1
|
Wang X, Shi SR, Sun MM, Zhang XY, Zhang XH, Song SL, Yin F, Guo SD. Mechanisms of action of Fucus vesiculosus-derived fucoidan on improving dyslipidemia in New Zealand rabbits fed a high-fat diet. Int J Biol Macromol 2025; 314:144148. [PMID: 40368205 DOI: 10.1016/j.ijbiomac.2025.144148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/30/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Dyslipidemia is a major contributor to various diseases, including atherosclerotic cardiovascular disease and obesity. Treatment strategies for dyslipidemia continue to evolve as our understanding of this metabolic disorder and potential therapeutic candidates advance. Notably, fucoidan demonstrates promising effects in ameliorating dyslipidemia in rodents, although their lipid metabolism differs significantly from humans. This study, investigates the lipid-regulatory effects of Fucus vesiculosus-derived fucoidan (FvF) and elucidates the underlying mechanisms of action using New Zealand rabbits fed a high-fat diet, whose lipid profiles closely resemble those of patients with dyslipidemia. The results demonstrate that FvF intervention ameliorates dyslipidemia and lipid deposition in a dose-dependent manner. Mechanistically, FvF intervention modulates the expression levels of multiple molecules involved in lipid transport, fatty acid synthesis and beta-oxidation, and redox balance, as revealed by quantitative reverse transcription polymerase chain reaction, western blotting, and proteomic analysis. This study is the first to report that FvF, consisting of alternating [→4)-α-L-Fucp(1 → 3)-α-L-Fucp(1→] glycosyls ameliorates dyslipidemia by directly modulating lipid metabolism and indirectly attenuating oxidative stress. These findings suggest that FvF holds significant potential as a candidate for the treatment of lipid disorder-related diseases.
Collapse
Affiliation(s)
- Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Min-Min Sun
- School of Stomatology, Shandong Second Medical University, Weifang 261053, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Xu-Hang Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shi-Lin Song
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Fan Yin
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
2
|
Meissner A, Liefke J, Matthes F, Morsing E, Ley D, Hedström E. Plasma biomarkers of inflammation associate with blood pressure and arterial stiffness in adolescents after very preterm birth. Cardiovasc Res 2024; 120:1992-1995. [PMID: 39196714 PMCID: PMC11646097 DOI: 10.1093/cvr/cvae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/01/2024] [Accepted: 07/05/2024] [Indexed: 08/30/2024] Open
Affiliation(s)
- Anja Meissner
- Department of Experimental Medical Science, Lund University, Klinikgatan 32, SE-22184 Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Klinikgatan 32, SE-22184 Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Universitätsstr. 2, DE-86159 Augsburg, Germany
| | - Jonas Liefke
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Clinical Physiology, Skåne University Hospital, Lund, Sweden
| | - Frank Matthes
- Department of Experimental Medical Science, Lund University, Klinikgatan 32, SE-22184 Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Klinikgatan 32, SE-22184 Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Universitätsstr. 2, DE-86159 Augsburg, Germany
| | - Eva Morsing
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - David Ley
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Erik Hedström
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Clinical Physiology, Skåne University Hospital, Lund, Sweden
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Radiology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
3
|
Matuskova H, Porschen LT, Matthes F, Lindgren AG, Petzold GC, Meissner A. Spatiotemporal sphingosine-1-phosphate receptor 3 expression within the cerebral vasculature after ischemic stroke. iScience 2024; 27:110031. [PMID: 38868192 PMCID: PMC11167442 DOI: 10.1016/j.isci.2024.110031] [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: 11/01/2023] [Revised: 03/29/2024] [Accepted: 05/16/2024] [Indexed: 06/14/2024] Open
Abstract
Sphingosine-1-phosphate receptors (S1PRs) are promising therapeutic targets in cardiovascular disease, including ischemic stroke. However, important spatiotemporal information for alterations of S1PR expression is lacking. Here, we investigated the role of S1PR3 in ischemic stroke in rodent models and patient samples. We show that S1PR3 is acutely upregulated in perilesional reactive astrocytes after stroke, and that stroke volume and behavioral deficits are improved in mice lacking S1PR3. Further, we find that administration of an S1PR3 antagonist at 4-h post-stroke, but not at later timepoints, improves stroke outcome. Lastly, we observed higher plasma S1PR3 concentrations in experimental stroke and in patients with ischemic stroke. Together, our results establish S1PR3 as a potential drug target and biomarker in ischemic stroke.
Collapse
Affiliation(s)
- Hana Matuskova
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Division of Vascular Neurology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Lisa T. Porschen
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Frank Matthes
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Arne G. Lindgren
- Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
- Department of Neurology, Rehabilitation Medicine, Memory Disorders and Geriatrics, Skåne University Hospital, Lund, Sweden
| | - Gabor C. Petzold
- Division of Vascular Neurology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Anja Meissner
- Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
4
|
Hsing V, Zhao HQ, Post M, Devine D, McVey MJ. Preservation of recipient plasma sphingosine-1-phosphate levels reduces transfusion-related acute lung injury. Am J Physiol Lung Cell Mol Physiol 2024; 326:L589-L595. [PMID: 38375568 PMCID: PMC11905805 DOI: 10.1152/ajplung.00388.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/30/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024] Open
Abstract
Cold-stored (CS) platelets are once again being reintroduced for clinical use. Transfused CS platelets offer benefits over room temperature-stored (RTS) platelets such as increased hemostatic effects and prolongation of shelf-life. Despite these advantages little is known about their association with transfusion-related acute lung injury (TRALI). TRALI is associated with prolonged storage of RTS platelets and has a mortality of >15%. Determining the safety of CS platelets is important considering their proposed use in TRALI-vulnerable populations with inflammation such as surgical patients or patients with trauma. Donor platelet-derived ceramide causes TRALI, whereas donor platelet sphingosine-1-phosphate (S1P) is barrier protective. Females have higher plasma levels of S1P than males. Cold temperatures increase S1P levels in cells. Therefore, we hypothesized that female (donors or recipients) and/or CS platelets would decrease TRALI. To test this, we compared how male and female donor and recipient allogeneic platelet transfusions of CS (4°C) versus RTS (23°C) platelets stored for 5 days influence murine TRALI. Transfusion of CS platelets significantly reduced recipient lung tissue wet-to-dry ratios, bronchoalveolar lavage total protein, lung tissue myeloperoxidase enzyme activity, histological lung injury scores, and increased plasma sphingosine-1-phosphate (S1P) levels compared with RTS platelet transfusions. Female as opposed to male recipients had less TRALI and higher plasma S1P levels. Female donor mouse platelets had higher S1P levels than males. Mouse and human CS platelets had increased S1P levels compared with RTS platelets. Higher recipient plasma S1P levels appear protective considering females, and males receiving platelets from females or male CS platelets had less TRALI.NEW & NOTEWORTHY Transfusion-related acute lung injury (TRALI) though relatively rare represents a severe lung injury. The sphingolipid sphingosine-1-phosphate (S1P) regulates the severity of platelet-mediated TRALI. Female platelet transfusion recipient plasmas or stored platelets from female donors have higher S1P levels than males, which reduces TRALI. Cold storage of murine platelets preserves platelet-S1P, which reduces TRALI in platelet-transfused recipients.
