:90-94. [PMID: 32341235 DOI: 10.4103/cjp.cjp_71_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
We investigated the expression of caveolin-1 (Cav-1) in Kawasaki disease (KD) and analyzed its relationship with coronary artery lesions (CALs). Cav-1 participated in the progression of CAL in KD. A total of 68 children with KD (23 with CALs), age matched with a fever control group (F, n = 28) and a normal control group (N, n = 24) were enrolled in this study. Cav-1 expression was detected using an enzyme-linked immunosorbent assay. The results are the following: (1) Compared with the F and N, Cav-1 expression was significantly increased in the children with KD (P < 0.05); there was no significant difference in Cav-1 between the F and N. (2) The serum level of Cav-1 was significantly higher in children with KD and CALs during the acute phase than in children with KD without CALs (P < 0.05). (3) Serum Cav-1 may be a biomarker that reflects CALs in children with KD based on a receiver operating characteristic (ROC) curve analysis. (4) Those children with KD who were given intravenous immunoglobulin (2 g/kg, 10-12 h) during the acute phase showed decreased expression of Cav-1 compared to the N. Conclusions are as follows: (1) The serum level of Cav-1 during the acute phase of KD increased significantly, while in KD patients with CALs the increase was even greater. (2) Based on our ROC curve analysis, Cav-1 may be a predictor of CALs in children with KD.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Child Healthcare, Wenzhou People's Hospital, Wenzhou, Zhejiang, China
| | - Jing Huang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuliang Wang
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ping Li
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yaoyao Yan
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunyun Zheng
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue'e He
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tingting Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue Ren
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rongzhou Wu
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Institute of Cardiovascular Development and Translational Medicine, The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
25
|
Inoh Y, Tsuchiya Y, Nakanishi Y, Yokawa S, Furuno T. Involvement of intracellular caveolin-1 distribution in the suppression of antigen-induced mast cell activation by cationic liposomes. Cell Biol Int 2020; 44:1068-1075. [PMID: 31889352 DOI: 10.1002/cbin.11297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/28/2019] [Indexed: 11/10/2022]
Abstract
Cationic liposomes are commonly used as vectors to effectively introduce foreign genes into target cells. In another function, we recently showed that cationic liposomes bound to the mast cell surface suppress the degranulation induced by the cross-linking of high-affinity immunoglobulin E receptor in a time- and dose-dependent manner. This suppression is mediated by the impairment of the sustained level of intracellular Ca2+ concentration ([Ca2+ ]i ) via the inhibition of store-operated Ca2+ entry. Further, we revealed that the mechanism underlying an impaired [Ca2+ ]i increase is the inhibition of the activation of the phosphatidylinositol 3-kinase (PI3K)-Akt pathway. Yet, how cationic liposomes inhibit the PI3K-Akt pathway is still unclear. Here, we focused on caveolin-1, a major component of caveolae, which is reported to be involved in the activation of the PI3K-Akt pathway in various cell lines. In this study, we showed that caveolin-1 translocated from the cytoplasm to the plasma membrane after the activation of mast cells and colocalized with the p85 subunit of PI3K, which seemed to be essential for PI3K activity. Meanwhile, cationic liposomes suppressed the translocation of caveolin-1 to the plasma membrane and the colocalization of caveolin-1 with PI3K p85 also at the plasma membrane. This finding provides new information for the development of therapies using cationic liposomes against allergies.