Collapse
Affiliation(s)
- Vanessa Hsing
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Han Qi Zhao
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Innovation, Canadian Blood Services, Vancouver, British Columbia, Canada
| | - Martin Post
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Dana Devine
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Innovation, Canadian Blood Services, Vancouver, British Columbia, Canada
| | - Mark J McVey
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Kim D, Tian W, Wu TTH, Xiang M, Vinh R, Chang JL, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Abnormal Lymphatic Sphingosine-1-Phosphate Signaling Aggravates Lymphatic Dysfunction and Tissue Inflammation. Circulation 2023; 148:1231-1249. [PMID: 37609838 PMCID: PMC10592179 DOI: 10.1161/circulationaha.123.064181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Lymphedema is a global health problem with no effective drug treatment. Enhanced T-cell immunity and abnormal lymphatic endothelial cell (LEC) signaling are promising therapeutic targets for this condition. Sphingosine-1-phosphate (S1P) mediates a key signaling pathway required for normal LEC function, and altered S1P signaling in LECs could lead to lymphatic disease and pathogenic T-cell activation. Characterizing this biology is relevant for developing much needed therapies. METHODS Human and mouse lymphedema was studied. Lymphedema was induced in mice by surgically ligating the tail lymphatics. Lymphedematous dermal tissue was assessed for S1P signaling. To verify the role of altered S1P signaling effects in lymphatic cells, LEC-specific S1pr1-deficient (S1pr1LECKO) mice were generated. Disease progression was quantified by tail-volumetric and -histopathologic measurements over time. LECs from mice and humans, with S1P signaling inhibition, were then cocultured with CD4 T cells, followed by an analysis of CD4 T-cell activation and pathway signaling. Last, animals were treated with a monoclonal antibody specific to P-selectin to assess its efficacy in reducing lymphedema and T-cell activation. RESULTS Human and experimental lymphedema tissues exhibited decreased LEC S1P signaling through S1P receptor 1 (S1PR1). LEC S1pr1 loss-of-function exacerbated lymphatic vascular insufficiency, tail swelling, and increased CD4 T-cell infiltration in mouse lymphedema. LECs, isolated from S1pr1LECKO mice and cocultured with CD4 T cells, resulted in augmented lymphocyte differentiation. Inhibiting S1PR1 signaling in human dermal LECs promoted T-helper type 1 and 2 (Th1 and Th2) cell differentiation through direct cell contact with lymphocytes. Human dermal LECs with dampened S1P signaling exhibited enhanced P-selectin, an important cell adhesion molecule expressed on activated vascular cells. In vitro, P-selectin blockade reduced the activation and differentiation of Th cells cocultured with shS1PR1-treated human dermal LECs. P-selectin-directed antibody treatment improved tail swelling and reduced Th1/Th2 immune responses in mouse lymphedema. CONCLUSIONS This study suggests that reduction of the LEC S1P signaling aggravates lymphedema by enhancing LEC adhesion and amplifying pathogenic CD4 T-cell responses. P-selectin inhibitors are suggested as a possible treatment for this pervasive condition.
Collapse
Affiliation(s)
- Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Ting-Hsuan Wu
- Stanford University School of Medicine, Stanford, California, USA
- Department of Biochemistry, Stanford Bio-X, Stanford, California, USA
| | - Menglan Xiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Jason Lon Chang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Shenbiao Gu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Seunghee Lee
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Yu Zhu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Torrey Guan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Emilie Claire Schneider
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Evan Bao
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Peter Kao
- Stanford University School of Medicine, Stanford, California, USA
| | - Junliang Pan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark Robert Nicolls
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Kim D, Tian W, Wu TTH, Xiang M, Vinh R, Chang J, Gu S, Lee S, Zhu Y, Guan T, Schneider EC, Bao E, Dixon JB, Kao P, Pan J, Rockson SG, Jiang X, Nicolls MR. Abnormal lymphatic S1P signaling aggravates lymphatic dysfunction and tissue inflammation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.08.23291175. [PMID: 37398237 PMCID: PMC10312855 DOI: 10.1101/2023.06.08.23291175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
BACKGROUND Lymphedema is a global health problem with no effective drug treatment. Enhanced T cell immunity and abnormal lymphatic endothelial cell (LEC) signaling are promising therapeutic targets for this condition. Sphingosine-1-phosphate (S1P) mediates a key signaling pathway required for normal LEC function, and altered S1P signaling in LECs could lead to lymphatic disease and pathogenic T cell activation. Characterizing this biology is relevant for developing much-needed therapies. METHODS Human and mouse lymphedema was studied. Lymphedema was induced in mice by surgically ligating the tail lymphatics. Lymphedematous dermal tissue was assessed for S1P signaling. To verify the role of altered S1P signaling effects in lymphatic cells, LEC-specific S1pr1 -deficient ( S1pr1 LECKO ) mice were generated. Disease progression was quantified by tail-volumetric and -histopathological measurements over time. LECs from mice and humans, with S1P signaling inhibition, were then co-cultured with CD4 T cells, followed by an analysis of CD4 T cell activation and pathway signaling. Finally, animals were treated with a monoclonal antibody specific to P-selectin to assess its efficacy in reducing lymphedema and T cell activation. RESULTS Human and experimental lymphedema tissues exhibited decreased LEC S1P signaling through S1PR1. LEC S1pr1 loss-of-function exacerbated lymphatic vascular insufficiency, tail swelling, and increased CD4 T cell infiltration in mouse lymphedema. LECs, isolated from S1pr1 LECKO mice and co-cultured with CD4 T cells, resulted in augmented lymphocyte differentiation. Inhibiting S1PR1 signaling in human dermal LECs (HDLECs) promoted T helper type 1 and 2 (Th1 and Th2) cell differentiation through direct cell contact with lymphocytes. HDLECs with dampened S1P signaling exhibited enhanced P-selectin, an important cell adhesion molecule expressed on activated vascular cells. In vitro , P-selectin blockade reduced the activation and differentiation of Th cells co-cultured with sh S1PR1 -treated HDLECs. P-selectin-directed antibody treatment improved tail swelling and reduced Th1/Th2 immune responses in mouse lymphedema. CONCLUSION This study suggests that reduction of the LEC S1P signaling aggravates lymphedema by enhancing LEC adhesion and amplifying pathogenic CD4 T cell responses. P-selectin inhibitors are suggested as a possible treatment for this pervasive condition. Clinical Perspective What is New?: Lymphatic-specific S1pr1 deletion exacerbates lymphatic vessel malfunction and Th1/Th2 immune responses during lymphedema pathogenesis. S1pr1 -deficient LECs directly induce Th1/Th2 cell differentiation and decrease anti-inflammatory Treg populations. Peripheral dermal LECs affect CD4 T cell immune responses through direct cell contact.LEC P-selectin, regulated by S1PR1 signaling, affects CD4 T cell activation and differentiation.P-selectin blockade improves lymphedema tail swelling and decreases Th1/Th2 population in the diseased skin.What Are the Clinical Implications?: S1P/S1PR1 signaling in LECs regulates inflammation in lymphedema tissue.S1PR1 expression levels on LECs may be a useful biomarker for assessing predisposition to lymphatic disease, such as at-risk women undergoing mastectomyP-selectin Inhibitors may be effective for certain forms of lymphedema.
Collapse
Affiliation(s)
- Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Timothy Ting-Hsuan Wu
- Stanford University School of Medicine, Stanford, California, USA
- Department of Biochemistry, Stanford Bio-X, Stanford, California, USA
| | - Menglan Xiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Jason Chang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Shenbiao Gu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Seunghee Lee
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Yu Zhu
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Torrey Guan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Emilie Claire Schneider
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Evan Bao
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Peter Kao
- Stanford University School of Medicine, Stanford, California, USA
| | - Junliang Pan
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark Robert Nicolls
- VA Palo Alto Health Care System, Palo Alto, California, USA
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
7
|
SenthilKumar G, Katunaric B, Bordas-Murphy H, Sarvaideo J, Freed JK. Estrogen and the Vascular Endothelium: The Unanswered Questions. Endocrinology 2023; 164:bqad079. [PMID: 37207450 PMCID: PMC10230790 DOI: 10.1210/endocr/bqad079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 05/21/2023]
Abstract
Premenopausal women have a lower incidence of cardiovascular disease (CVD) compared with their age-matched male counterparts; however, this discrepancy is abolished following the transition to menopause or during low estrogen states. This, combined with a large amount of basic and preclinical data indicating that estrogen is vasculoprotective, supports the concept that hormone therapy could improve cardiovascular health. However, clinical outcomes in individuals undergoing estrogen treatment have been highly variable, challenging the current paradigm regarding the role of estrogen in the fight against heart disease. Increased risk for CVD correlates with long-term oral contraceptive use, hormone replacement therapy in older, postmenopausal cisgender females, and gender affirmation treatment for transgender females. Vascular endothelial dysfunction serves as a nidus for the development of many cardiovascular diseases and is highly predictive of future CVD risk. Despite preclinical studies indicating that estrogen promotes a quiescent, functional endothelium, it still remains unclear why these observations do not translate to improved CVD outcomes. The goal of this review is to explore our current understanding of the effect of estrogen on the vasculature, with a focus on endothelial health. Following a discussion regarding the influence of estrogen on large and small artery function, critical knowledge gaps are identified. Finally, novel mechanisms and hypotheses are presented that may explain the lack of cardiovascular benefit in unique patient populations.
Collapse
Affiliation(s)
- Gopika SenthilKumar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovasular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI 53226, USA
| | - Boran Katunaric
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI 53226, USA
| | - Henry Bordas-Murphy
- Cardiovasular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI 53226, USA
| | - Jenna Sarvaideo
- Divison of Endocrinology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Julie K Freed
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovasular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee WI 53226, USA
| |
Collapse
|
8
|
Pepe G, Capocci L, Marracino F, Realini N, Lenzi P, Martinello K, Bovier TF, Bichell TJ, Scarselli P, Di Cicco C, Bowman AB, Digilio FA, Fucile S, Fornai F, Armirotti A, Parlato R, Di Pardo A, Maglione V. Treatment with THI, an inhibitor of sphingosine-1-phosphate lyase, modulates glycosphingolipid metabolism and results therapeutically effective in experimental models of Huntington's disease. Mol Ther 2023; 31:282-299. [PMID: 36116006 PMCID: PMC9840122 DOI: 10.1016/j.ymthe.2022.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/05/2022] [Accepted: 09/06/2022] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder with no effective cure currently available. Over the past few years our research has shown that alterations in sphingolipid metabolism represent a critical determinant in HD pathogenesis. In particular, aberrant metabolism of sphingosine-1-phosphate (S1P) has been reported in multiple disease settings, including human postmortem brains from HD patients. In this study, we investigate the potential therapeutic effect of the inhibition of S1P degradative enzyme SGPL1, by the chronic administration of the 2-acetyl-5-tetrahydroxybutyl imidazole (THI) inhibitor. We show that THI mitigated motor dysfunctions in both mouse and fly models of HD. The compound evoked the activation of pro-survival pathways, normalized levels of brain-derived neurotrophic factor, preserved white matter integrity, and stimulated synaptic functions in HD mice. Metabolically, THI restored normal levels of hexosylceramides and stimulated the autophagic and lysosomal machinery, facilitating the reduction of nuclear inclusions of both wild-type and mutant huntingtin proteins.
Collapse
Affiliation(s)
| | | | | | - Natalia Realini
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | - Tiziana Francesca Bovier
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; Department of Pediatrics Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York 10032, NY, USA
| | - Terry Jo Bichell
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA
| | | | | | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Filomena A Digilio
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Sergio Fucile
- IRCCS Neuromed, Pozzilli (IS) 86077, Italy; Department of Physiology and Pharmacology, Sapienza Rome University, Rome 00185, Italy
| | - Francesco Fornai
- IRCCS Neuromed, Pozzilli (IS) 86077, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Rosanna Parlato
- Division for Neurodegenerative Diseases, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim Heidelberg University, Mannheim 68167, Germany
| | | | | |
Collapse
|
9
|
Crivelli SM, Luo Q, Kruining DV, Giovagnoni C, Mané-Damas M, den Hoedt S, Berkes D, De Vries HE, Mulder MT, Walter J, Waelkens E, Derua R, Swinnen JV, Dehairs J, Wijnands EPM, Bieberich E, Losen M, Martinez-Martinez P. FTY720 decreases ceramides levels in the brain and prevents memory impairments in a mouse model of familial Alzheimer's disease expressing APOE4. Biomed Pharmacother 2022; 152:113240. [PMID: 35689862 DOI: 10.1016/j.biopha.2022.113240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 11/24/2022] Open
Abstract
The protection mediated by the bioactive sphingolipid sphingosine-1-phosphate (S1P) declines during Alzheimer's disease (AD) progression, especially in patients carrying the apolipoprotein E ε4 (APOE4) isoform. The drug FTY720 mimics S1P bioactivity, but its efficacy in treating AD is unclear. Two doses of FTY720 (0.1 mg / kg and 0.5 mg / kg daily) were given by oral gavage for 15 weeks to transgenic mouse models of familial AD carrying human apolipoprotein E (APOE) APOE3 (E3FAD) or APOE4 (E4FAD). After 12 weeks of treatment, animals were subjected to behavioral tests for memory, locomotion, and anxiety. Blood was withdrawn at different time points and brains were collected for sphingolipids analysis by mass spectrometry, gene expression by RT-PCR and Aβ quantification by ELISA. We discovered that low levels of S1P in the plasma is associated with a higher probability of failing the memory test and that FTY720 prevents memory impairments in E4FAD. The beneficial effect of FTY720 was induced by a shift of the sphingolipid metabolism in the brain towards a lower production of toxic metabolites, like ceramide d18:1/16:0 and d18:1/22:0, and reduction of amyloid-β burden and inflammation. In conclusion, we provide further evidence of the druggability of the sphingolipid system in AD.
Collapse
Affiliation(s)
- Simone M Crivelli
- Maastricht University, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht 6200MD, the Netherlands; Department of Physiology, University of Kentucky College of Medicine, Lexington 40506, KY, USA.
| | - Qian Luo
- Maastricht University, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht 6200MD, the Netherlands
| | - Daan van Kruining
- Maastricht University, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht 6200MD, the Netherlands
| | - Caterina Giovagnoni
- Maastricht University, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht 6200MD, the Netherlands
| | - Marina Mané-Damas
- Maastricht University, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht 6200MD, the Netherlands
| | - Sandra den Hoedt
- Department of Internal Medicine, Laboratory Vascular Medicine, Erasmus MC University Medical Center, Rotterdam 3000CA, the Netherlands
| | - Dusan Berkes
- Department of Organic Chemistry, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovak Republic
| | - Helga E De Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam 1007MB, the Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Laboratory Vascular Medicine, Erasmus MC University Medical Center, Rotterdam 3000CA, the Netherlands
| | - Jochen Walter
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn D-53127, Germany
| | - Etienne Waelkens
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven 3000, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven 3000, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven, Leuven 3000, Belgium
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, KU Leuven, Leuven 3000, Belgium
| | - Erwin P M Wijnands
- Department of Pathology, Maastricht University, Maastricht 6200MD, the Netherlands
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington 40506, KY, USA; Veterans Affairs Medical Center, Lexington, KY 40502, USA
| | - Mario Losen
- Maastricht University, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht 6200MD, the Netherlands
| | - Pilar Martinez-Martinez
- Maastricht University, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht 6200MD, the Netherlands.