Collapse
Affiliation(s)
- Yoshikazu Inoh
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Yuuki Tsuchiya
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Yokiko Nakanishi
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Satoru Yokawa
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Tadahide Furuno
- School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| |
Collapse
|
26
|
Xu Y, Du X, Turner N, Brown AJ, Yang H. Enhanced acyl-CoA:cholesterol acyltransferase activity increases cholesterol levels on the lipid droplet surface and impairs adipocyte function. J Biol Chem 2019; 294:19306-19321. [PMID: 31727739 DOI: 10.1074/jbc.ra119.011160] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/11/2019] [Indexed: 01/21/2023] Open
Abstract
Cholesterol plays essential structural and signaling roles in mammalian cells, but too much cholesterol can cause cytotoxicity. Acyl-CoA:cholesterol acyltransferases 1 and 2 (ACAT1/2) convert cholesterol into its storage form, cholesteryl esters, regulating a key step in cellular cholesterol homeostasis. Adipose tissue can store >50% of whole-body cholesterol. Interestingly, however, almost no ACAT activity is present in adipose tissue, and most adipose cholesterol is stored in its free form. We therefore hypothesized that increased cholesterol esterification may have detrimental effects on adipose tissue function. Here, using several approaches, including protein overexpression, quantitative RT-PCR, immunofluorescence, and various biochemical assays, we found that ACAT1 expression is significantly increased in the adipose tissue of the ob/ob mice. We further demonstrated that ACAT1/2 overexpression partially inhibited the differentiation of 3T3-L1 preadipocytes. In mature adipocytes, increased ACAT activity reduced the size of lipid droplets (LDs) and inhibited lipolysis and insulin signaling. Paradoxically, the amount of free cholesterol increased on the surface of LDs in ACAT1/2-overexpressing adipocytes, accompanied by increased LD localization of caveolin-1. Moreover, cholesterol depletion in adipocytes by treating the cells with cholesterol-deficient media or β-cyclodextrins induced changes in cholesterol distribution that were similar to those caused by ACAT1/2 overexpression. Our results suggest that ACAT1/2 overexpression increases the level of free cholesterol on the LD surface, thereby impeding adipocyte function. These findings provide detailed insights into the role of free cholesterol in LD and adipocyte function and suggest that ACAT inhibitors have potential utility for managing disorders associated with extreme obesity.
Collapse
Affiliation(s)
- Yanqing Xu
- School of Biotechnology and Biomolecular Sciences, the University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, the University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Nigel Turner
- School of Medical Sciences, the University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, the University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, the University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
27
|
Farràs M, Arranz S, Carrión S, Subirana I, Muñoz-Aguayo D, Blanchart G, Kool M, Solà R, Motilva MJ, Escolà-Gil JC, Rubió L, Fernández-Castillejo S, Pedret A, Estruch R, Covas MI, Fitó M, Hernáez Á, Castañer O. A Functional Virgin Olive Oil Enriched with Olive Oil and Thyme Phenolic Compounds Improves the Expression of Cholesterol Efflux-Related Genes: A Randomized, Crossover, Controlled Trial. Nutrients 2019; 11:nu11081732. [PMID: 31357534 PMCID: PMC6723782 DOI: 10.3390/nu11081732] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022] Open
Abstract
The consumption of antioxidant-rich foods such as virgin olive oil (VOO) promotes high-density lipoprotein (HDL) anti-atherogenic capacities. Intake of functional VOOs (enriched with olive/thyme phenolic compounds (PCs)) also improves HDL functions, but the gene expression changes behind these benefits are not fully understood. Our aim was to determine whether these functional VOOs could enhance the expression of cholesterol efflux-related genes. In a randomized, double-blind, crossover, controlled trial, 22 hypercholesterolemic subjects ingested for three weeks 25 mL/day of: (1) a functional VOO enriched with olive oil PCs (500 mg/kg); (2) a functional VOO enriched with olive oil (250 mg/kg) and thyme PCs (250 mg/kg; FVOOT), and; (3) a natural VOO (olive oil PCs: 80 mg/kg, control intervention). We assessed whether these interventions improved the expression of cholesterol efflux-related genes in peripheral blood mononuclear cells by quantitative reverse-transcription polymerase chain reactions. The FVOOT intervention upregulated the expression of CYP27A1 (p = 0.041 and p = 0.053, versus baseline and the control intervention, respectively), CAV1 (p = 0.070, versus the control intervention), and LXRβ, RXRα, and PPARβ/δ (p = 0.005, p = 0.005, and p = 0.038, respectively, relative to the baseline). The consumption of a functional VOO enriched with olive oil and thyme PCs enhanced the expression of key cholesterol efflux regulators, such as CYP27A1 and nuclear receptor-related genes.
Collapse
Affiliation(s)
- Marta Farràs
- Molecular Bases of Cardiovascular Risk Group, IIB-Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Sara Arranz
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Sílvia Carrión
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Isaac Subirana
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Cardiovascular Genetics and Epidemiology Research Group, IMIM, 08003 Barcelona, Spain
| | - Daniel Muñoz-Aguayo
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
| | - Gemma Blanchart
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Marjon Kool
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Rosa Solà
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - María José Motilva
- Instituto de Ciencias de la Vid y el Vino, CSIC-Universidad de la Rioja, 26007 Logroño, Spain
| | - Joan Carles Escolà-Gil
- Molecular Bases of Cardiovascular Risk Group, IIB-Sant Pau, 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Laura Rubió
- Food Technology Department, Agrotecnio Center, University of Lleida, 25198 Lleida, Spain
| | - Sara Fernández-Castillejo
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili, 43204 Reus, Spain
| | - Anna Pedret
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili, 43204 Reus, Spain
| | - Ramón Estruch
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
- Internal Medicine Service, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
- Cardiovascular Risk, Nutrition and Aging Research Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - María Isabel Covas
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
- NUPROAS Handelsbolag, Nackă, Sweden
| | - Montserrat Fitó
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
| | - Álvaro Hernáez
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain.
- Cardiovascular Risk, Nutrition and Aging Research Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.
| | - Olga Castañer
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain.
| |
Collapse
|
28
|
An endosomal LAPF is required for macrophage endocytosis and elimination of bacteria. Proc Natl Acad Sci U S A 2019; 116:12958-12963. [PMID: 31189603 DOI: 10.1073/pnas.1903896116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Macrophages can internalize the invading pathogens by raft/caveolae and/or clathrin-dependent endocytosis and elicit an immune response against infection. However, the molecular mechanism for macrophage endocytosis remains elusive. Here we report that LAPF (lysosome-associated and apoptosis-inducing protein containing PH and FYVE domains) is required for caveolae-mediated endocytosis. Lapf -deficient macrophages have impaired capacity to endocytose and eliminate bacteria. Macrophage-specific Lapf-deficient mice are more susceptible to Escherichia coli (E. coli) infection with higher bacterial loads. Moreover, Lapf deficiency impairs TLR4 endocytosis, resulting in attenuated production of TLR-triggered proinflammatory cytokines. LAPF is localized to early endosomes and interacts with caveolin-1. Phosphorylation of LAPF by the tyrosine kinase Src is required for LAPF-Src-Caveolin complex formation and endocytosis and elimination of bacteria. Collectively, our work demonstrates that LAPF is critical for endocytosis of bacteria and induction of inflammatory responses, suggesting that LAPF and Src could be potential targets for the control of infectious diseases.
Collapse
|
29
|
Erchen Decoction Ameliorates Lipid Metabolism by the Regulation of the Protein CAV-1 and the Receptors VLDLR, LDLR, ABCA1, and SRB1 in a High-Fat Diet Rat Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5309490. [PMID: 30402126 PMCID: PMC6196931 DOI: 10.1155/2018/5309490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/07/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022]
Abstract
Lipid metabolism disorder is a common metabolic disorder characterized by abnormal lipid levels in blood. Erchen decoction (ECD) is a traditional Chinese medicine prescription, which is used for the treatment of diseases caused by retention of phlegm dampness. It has been reported to ameliorate the disorder of lipid metabolism. The aim of the present study was to investigate the effects and underlying mechanisms of ECD in lipid metabolism disorder induced by a high-fat diet (HFD) in rats. ECD (4.35g/kg/d) and atorvastatin (10mg/kg/d, positive control) were orally administered to HFD-fed rats for four weeks. The parameters, food, water consumption, body weight, body length, liver, and visceral fat weight and the content of serum lipids and lipid transporters were assessed. The effects of ECD on the mRNA and protein expression levels of lipid transport factors were measured by real-time PCR and western blotting. The present study demonstrated that ECD improved the disorders of serum lipid and lipid transporters in HFD-fed rats, TG (0.70±0.08 mmol/L, p<0.01), LDL-C (1.50±0.19 mmol/L, p<0.01), LDL (1.38±0.21 mmol/L, p<0.05), and oxLDL (1.77±0.39 ng/mL, p<0.05) were downregulated, while HDL-C (0.87±0.13 mmol/L, p<0.01), FFA (0.62±0.13 mmol/L, p<0.05), HDL (38.8±4.0 mg/dL, p<0.05), and CETP (903.6±120.0 ng/mL, p<0.05) were upregulated. But ECD obviously had no effects on the indices food/water/energy intake, body/tissue (liver and fat) weight, and BMI (p>0.05). Concomitantly, ECD reversed the abnormal expressions of those lipid transport factors in the liver and visceral fat.