| |
Collapse
|
10
|
Deng S, Ao Z, Liu B, She Q, Du J, Liu Y, Jing X. Correlation between plasma sphingosine-1-phosphate and the occurrence and severity of coronary heart disease in postmenopausal women. Menopause 2022; 29:920-925. [PMID: 35881936 DOI: 10.1097/gme.0000000000002004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Sphingosine-1-phosphate (S1P) is a bioactive sphingosine with antiatherosclerotic effects. The incidence of coronary heart disease (CHD) increases significantly among women after menopause. We explored the relationship between plasma S1P levels and the occurrence and severity of CHD in postmenopausal women. METHODS Postmenopausal women admitted to our hospital for coronary angiography because of chest pain-like symptoms were included in our study. By 1:1 age matching (age difference ≤5 y), 166 women in the CHD group and control group were enrolled. The plasma S1P concentration was determined, and the Gensini score was calculated to decide the severity of CHD. RESULTS Plasma S1P levels were significantly lower in the CHD group of postmenopausal women ( P < 0.001). S1P (odds ratio, 0.952; 95% CI, 0.934-0.970) was an independent predictor of the occurrence of CHD in postmenopausal women. The area under the curve for S1P to predict the occurrence of CHD was 0.653 (95% CI, 0.595-0.712), and the cutoff value was 96.89 ng/mL. The plasma S1P level was the lowest in the high-tertile group of the Gensini score ( P < 0.001), and the plasma S1P (odds ratio, 0.948; 95% CI, 0.926-0.970) was an independent predictor of a high Gensini score in postmenopausal women with CHD. CONCLUSIONS Plasma S1P is an independent risk factor of the occurrence and severity of CHD in postmenopausal women. The occurrence and aggravation of CHD in postmenopausal women may be related to levels of S1P.
Collapse
Affiliation(s)
- Songbai Deng
- From the Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zehui Ao
- the Department of Cardiology, People's Hospital of Xiushan Country, Chongqing, China
| | - Bin Liu
- From the Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang She
- From the Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- From the Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- From the Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- From the Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Kasaven LS, Saso S, Getreu N, O'Neill H, Bracewell-Milnes T, Shakir F, Yazbek J, Thum MY, Nicopoullos J, Ben Nagi J, Hardiman P, Diaz-Garcia C, Jones BP. Age-related fertility decline: is there a role for elective ovarian tissue cryopreservation? Hum Reprod 2022; 37:1970-1979. [PMID: 35734904 PMCID: PMC9433842 DOI: 10.1093/humrep/deac144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/29/2022] [Indexed: 11/21/2022] Open
Abstract
Age-related fertility decline (ARFD) is a prevalent concern amongst western cultures due to the increasing age of first-time motherhood. Elective oocyte and embryo cryopreservation remain the most established methods of fertility preservation, providing women the opportunity of reproductive autonomy to preserve their fertility and extend their childbearing years to prevent involuntary childlessness. Whilst ovarian cortex cryopreservation has been used to preserve reproductive potential in women for medical reasons, such as in pre- or peripubertal girls undergoing gonadotoxic chemotherapy, it has not yet been considered in the context of ARFD. As artificial reproductive technology (ART) and surgical methods of fertility preservation continue to evolve, it is a judicious time to review current evidence and consider alternative options for women wishing to delay their fertility. This article critically appraises elective oocyte cryopreservation as an option for women who use it to mitigate the risk of ARFD and introduces the prospect of elective ovarian cortex cryopreservation as an alternative.
Collapse
Affiliation(s)
- Lorraine S Kasaven
- West London Gynaecological Cancer Centre, Hammersmith Hospital, Imperial College NHS Trust, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK.,Cutrale Perioperative and Ageing Group, Sir Michael Uren Hub, Imperial College London, London, UK
| | - Srdjan Saso
- West London Gynaecological Cancer Centre, Hammersmith Hospital, Imperial College NHS Trust, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Natalie Getreu
- Translational Ovarian Physiology and Pathophysiology, Institute for Women's Health, University College London, London, UK
| | - Helen O'Neill
- Genome Editing and Reproductive Genetics Group, Institute for Women's Health, University College London, London, UK
| | | | - Fevzi Shakir
- Royal Free London NHS Foundation Trust, London, UK
| | - Joseph Yazbek
- West London Gynaecological Cancer Centre, Hammersmith Hospital, Imperial College NHS Trust, London, UK
| | - Meen-Yau Thum
- Lister Fertility Clinic, The Lister Hospital, London, UK
| | | | - Jara Ben Nagi
- Centre for Reproductive and Genetic Health, London, UK
| | | | - Cesar Diaz-Garcia
- IVI London, IVIRMA Global, London, UK.,EGA Institute for Women's Health, University College London, London, UK
| | - Benjamin P Jones
- West London Gynaecological Cancer Centre, Hammersmith Hospital, Imperial College NHS Trust, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
12
|
Montefusco D, Jamil M, Maczis MA, Schroeder W, Levi M, Ranjit S, Allegood J, Bandyopadhyay D, Retnam R, Spiegel S, Cowart LA. Sphingosine Kinase 1 Mediates Sexual Dimorphism in Fibrosis in a Mouse Model of NASH. Mol Metab 2022; 62:101523. [PMID: 35671973 PMCID: PMC9194589 DOI: 10.1016/j.molmet.2022.101523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/04/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Men with non-alcoholic fatty liver disease (NAFLD) are more likely to progress to nonalcoholic steatohepatitis (NASH) and liver fibrosis than women. However, the underlying molecular mechanisms of this dimorphism is unclear. We have previously shown that mice with global deletion of SphK1, the enzyme that produces the bioactive sphingolipid metabolite sphingosine 1-phosphate (S1P), were protected from development of NASH. The aim of this study was to elucidate the role of hepatocyte-specific SphK1 in development of NASH and to compare its contribution to hepatosteatosis in male and female mice. RESULTS We generated hepatocyte-specific SphK1 knockout mice (SphK1-hKO). Unlike the global knockout, SphK1-hKO male mice were not protected from diet-induced steatosis, inflammation, or fibrogenesis. In contrast, female SphK1-hKO mice were protected from inflammation. Surprisingly, however, in these female mice, there was a ∼10-fold increase in the fibrosis markers Col1α1 and 2-3 fold induction of alpha smooth muscle actin and the pro-fibrotic chemokine CCL5. Because increased fibrosis in female SphK1-hKO mice occurred despite an attenuated inflammatory response, we investigated the crosstalk between hepatocytes and hepatic stellate cells, central players in fibrosis. We found that estrogen stimulated release of S1P from female hepatocytes preventing TGFβ-induced expression of Col1α1 in HSCs via S1PR3. CONCLUSIONS The results revealed a novel pathway of estrogen-mediated cross-talk between hepatocytes and HSCs that may contribute to sex differences in NAFLD through an anti-fibrogenic function of the S1P/S1PR3 axis. This pathway is susceptible to pharmacologic manipulation, which may lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- David Montefusco
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, VA, USA.
| | - Maryam Jamil
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, VA, USA
| | - Melissa A Maczis
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, VA, USA
| | - William Schroeder
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, VA, USA
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, USA
| | - Suman Ranjit
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, USA
| | - Jeremy Allegood
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, VA, USA
| | | | - Reuben Retnam
- Virginia Commonwealth University Department of Biostatistics, VA, USA
| | - Sarah Spiegel
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, VA, USA
| | - L Ashley Cowart
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, VA, USA; Hunter Holmes McGuire VAMC, Richmond, VA, USA
| |
Collapse
|
13
|
Sex Differences in Cardiovascular Diseases: A Matter of Estrogens, Ceramides, and Sphingosine 1-Phosphate. Int J Mol Sci 2022; 23:ijms23074009. [PMID: 35409368 PMCID: PMC8999971 DOI: 10.3390/ijms23074009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/30/2022] Open
Abstract
The medical community recognizes sex-related differences in pathophysiology and cardiovascular disease outcomes (CVD), culminating with heart failure. In general, pre-menopausal women tend to have a better prognosis than men. Explaining why this occurs is not a simple matter. For decades, sex hormones like estrogens (Es) have been identified as one of the leading factors driving these sex differences. Indeed, Es seem protective in women as their decline, during and after menopause, coincides with an increased CV risk and HF development. However, clinical trials demonstrated that E replacement in post-menopause women results in adverse cardiac events and increased risk of breast cancer. Thus, a deeper understanding of E-related mechanisms is needed to provide a vital gateway toward better CVD prevention and treatment in women. Of note, sphingolipids (SLs) and their metabolism are strictly related to E activities. Among the SLs, ceramide and sphingosine 1-phosphate play essential roles in mammalian physiology, particularly in the CV system, and appear differently modulated in males and females. In keeping with this view, here we explore the most recent experimental and clinical observations about the role of E and SL metabolism, emphasizing how these factors impact the CV system.
Collapse
|
14
|
den Hoedt S, Crivelli SM, Leijten FPJ, Losen M, Stevens JAA, Mané-Damas M, de Vries HE, Walter J, Mirzaian M, Sijbrands EJG, Aerts JMFG, Verhoeven AJM, Martinez-Martinez P, Mulder MT. Effects of Sex, Age, and Apolipoprotein E Genotype on Brain Ceramides and Sphingosine-1-Phosphate in Alzheimer's Disease and Control Mice. Front Aging Neurosci 2021; 13:765252. [PMID: 34776936 PMCID: PMC8579780 DOI: 10.3389/fnagi.2021.765252] [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: 08/26/2021] [Accepted: 09/29/2021] [Indexed: 11/28/2022] Open
Abstract
Apolipoprotein ε4 (APOE)4 is a strong risk factor for the development of Alzheimer’s disease (AD) and aberrant sphingolipid levels have been implicated in AD. We tested the hypothesis that the APOE4 genotype affects brain sphingolipid levels in AD. Seven ceramides and sphingosine-1-phosphate (S1P) were quantified by LC-MSMS in hippocampus, cortex, cerebellum, and plasma of <3 months and >5 months old human APOE3 and APOE4-targeted replacement mice with or without the familial AD (FAD) background of both sexes (145 animals). APOE4 mice had higher Cer(d18:1/24:0) levels in the cortex (1.7-fold, p = 0.002) than APOE3 mice. Mice with AD background showed higher levels of Cer(d18:1/24:1) in the cortex than mice without (1.4-fold, p = 0.003). S1P levels were higher in all three brain regions of older mice than of young mice (1.7-1.8-fold, all p ≤ 0.001). In female mice, S1P levels in hippocampus (r = −0.54 [−0.70, −0.35], p < 0.001) and in cortex correlated with those in plasma (r = −0.53 [−0.71, −0.32], p < 0.001). Ceramide levels were lower in the hippocampus (3.7–10.7-fold, all p < 0.001), but higher in the cortex (2.3–12.8-fold, p < 0.001) of female than male mice. In cerebellum and plasma, sex effects on individual ceramides depended on acyl chain length (9.5-fold lower to 11.5-fold higher, p ≤ 0.001). In conclusion, sex is a stronger determinant of brain ceramide levels in mice than APOE genotype, AD background, or age. Whether these differences impact AD neuropathology in men and women remains to be investigated.
Collapse
Affiliation(s)
- Sandra den Hoedt
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Simone M Crivelli
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Frank P J Leijten
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Mario Losen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Jo A A Stevens
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Marina Mané-Damas
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, VU Medical Center, Amsterdam UMC, Amsterdam, Netherlands
| | - Jochen Walter
- Department of Neurology, University Hospital Bonn, Venusberg Campus, Bonn, Germany
| | - Mina Mirzaian
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Adrie J M Verhoeven
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pilar Martinez-Martinez
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
15
|
Jujic A, Matthes F, Vanherle L, Petzka H, Orho-Melander M, Nilsson PM, Magnusson M, Meissner A. Plasma S1P (Sphingosine-1-Phosphate) Links to Hypertension and Biomarkers of Inflammation and Cardiovascular Disease: Findings From a Translational Investigation. Hypertension 2021; 78:195-209. [PMID: 33993723 DOI: 10.1161/hypertensionaha.120.17379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Amra Jujic
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Lund University Diabetes Centre (A.J.), Lund University, Malmö, Sweden
| | - Frank Matthes
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Department of Experimental Medical Sciences (F.M., L.V., A.M.), Lund University, Malmö, Sweden
| | - Lotte Vanherle
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Department of Experimental Medical Sciences (F.M., L.V., A.M.), Lund University, Malmö, Sweden
| | - Henning Petzka
- Department of Mathematics, Lund Technical University, Sweden (H.P.)
| | - Marju Orho-Melander
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Department of Internal Medicine, Clinical Research Unit, Malmö, Sweden (P.M.N.)
| | - Martin Magnusson
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Hypertension in Africa Research Team, North West University Potchefstroom, South Africa (M.M.)
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden (M.M.)
| | - Anja Meissner
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Department of Experimental Medical Sciences (F.M., L.V., A.M.), Lund University, Malmö, Sweden
| |
Collapse
|
16
|
Pitman M, Oehler MK, Pitson SM. Sphingolipids as multifaceted mediators in ovarian cancer. Cell Signal 2021; 81:109949. [PMID: 33571664 DOI: 10.1016/j.cellsig.2021.109949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/19/2022]
Abstract
Ovarian cancer is the most lethal gynaecological malignancy. It is commonly diagnosed at advanced stage when it has metastasised to the abdominal cavity and treatment becomes very challenging. While current standard therapy involving debulking surgery and platinum + taxane-based chemotherapy is associated with high response rates initially, the large majority of patients relapse and ultimately succumb to chemotherapy-resistant disease. In order to improve survival novel strategies for early detection and therapeutics against treatment-refractory disease are urgently needed. A promising new target against ovarian cancer is the sphingolipid pathway which is commonly hijacked in cancer to support cell proliferation and survival and has been shown to promote chemoresistance and metastasis in a wide range of malignant neoplasms. In particular, the sphingosine kinase 1-sphingosine 1-phosphate receptor 1 axis has been shown to be altered in ovarian cancer in multiple ways and therefore represents an attractive therapeutic target. Here we review the roles of sphingolipids in ovarian cancer progression, metastasis and chemoresistance, highlighting novel strategies to target this pathway that represent potential avenues to improve patient survival.
Collapse
Affiliation(s)
- MelissaR Pitman
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5000, Australia.