Collapse
|
30
|
Aguirre-Portolés C, Feliu J, Reglero G, Ramírez de Molina A. ABCA1 overexpression worsens colorectal cancer prognosis by facilitating tumour growth and caveolin-1-dependent invasiveness, and these effects can be ameliorated using the BET inhibitor apabetalone. Mol Oncol 2018; 12:1735-1752. [PMID: 30098223 PMCID: PMC6166002 DOI: 10.1002/1878-0261.12367] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/26/2018] [Accepted: 07/29/2018] [Indexed: 02/05/2023] Open
Abstract
At the time of diagnosis, 20% of patients with colorectal cancer present metastasis. Among individuals with primary lesions, 50% of them will develop distant tumours with time. Therefore, early diagnosis and prediction of aggressiveness is crucial for therapy design and disease prognosis. Tumoral cells must undergo significant changes in energy metabolism to meet increased structural and energetic demands for cell proliferation, and metabolic alterations are considered to be a hallmark of cancer. Here, we present the ATP‐binding cassette transporter (ABCA1), a regulator of cholesterol transport, as a new marker for invasion and colorectal cancer survival. ABCA1 is significantly overexpressed in patients at advanced stages of colorectal cancer, and its overexpression confers proliferative advantages together with caveolin‐1 dependent‐increased migratory and invasive capacities. Thus, intracellular cholesterol imbalances mediated by ABCA1 overexpression may contribute to primary tumour growth and dissemination to distant locations. Furthermore, we demonstrate here that increased levels of apolipoprotein A1 (APOA1), a protein involved in cholesterol efflux and high‐density lipoprotein constitution, in the extracellular compartment modulates expression of ABCA1 by regulating COX‐2, and compensate for ABCA1‐dependent excessive export of cholesterol. APOA1 emerges as a new therapeutic option to inhibit the promotion of colorectal cancer to metastasis by modulating intracellular cholesterol metabolism. Furthermore, we propose apabetalone, an orally available small molecule that is currently being evaluated in clinical trials for the treatment of atherosclerosis, as a new putative therapeutic option to prevent colorectal cancer progression by increasing APOA1 expression and regulating reverse transport of cholesterol.
Collapse
Affiliation(s)
| | - Jaime Feliu
- Medical Oncology, La Paz University Hospital (IdiPAZ), CIBERONC, cátedra UAM-AMGEN, Madrid, Spain
| | - Guillermo Reglero
- Molecular Oncology, IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | | |
Collapse
|
31
|
Wang J, Bai Y, Zhao X, Ru J, Kang N, Tian T, Tang L, An Y, Li P. oxLDL-mediated cellular senescence is associated with increased NADPH oxidase p47phox recruitment to caveolae. Biosci Rep 2018; 38:BSR20180283. [PMID: 29695496 PMCID: PMC5997791 DOI: 10.1042/bsr20180283] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/05/2018] [Accepted: 04/24/2018] [Indexed: 01/26/2023] Open
Abstract
Atherosclerosis develops as a consequence of inflammation and cell senescence. In critical factors involved in the atherosclerotic changes, reactive oxygen species (ROS) generation is considered a leading cause. While NADPH oxidases, particularly NOX2, are the main sources of ROS, how they are regulated in the disease is incompletely understood. In addition, how caveolae, the membrane structure implicated in oxLDL deposition under vascular endothelia, is involved in the oxLDL-mediated ROS production remains mostly elusive. We report here that macrophages exposed to oxLDL up-regulate its caveolin-1 expression, and the latter in turn up-regulates NOX2 p47phox level. This combination effect results in increased cellular senescence. Interestingly, oxLDL treatment causes the p47phox residing in the cytosol to translocate to the caveolae. Immunoprecipitation assays confirms that cavelin-1 is in high degree association with p47phox. These results suggest caveolin-1 may serve as the membrane target for p47phox and as a switch for ROS production following oxLDL exposure. Our results reveal a previously unknown molecular event in oxLDL-mediated cellular ageing, and may provide a target for clinical intervention for atherosclerosis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Yuzhi Bai
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Xia Zhao
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Jing Ru
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Ning Kang
- Institute for Immunology, Department of Basic Medical Sciences, School of Medicine, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tian Tian
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Liying Tang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Yun An
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| | - Pei Li