| | - Martin K Oehler
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5000, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
17
|
Bergougnan L, Andersen G, Plum-Mörschel L, Evaristi MF, Poirier B, Tardat A, Ermer M, Herbrand T, Arrubla J, Coester HV, Sansone R, Heiss C, Vitse O, Hurbin F, Boiron R, Benain X, Radzik D, Janiak P, Muslin AJ, Hovsepian L, Kirkesseli S, Deutsch P, Parkar AA. Endothelial-protective effects of a G-protein-biased sphingosine-1 phosphate receptor-1 agonist, SAR247799, in type-2 diabetes rats and a randomized placebo-controlled patient trial. Br J Clin Pharmacol 2020; 87:2303-2320. [PMID: 33125753 PMCID: PMC8247405 DOI: 10.1111/bcp.14632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/12/2022] Open
Abstract
Aims SAR247799 is a G‐protein‐biased sphingosine‐1 phosphate receptor‐1 (S1P1) agonist designed to activate endothelial S1P1 and provide endothelial‐protective properties, while limiting S1P1 desensitization and consequent lymphocyte‐count reduction associated with higher doses. The aim was to show whether S1P1 activation can promote endothelial effects in patients and, if so, select SAR247799 doses for further clinical investigation. Methods Type‐2 diabetes patients, enriched for endothelial dysfunction (flow‐mediated dilation, FMD <7%; n = 54), were randomized, in 2 sequential cohorts, to 28‐day once‐daily treatment with SAR247799 (1 or 5 mg in ascending cohorts), placebo or 50 mg sildenafil (positive control) in a 5:2:2 ratio per cohort. Endothelial function was assessed by brachial artery FMD. Renal function, biomarkers and lymphocytes were measured following 5‐week SAR247799 treatment (3 doses) to Zucker diabetic fatty rats and the data used to select the doses for human testing. Results The maximum FMD change from baseline vs placebo for all treatments was reached on day 35; mean differences vs placebo were 0.60% (95% confidence interval [CI] −0.34 to 1.53%; P = .203) for 1 mg SAR247799, 1.07% (95% CI 0.13 to 2.01%; P = .026) for 5 mg SAR247799 and 0.88% (95% CI −0.15 to 1.91%; P = .093) for 50 mg sildenafil. Both doses of SAR247799 were well tolerated, did not affect blood pressure, and were associated with minimal‐to‐no lymphocyte reduction and small‐to‐moderate heart rate decrease. Conclusion These data provide the first human evidence suggesting endothelial‐protective properties of S1P1 activation, with SAR247799 being as effective as the clinical benchmark, sildenafil. Further clinical testing of SAR247799, at sub‐lymphocyte‐reducing doses (≤5 mg), is warranted in vascular diseases associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Luc Bergougnan
- Sanofi R&D, 1 Avenue Pierre Brossolette, Chilly Mazarin, France
| | | | | | | | - Bruno Poirier
- Sanofi R&D, 1 Avenue Pierre Brossolette, Chilly Mazarin, France
| | - Agnes Tardat
- Sanofi R&D, 371 Rue du Professeur Blayac, Montpellier, France
| | | | | | | | | | - Roberto Sansone
- Division of Cardiology, Pulmonary diseases and Vascular medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christian Heiss
- Department of Clinical and Experimental Medicine, University of Surrey, Stag Hill, Guildford, UK
| | - Olivier Vitse
- Sanofi R&D, 371 Rue du Professeur Blayac, Montpellier, France
| | - Fabrice Hurbin
- Sanofi R&D, 371 Rue du Professeur Blayac, Montpellier, France
| | - Rania Boiron
- Sanofi R&D, 1 Avenue Pierre Brossolette, Chilly Mazarin, France
| | - Xavier Benain
- Sanofi R&D, 371 Rue du Professeur Blayac, Montpellier, France
| | - David Radzik
- Sanofi R&D, 1 Avenue Pierre Brossolette, Chilly Mazarin, France
| | - Philip Janiak
- Sanofi R&D, 1 Avenue Pierre Brossolette, Chilly Mazarin, France
| | | | | | | | | | | |
Collapse
|
18
|
Dhangadamajhi G, Singh S. Sphingosine 1-Phosphate in Malaria Pathogenesis and Its Implication in Therapeutic Opportunities. Front Cell Infect Microbiol 2020; 10:353. [PMID: 32923406 PMCID: PMC7456833 DOI: 10.3389/fcimb.2020.00353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/08/2020] [Indexed: 11/13/2022] Open
Abstract
Sphingosine 1-Phosphate (S1P) is a bioactive lipid intermediate in the sphingolipid metabolism, which exist in two pools, intracellular and extracellular, and each pool has a different function. The circulating extracellular pool, specifically the plasma S1P is shown to be important in regulating various physiological processes related to malaria pathogenesis in recent years. Although blood cells (red blood cells and platelets), vascular endothelial cells and hepatocytes are considered as the important sources of plasma S1P, their extent of contribution is still debated. The red blood cells (RBCs) and platelets serve as a major repository of intracellular S1P due to lack, or low activity of S1P degrading enzymes, however, contribution of platelets toward maintaining plasma S1P is shown negligible under normal condition. Substantial evidences suggest platelets loss during falciparum infection as a contributing factor for severe malaria. However, platelets function as a source for plasma S1P in malaria needs to be examined experimentally. RBC being the preferential site for parasite seclusion, and having the ability of trans-cellular S1P transportation to EC upon tight cell-cell contact, might play critical role in differential S1P distribution and parasite growth. In the present review, we have summarized the significance of both the S1P pools in the context of malaria, and how the RBC content of S1P can be channelized in better ways for its possible implication in therapeutic opportunities to control malaria.
Collapse
Affiliation(s)
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
19
|
Plasma sphingosine 1-phosphate concentrations and cardiovascular autonomic neuropathy in individuals with type 2 diabetes. Sci Rep 2020; 10:12768. [PMID: 32728147 PMCID: PMC7391653 DOI: 10.1038/s41598-020-69566-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to test the hypothesis that plasma sphingosine 1-phosphate (S1P) levels are associated with the risk of cardiovascular autonomic neuropathy (CAN) in type 2 diabetes patients. This cross-sectional study included 287 individuals with type 2 diabetes. CAN was evaluated using cardiovascular reflex tests. Logistic regression analyses were conducted to assess the relationship between plasma S1P levels and CAN. Plasma S1P concentrations were significantly lower in individuals with CAN than in those without CAN. There was a significant interaction between plasma S1P levels and sex with respect to CAN (p for interaction = 0.003). When stratified by sex, the association between plasma S1P levels and CAN exhibited a sex difference; in multivariable analysis, plasma S1P levels were significantly associated with CAN in women (odds ratio per standard deviation increase in the log-transformed value, 0.40; 95% confidence interval, 0.23–0.70, p = 0.001). However, there was no significant association between plasma S1P and CAN in men. Plasma S1P concentrations were inversely associated with CAN only in women with type 2 diabetes.
Collapse
|
20
|
Zhang W, Liu XD, Wang JW, Meng LF, Zhang YG, Wang JY. The sphingosine-1-phosphate/RhoA/Rho associated kinases/myosin light chain pathway in detrusor of female rats is down-regulated in response to ovariectomy. Chin Med J (Engl) 2020; 133:1203-1210. [PMID: 32433052 PMCID: PMC7249712 DOI: 10.1097/cm9.0000000000000767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Dysuria is one of the main symptoms of genitourinary syndrome of menopause, which causes serious disruption to the normal life of peri-menopausal women. Studies have shown that it is related to decrease of detrusor contractile function, but the exact mechanism is still poorly understood. Previous results have suggested that the sphingosine-1-phosphate (S1P) pathway can regulate detrusor contraction, and this pathway is affected by estrogen in various tissues. However, how estrogen affects this pathway in the detrusor has not been investigated. In this study, we detected changes of the S1P/RhoA/Rho associated kinases (ROCK)/myosin light chain (MLC) pathway in the detrusor of ovariectomized rats in order to explore the underlying mechanism of dysuria during peri-menopause. METHODS Thirty-six female Sprague-Dawley rats were randomly divided into SHAM (sham operation), OVX (ovariectomy), and E groups (ovariectomy + estrogen), with 12 rats in each group. We obtained bladder detrusor tissues from each group and examined the mRNA and protein levels of the major components of the S1P/RhoA/ROCK/MLC pathway using quantitative real-time polymerase chain reaction and Western blotting, respectively. We also quantified the content of S1P in the detrusor using an enzyme linked immunosorbent assay. Finally, we compared results between the groups with one-way analysis of variance. RESULTS The components of the S1P pathway and the RhoA/ROCK/MLC pathway of the OVX group were significantly decreased, as compared with SHAM group. The percent decreases of the components in the S1P pathway were as follows: sphingosine kinase 1 (mRNA: 39%, protein: 45%) (both P < 0.05), S1P (21.73 ± 1.09 nmol/g vs. 18.86 ± 0.69 nmol/g) (P < 0.05), and S1P receptor 2/3 (S1PR2/3) (mRNA: 25%, 27%, respectively) (P < 0.05). However, the protein expression levels of S1PR2/3 and the protein and mRNA levels of SphK2 and S1PR1 did not show significant differences between groups (P > 0.05). The percent decreases of the components in the RhoA/ROCK/MLC pathway were as follows: ROCK2 (protein: 41%, mRNA: 36%) (both P < 0.05), p-MYPT1 (protein: 54%) (P < 0.05), and p-MLC20 (protein: 47%) (P < 0.05), but there were no significant differences in the mRNA and protein levels of RhoA, ROCK1, MYPT1, and MLC20 (all P > 0.05). In addition, all of the above-mentioned decreases could be reversed after estrogen supplementation (E group vs. SHAM group) (all P > 0.05). CONCLUSION In this study, we confirmed that ovariectomy is closely associated with the down-regulation of the S1P/RhoA/ROCK/MLC pathway in the rat detrusor, which may be one mechanism of dysuria caused by decreased contractile function of the female detrusor during peri-menopause.