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China
| |
Collapse
|
32
|
Zeng Z, Cao B, Guo X, Li W, Li S, Chen J, Zhou W, Zheng C, Wei Y. Apolipoprotein B-100 peptide 210 antibody inhibits atherosclerosis by regulation of macrophages that phagocytize oxidized lipid. Am J Transl Res 2018; 10:1817-1828. [PMID: 30018722 PMCID: PMC6038070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Immunization with peptides derived from apolipoprotein B-100 (ApoB-100) has been shown to ameliorate atherosclerosis in apolipoprotein E knockout (ApoE-/-) mice. However, the exact mechanism underlying the therapeutic effects remains elusive. To shed light on this mechanism, we immunized ApoE-/- mice that were fed a Western diet with either malondialdehyde-modified ApoB-100 peptide 210 (P210) emulsified in Freund's adjuvant or anti-malondialdehyde-modified P210 antibody (P210-Ab). Mice immunized with Freund's adjuvant or bovine serum albumin served as controls. Macrophages were incubated in vitro with oxidized low-density lipoprotein (ox-LDL) or ox-LDL plus P210-Ab. Our results show that P210-Ab promoted cholesterol efflux, inhibited lipid accumulation in vitro, and reduced plasma levels of high-sensitivity C-reactive protein (hsCRP), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6). Furthermore, dramatically increased the expression of Fc receptors (FcR) on peripheral blood mononuclear macrophages, suggesting that the mechanism of phagocytosis of ox-LDL by mononuclear macrophages may rely more on FcR than the cluster of differentiation 36 (CD36) scavenger receptor with P210-Ab. Both in vitro and in vivo, P210-Ab triggered the promoter of ATP-binding cassette transporter A1 (ABCA1) to increase peroxisome proliferator-activated receptor alpha (α) activity and inhibit the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. In addition, P210-Ab significantly attenuated macrophage infiltration and markedly improved the stability of atheromatous plaque. In conclusion, the anti-atherosclerotic effect of P210-Ab is related to its preferential inhibition of inflammation and reversion of cholesterol transportation by altering the pathway by which macrophages phagocytize ox-LDL.
Collapse
Affiliation(s)
- Zhuanglin Zeng
- Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of The Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Bingxin Cao
- Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of The Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Xiaopeng Guo
- Department of Interventional Radiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Weijuan Li
- Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of The Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Songhai Li
- Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of The Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Juan Chen
- Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of The Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Wenping Zhou
- Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of The Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Chuansheng Zheng
- Department of Interventional Radiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| | - Yumiao Wei
- Laboratory of Cardiovascular Immunology, Key Laboratory of Molecular Targeted Therapies of The Ministry of Education, Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
| |
Collapse
|
33
|
Haczeyni F, Bell-Anderson KS, Farrell GC. Causes and mechanisms of adipocyte enlargement and adipose expansion. Obes Rev 2018; 19:406-420. [PMID: 29243339 DOI: 10.1111/obr.12646] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/28/2017] [Accepted: 10/23/2017] [Indexed: 02/06/2023]
Abstract
Adipose tissue plays a significant role in whole body energy homeostasis. Obesity-associated diabetes, fatty liver and metabolic syndrome are closely linked to adipose stress and dysfunction. Genetic predisposition, overeating and physical inactivity influence the expansion of adipose tissues. Under conditions of constant energy surplus, adipocytes become hypertrophic and adipose tissues undergo hyperplasia so as to increase their lipid storage capacity, thereby keeping circulating blood glucose and fatty acids below toxic levels. Nonetheless, adipocytes have a saturation point where they lose capacity to store more lipids. At this stage, when adipocytes are fully lipid-engorged, they express stress signals. Adipose depots (particularly visceral compartments) from obese individuals with a severe metabolic phenotype are characterized by the high proportion of hypertrophic adipocytes. This review focuses on the mechanisms of adipocyte enlargement in relation to adipose fatty acid and cholesterol metabolism, and considers how this may be related to adipose dysfunction.
Collapse
Affiliation(s)
- F Haczeyni
- Liver Research Group, Australian National University Medical School at The Canberra Hospital, Canberra, ACT, Australia
| | - K S Bell-Anderson
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - G C Farrell
- Liver Research Group, Australian National University Medical School at The Canberra Hospital, Canberra, ACT, Australia
| |
Collapse
|
34
|
Hsu CP, Lin CH, Kuo CY. Endothelial-cell inflammation and damage by reactive oxygen species are prevented by propofol via ABCA1-mediated cholesterol efflux. Int J Med Sci 2018; 15:978-985. [PMID: 30013438 PMCID: PMC6036153 DOI: 10.7150/ijms.24659] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 05/27/2018] [Indexed: 01/06/2023] Open
Abstract
Background: Cholesterol efflux efficiency, reactive oxygen species, and inflammation are closely related to cardiovascular diseases. Our aim was to investigate the effect of propofol on cholesterol-loaded rat aortic endothelial cells after high-density lipoprotein treatment in vitro. Methods and Results: The results showed that propofol promoted cholesterol efflux and ameliorated inflammation and reactive oxygen species overproduction according to the analysis of p65 nuclear translocation and a 2',7'-dichlorofluorescin diacetate assay, respectively. Conclusions: These results provide a possible explanation for the anti-inflammatory, antioxidant, and cholesterol efflux-promoting effects of propofol on rat aortic endothelial cells after incubation with high-density lipoprotein.
Collapse
Affiliation(s)
- Chih-Peng Hsu
- Department of Cardiology, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Chih-Hung Lin
- Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Chan-Yen Kuo
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Chungli, Taiwan.,Department of Ophthalmology, Hsin Sheng Junior College of Medical Care and Management, Longtan, Taiwan
| |
Collapse
|
35
|
Codrici E, Albulescu L, Popescu ID, Mihai S, Enciu AM, Albulescu R, Tanase C, Hinescu ME. Caveolin-1-Knockout Mouse as a Model of Inflammatory Diseases. J Immunol Res 2018; 2018:2498576. [PMID: 30246033 PMCID: PMC6136523 DOI: 10.1155/2018/2498576] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/05/2018] [Indexed: 02/07/2023] Open
Abstract
Caveolin-1 (CAV1) is the scaffold protein of caveolae, which are minute invaginations of the cell membrane that are involved in endocytosis, cell signaling, and endothelial-mediated inflammation. CAV1 has also been reported to have a dual role as either a tumor suppressor or tumor promoter, depending on the type of cancer. Inflammation is an important player in tumor progression, but the role of caveolin-1 in generating an inflammatory milieu remains poorly characterized. We used a caveolin-1-knockout (CAV1-/-) mouse model to assess the inflammatory status via the quantification of the pro- and anti-inflammatory cytokine levels, as well as the ability of circulating lymphocytes to respond to nonspecific stimuli by producing cytokines. Here, we report that the CAV1-/- mice were characterized by a low-grade systemic proinflammatory status, with a moderate increase in the IL-6, TNF-α, and IL-12p70 levels. CAV1-/- circulating lymphocytes were more prone to cytokine production upon nonspecific stimulation than the wild-type lymphocytes. These results show that CAV1 involvement in cell homeostasis is more complex than previously revealed, as it plays a role in the inflammatory process. These findings indicate that the CAV1-/- mouse model could prove to be a useful tool for inflammation-related studies.