Collapse
Affiliation(s)
- Wei Zhang
- Peking University Fifth School of Clinical Medicine, Beijing 100730, China
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Xiao-Dong Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Jia-Wen Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Ling-Feng Meng
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Yao-Guang Zhang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Jian-Ye Wang
- Peking University Fifth School of Clinical Medicine, Beijing 100730, China
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| |
Collapse
|
21
|
Li Y, Zhang W, Li J, Sun Y, Yang Q, Wang S, Luo X, Wang W, Wang K, Bai W, Zhang H, Qin L. The imbalance in the aortic ceramide/sphingosine-1-phosphate rheostat in ovariectomized rats and the preventive effect of estrogen. Lipids Health Dis 2020; 19:95. [PMID: 32430006 PMCID: PMC7236922 DOI: 10.1186/s12944-020-01279-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background The prevalence of hypertension in young women is lower than that in age-matched men while the prevalence of hypertension in women is significantly increased after the age of 50 (menopause) and is greater than that in men. It is already known that sphingosine-1-phosphate (S1P) and ceramide regulate vascular tone with opposing effects. This study aimed to explore the effects of ovariectomy and estrogen supplementation on the ceramide/S1P rheostat of the aorta in rats, and to explore a potential mechanism for perimenopausal hypertension and a brand-new target for menopausal hormone therapy to protect vessels. Methods In total, 30 female adult SD rats were randomly divided into three groups: The sham operation group (SHAM), ovariectomy group (OVX) and ovariectomy plus estrogen group (OVX + E). After 4 weeks of treatment, the blood pressure (BP) of the rats was monitored by a noninvasive system; the sphingolipid content (e.g., ceramide and S1P) was detected by liquid chromatography-mass spectrometry (LC-MS); the expression of the key enzymes involved in ceramide anabolism and catabolism was measured by real-time fluorescence quantitative polymerase chain reaction (qPCR); and the expression of key enzymes and proteins in the sphingosine kinase 1/2 (SphK1/2)-S1P-S1P receptor 1/2/3 (S1P1/2/3) signaling pathway was detected by qPCR and western blotting. Results In the OVX group compared with the SHAM group, the systolic BP (SBP), diastolic BP (DBP) and pulse pressure (PP) increased significantly, especially the SBP and PP (P < 0.001). For aortic ceramide metabolism, the mRNA level of key enzymes involved in anabolism and catabolism decreased in parallel 2–3 times, while the contents of total ceramide and certain long-chain subtypes increased significantly (P < 0.05). As for the S1P signaling pathway, SphK1/2, the key enzymes involved in S1P synthesis, decreased significantly, and the content of S1P decreased accordingly (P < 0.01). The S1P receptors showed various trends: S1P1 was significantly down-regulated, S1P2 was significantly up-regulated, and S1P3 showed no significant difference. No significant difference existed between the SHAM and OVX + E groups for most of the above parameters (P > 0.05). Conclusions Ovariectomy resulted in the imbalance of the aortic ceramide/S1P rheostat in rats, which may be a potential mechanism underlying the increase in SBP and PP among perimenopausal women. Besides, the ceramide/S1P rheostat may be a novel mechanism by which estrogen protects vessels.
Collapse
Affiliation(s)
- Yao Li
- Department of Cardiology, Peking University People's Hospital, No. 11 South Avenue, Beijing, 100044, Xi Zhi Men Xicheng District, China
| | - Wei Zhang
- Department of Urology, Peking University Fifth School of Clinical Medicine, Beijing, 100730, China
| | - Junlei Li
- Department of Cardiology, Peking University People's Hospital, No. 11 South Avenue, Beijing, 100044, Xi Zhi Men Xicheng District, China
| | - Yanrong Sun
- Department of Anatomy and Embryology, Peking University Health Science Center, No. 38, Xueyuan Road, Beijing, 100191, Haidian District, China
| | - Qiyue Yang
- Department of Anatomy and Embryology, Peking University Health Science Center, No. 38, Xueyuan Road, Beijing, 100191, Haidian District, China
| | - Sinan Wang
- Department of Stomatology, General Hospital of Armed Police, Beijing, 100039, China
| | - Xiaofeng Luo
- Department of Stomatology, General Hospital of Armed Police, Beijing, 100039, China
| | - Wenjuan Wang
- Department of Anatomy and Embryology, Peking University Health Science Center, No. 38, Xueyuan Road, Beijing, 100191, Haidian District, China
| | - Ke Wang
- Department of Anatomy and Embryology, Peking University Health Science Center, No. 38, Xueyuan Road, Beijing, 100191, Haidian District, China
| | - Wenpei Bai
- Department of Obstetrics and Gynecology, Shijitan Hospital, Beijing, 100038, China
| | - Haicheng Zhang
- Department of Cardiology, Peking University People's Hospital, No. 11 South Avenue, Beijing, 100044, Xi Zhi Men Xicheng District, China.
| | - Lihua Qin
- Department of Anatomy and Embryology, Peking University Health Science Center, No. 38, Xueyuan Road, Beijing, 100191, Haidian District, China.
| |
Collapse
|
22
|
Li T, Hu S, Pang X, Wang J, Yin J, Li F, Wang J, Yang X, Xia B, Liu Y, Song W, Guo S. The marine-derived furanone reduces intracellular lipid accumulation in vitro by targeting LXRα and PPARα. J Cell Mol Med 2020; 24:3384-3398. [PMID: 31981312 PMCID: PMC7131916 DOI: 10.1111/jcmm.15012] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 12/22/2022] Open
Abstract
Recent studies have demonstrated that commercially available lipid-lowering drugs cause various side effects; therefore, searching for anti-hyperlipidaemic compounds with lower toxicity is a research hotspot. This study was designed to investigate whether the marine-derived compound, 5-hydroxy-3-methoxy-5-methyl-4-butylfuran-2(5H)-one, has an anti-hyperlipidaemic activity, and the potential underlying mechanism in vitro. Results showed that the furanone had weaker cytotoxicity compared to positive control drugs. In RAW 264.7 cells, the furanone significantly lowered ox-LDL-induced lipid accumulation (~50%), and its triglyceride (TG)-lowering effect was greater than that of liver X receptor (LXR) agonist T0901317. In addition, it significantly elevated the protein levels of peroxisome proliferator-activated receptors (PPARα) and ATP-binding cassette (ABC) transporters, which could be partially inhibited by LXR antagonists, GSK2033 and SR9243. In HepG2 cells, it significantly decreased oleic acid-induced lipid accumulation, enhanced the protein levels of low-density lipoprotein receptor (LDLR), ABCG5, ABCG8 and PPARα, and reduced the expression of sterol regulatory element-binding protein 2 (~32%). PPARα antagonists, GW6471 and MK886, could significantly inhibit the furanone-induced lipid-lowering effect. Furthermore, the furanone showed a significantly lower activity on the activation of the expression of lipogenic genes compared to T0901317. Taken together, the furanone exhibited a weak cytotoxicity but had powerful TC- and TG-lowering effects most likely through targeting LXRα and PPARα, respectively. These findings indicate that the furanone has a potential application for the treatment of dyslipidaemia.