Collapse
Affiliation(s)
- Elena Codrici
- 1Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Lucian Albulescu
- 1Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | | | - Simona Mihai
- 1Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Ana-Maria Enciu
- 1Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- 2Carol Davila University of Medicine and Pharmacy, 050047 Bucharest, Romania
| | - Radu Albulescu
- 1Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- 3National Institute for Chemical Pharmaceutical R&D, Bucharest, Romania
| | - Cristiana Tanase
- 1Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- 4Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Mihail E. Hinescu
- 1Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- 2Carol Davila University of Medicine and Pharmacy, 050047 Bucharest, Romania
| |
Collapse
|
36
|
Russell J, Du Toit EF, Peart JN, Patel HH, Headrick JP. Myocyte membrane and microdomain modifications in diabetes: determinants of ischemic tolerance and cardioprotection. Cardiovasc Diabetol 2017; 16:155. [PMID: 29202762 PMCID: PMC5716308 DOI: 10.1186/s12933-017-0638-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease, predominantly ischemic heart disease (IHD), is the leading cause of death in diabetes mellitus (DM). In addition to eliciting cardiomyopathy, DM induces a ‘wicked triumvirate’: (i) increasing the risk and incidence of IHD and myocardial ischemia; (ii) decreasing myocardial tolerance to ischemia–reperfusion (I–R) injury; and (iii) inhibiting or eliminating responses to cardioprotective stimuli. Changes in ischemic tolerance and cardioprotective signaling may contribute to substantially higher mortality and morbidity following ischemic insult in DM patients. Among the diverse mechanisms implicated in diabetic impairment of ischemic tolerance and cardioprotection, changes in sarcolemmal makeup may play an overarching role and are considered in detail in the current review. Observations predominantly in animal models reveal DM-dependent changes in membrane lipid composition (cholesterol and triglyceride accumulation, fatty acid saturation vs. reduced desaturation, phospholipid remodeling) that contribute to modulation of caveolar domains, gap junctions and T-tubules. These modifications influence sarcolemmal biophysical properties, receptor and phospholipid signaling, ion channel and transporter functions, contributing to contractile and electrophysiological dysfunction, cardiomyopathy, ischemic intolerance and suppression of protective signaling. A better understanding of these sarcolemmal abnormalities in types I and II DM (T1DM, T2DM) can inform approaches to limiting cardiomyopathy, associated IHD and their consequences. Key knowledge gaps include details of sarcolemmal changes in models of T2DM, temporal patterns of lipid, microdomain and T-tubule changes during disease development, and the precise impacts of these diverse sarcolemmal modifications. Importantly, exercise, dietary, pharmacological and gene approaches have potential for improving sarcolemmal makeup, and thus myocyte function and stress-resistance in this ubiquitous metabolic disorder.
Collapse
Affiliation(s)
- Jake Russell
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Eugene F Du Toit
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, San Diego, USA
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia. .,School of Medical Science, Griffith University, Southport, QLD, 4217, Australia.
| |
Collapse
|
37
|
Li J, Yu C, Wang R, Xu J, Chi Y, Qin J, Liu Q. The ω-carboxyl group of 7-ketocholesteryl-9-carboxynonanoate mediates the binding of oxLDL to CD36 receptor and enhances caveolin-1 expression in macrophages. Int J Biochem Cell Biol 2017; 90:121-135. [PMID: 28789920 DOI: 10.1016/j.biocel.2017.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 07/24/2017] [Accepted: 07/31/2017] [Indexed: 01/06/2023]
Abstract
CD36 signal transduction modulates the uptake of oxidized low-density lipoprotein (oxLDL) and foam cell formation. We previously observed that 7-ketocholesteryl-9-carboxynonanoate (oxLig-1), the lipid moiety of oxLDL, activates the CD36-Src-JNK/ERK1/2 signalling pathway. In this study, we assessed the role of the ω-carboxyl group in the binding of oxLig-1 to CD36 and investigated whether the binding of the ω-carboxyl group to CD36 triggers CD36-mediated signalling, thereby resulting in the upregulation of caveolin-1 expression. Our results showed that oxLig-1 bound to CD36 and that the ω-carboxyl group was critical for this binding. Furthermore, immunoprecipitation and Western blot analyses showed that interaction between the ω-carboxyl group of oxLig-1 and CD36 triggered intracellular Src-JNK/ERK1/2 signal transduction. Moreover, the binding of the ω-carboxyl group to CD36 induced caveolin-1 expression and translocation to the membrane in macrophages. Additionally, inhibitors of Src, JNK and ERK and siRNA targeting CD36 and NF-κB significantly suppressed the enhanced caveolin-1 expression induced by oxLig-1. In conclusion, these observations suggest that oxLig-1 is a critical epitope of oxLDL that mediates the binding of oxLDL to CD36 and activates downstream Src-JNK/ERK1/2-NF-κB signal transduction, resulting in upregulation of caveolin-1 expression in macrophages.
Collapse
Affiliation(s)
- Jingda Li
- Key Laboratory of Carbohydrate and Lipid Metabolism Research, College of Life Science and Technology, Dalian University, 10-Xuefu Avenue, Dalian Economical and Technological Development Zone, Liaoning 116622, China; School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, Liaoning, China
| | - Chengjie Yu
- Key Laboratory of Carbohydrate and Lipid Metabolism Research, College of Life Science and Technology, Dalian University, 10-Xuefu Avenue, Dalian Economical and Technological Development Zone, Liaoning 116622, China
| | - Renjun Wang
- Key Laboratory of Carbohydrate and Lipid Metabolism Research, College of Life Science and Technology, Dalian University, 10-Xuefu Avenue, Dalian Economical and Technological Development Zone, Liaoning 116622, China
| | - Jianrong Xu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan Chi
- Key Laboratory of Carbohydrate and Lipid Metabolism Research, College of Life Science and Technology, Dalian University, 10-Xuefu Avenue, Dalian Economical and Technological Development Zone, Liaoning 116622, China
| | - Jianzhong Qin
- Key Laboratory of Carbohydrate and Lipid Metabolism Research, College of Life Science and Technology, Dalian University, 10-Xuefu Avenue, Dalian Economical and Technological Development Zone, Liaoning 116622, China
| | - Qingping Liu
- Key Laboratory of Carbohydrate and Lipid Metabolism Research, College of Life Science and Technology, Dalian University, 10-Xuefu Avenue, Dalian Economical and Technological Development Zone, Liaoning 116622, China.
| |
Collapse
|
38
|
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: 2.4] [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
|
39
|
Shyu HY, Chen MH, Hsieh YH, Shieh JC, Yen LR, Wang HW, Cheng CW. Association of eNOS and Cav-1 gene polymorphisms with susceptibility risk of large artery atherosclerotic stroke. PLoS One 2017; 12:e0174110. [PMID: 28346478 PMCID: PMC5367681 DOI: 10.1371/journal.pone.0174110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/03/2017] [Indexed: 01/25/2023] Open
Abstract
Endothelial nitric oxide synthase (eNOS) is localized in caveole and has important effects on caveolar coordination through its interaction with caveolin-1 (Cav-1), which supports normal functioning of vascular endothelial cells. However, the relationship between genotypic polymorphisms of e-NOS and Cav-1 genes and ischemic stroke (IS) remains lesser reported. This hospital-based case-control study aimed to determine the genetic polymorphisms of the eNOS (Glu298Asp) and Cav-1 (G14713A and T29107A) genes in association with susceptibility risk in patients who had suffered from a large artery atherosclerotic (LAA) stroke. Genotyping determination for these variant alleles was performed using the TaqMan assay. The distributions of observed allelic and genotypic frequencies for the polymorphisms were in Hardy-Weinberg equilibrium in healthy controls. The risk for an LAA stroke in the Asp298 variant was 1.72 (95% CI = 1.09–2.75) versus Glu298 of the eNOS. In the GA/AA (rs3807987) variant, it was 1.79 (95% CI = 1.16–2.74) versus GG and in TA/AA (rs7804372) was 1.61 (95% CI = 1.06–2.43) versus TT of the Cav-1, respectively. A tendency toward an increased LAA stroke risk was significant in carriers with the eNOS Glu298Asp variant in conjunction with the G14713 A and T29107A polymorphisms of the Cav-1 (aOR = 2.03, P-trend = 0.002). A synergistic effect between eNOS and Cav-1 polymorphisms on IS risk elevation was significantly influenced by alcohol drinking, heavy cigarette smoking (P-trend<0.01), and hypercholesterolemia (P-trend < 0.001). In conclusion, genotypic polymorphisms of the eNOS Glu298Asp and Cav-1 14713A/29107A polymorphisms are associated with the elevated risk of LAA stroke among Han Chinese in Taiwan.
Collapse
Affiliation(s)
- Hann-Yeh Shyu
- Section of Neurology, Department of Internal Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
- Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hua Chen
- Section of Neurology, Department of Internal Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Jia-Ching Shieh
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Ling-Rong Yen
- Section of Neurology, Department of Internal Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
| | - Hsiao-Wei Wang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail:
| |
Collapse
|