Collapse
Affiliation(s)
- Ting Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shu‐Mei Hu
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Yan Pang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jun‐feng Wang
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Jia‐Yu Yin
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Fa‐Hui Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jin Wang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Qian Yang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Bin Xia
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Yong‐Hong Liu
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Wei‐Guo Song
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shou‐Dong Guo
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| |
Collapse
|
23
|
Józefczuk E, Nosalski R, Saju B, Crespo E, Szczepaniak P, Guzik TJ, Siedlinski M. Cardiovascular Effects of Pharmacological Targeting of Sphingosine Kinase 1. Hypertension 2020; 75:383-392. [PMID: 31838904 PMCID: PMC7055939 DOI: 10.1161/hypertensionaha.119.13450] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/11/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023]
Abstract
High blood pressure is a risk factor for cardiovascular diseases. Ang II (angiotensin II), a key pro-hypertensive hormone, mediates target organ consequences such as endothelial dysfunction and cardiac hypertrophy. S1P (sphingosine-1-phosphate), produced by Sphk1 (sphingosine kinase 1), plays a pivotal role in the pathogenesis of hypertension and downstream organ damage, as it controls vascular tone and regulates cardiac remodeling. Accordingly, we aimed to examine if pharmacological inhibition of Sphk1 using selective inhibitor PF543 can represent a useful vasoprotective and cardioprotective anti-hypertensive strategy in vivo. PF543 was administered intraperitoneally throughout a 14-day Ang II-infusion in C57BL6/J male mice. Pharmacological inhibition of Sphk1 improved endothelial function of arteries of hypertensive mice that could be mediated via decrease in eNOS (endothelial nitric oxide synthase) phosphorylation at T495. This effect was independent of blood pressure. Importantly, PF543 also reduced cardiac hypertrophy (heart to body weight ratio, 5.6±0.2 versus 6.4±0.1 versus 5.9±0.2 mg/g; P<0.05 for Sham, Ang II+placebo, and Ang II+PF543-treated mice, respectively). Mass spectrometry revealed that PF543 elevated cardiac sphingosine, that is, Sphk1 substrate, content in vivo. Mechanistically, RNA-Seq indicated a decreased expression of cardiac genes involved in actin/integrin organization, S1pr1 signaling, and tissue remodeling. Indeed, downregulation of Rock1 (Rho-associated coiled-coil containing protein kinase 1), Stat3 (signal transducer and activator of transcription 3), PKC (protein kinase C), and ERK1/2 (extracellular signal-regulated kinases 1/2) level/phosphorylation by PF543 was observed. In summary, pharmacological inhibition of Sphk1 partially protects against Ang II-induced cardiac hypertrophy and endothelial dysfunction. Therefore, it may represent a promising target for harnessing residual cardiovascular risk in hypertension.
Collapse
Affiliation(s)
- Ewelina Józefczuk
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| | - Ryszard Nosalski
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Blessy Saju
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Eva Crespo
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Piotr Szczepaniak
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| | - Tomasz Jan Guzik
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Mateusz Siedlinski
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| |
Collapse
|
24
|
Del Gaudio I, Sreckovic I, Zardoya-Laguardia P, Bernhart E, Christoffersen C, Frank S, Marsche G, Illanes SE, Wadsack C. Circulating cord blood HDL-S1P complex preserves the integrity of the feto-placental vasculature. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158632. [PMID: 31954174 DOI: 10.1016/j.bbalip.2020.158632] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/08/2020] [Accepted: 01/11/2020] [Indexed: 12/14/2022]
Abstract
Perinatal and long-term offspring morbidities are strongly dependent on the preservation of placental vascular homeostasis during pregnancy. In adults, the HDL-apoM-S1P complex protects the endothelium and maintains vascular integrity. However, the metabolism and biology of cord blood-derived HDLs (referred to as neonatal HDL, nHDL) strikingly differ from those in adults. Here, we investigate the role of neonatal HDLs in the regulation of placental vascular function. We show that nHDL is a major carrier of sphingosine-1-phosphate (S1P), which is anchored to the particle through apoM (rs = 0.90, p < 0.0001) in the fetal circulation. Furthermore, this complex interacts with S1P receptors on the feto-placental endothelium and activates specifically extracellular signal-regulated protein kinases 1 and 2 (ERK) and phospholipase C (PLC) downstream signaling, promotes endothelial cell proliferation and calcium flux. Notably, the nHDL-S1P complex triggers actin filaments reorganization, leading to an enhancement of placental endothelial barrier function. Additionally, nHDL induces vasorelaxation of isolated placental chorionic arteries. Taken together, these results suggest that circulating nHDL exerts vasoprotective effects on the feto-placental endothelial barrier mainly via S1P signaling.
Collapse
Affiliation(s)
- Ilaria Del Gaudio
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Ivana Sreckovic
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | | | - Eva Bernhart
- Department of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Christina Christoffersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Saša Frank
- Department of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Sebastian E Illanes
- Laboratory of Reproductive Biology, Center for Biomedical Research, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile; Department of Obstetrics and Gynecology, Clinica Davila, Santiago, Chile
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
25
|
The fucoidan from sea cucumber Apostichopus japonicus attenuates lipopolysaccharide-challenged liver injury in C57BL/6J mice. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103493] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
26
|
Isolation and Quantification of Sphingosine and Sphinganine from Rat Serum Revealed Gender Differences. Biomolecules 2019; 9:biom9090459. [PMID: 31500283 PMCID: PMC6770419 DOI: 10.3390/biom9090459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 09/04/2019] [Indexed: 12/03/2022] Open
Abstract
Sphingolipids are an important group of lipids that play crucial roles in living cells, facilitating cell recognition, signal transduction and endocytosis. The concentration of sphingosine and some of its derivatives like sphinganine may serve as a biomarker for the diagnosis of sphingolipidoses or be used for further research into similar diseases. In this study, a sphingolipid extraction and a high resolution detection method specific for sphingosine and sphinganine was adapted and tested. Lipids were extracted from rats’ serum, coupled to o-phthalaldehyde and detected with a fluorescence detector after running through a silica gel column in a high performance liquid chromatography system. With this method, we analysed 20 male and 20 female rat serum samples and compared the concentrations of sphingosine and sphinganine. The results showed a significant difference between the sphingosine concentrations in the male and female rats. The sphingosine concentration in female rats was 805 ng/mL (standard deviation, SD ± 549), while that in males was significantly lower at (75 ng/mL (SD ± 40)). Furthermore, the sphingosine:sphinganine ratio was almost 15-fold higher in the females’ samples. The method presented here facilitates the accurate quantification of sphingosine and sphinganine concentrations down to 2.6 ng and 3.0 ng, respectively, and their ratio in small amounts of rat serum samples to study the sphingolipid metabolism and its potential modulation due to gene mutations or the effect of prevalent toxins.
Collapse
|
27
|
Yang Z, Yin J, Wang Y, Wang J, Xia B, Li T, Yang X, Hu S, Ji C, Guo S. The fucoidan A3 from the seaweed Ascophyllum nodosum enhances RCT-related genes expression in hyperlipidemic C57BL/6J mice. Int J Biol Macromol 2019; 134:759-769. [PMID: 31100394 DOI: 10.1016/j.ijbiomac.2019.05.070] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/03/2019] [Accepted: 05/11/2019] [Indexed: 12/13/2022]
Abstract
Reverse cholesterol transport (RCT) has been demonstrated to reduce hyperlipidemia, and fucoidans are found to possess hypolipidemic effect. This study was designed to investigate the lipid-lowering effect of the fucoidan from the brown seaweed A. nodosum and whether it improves RCT-related genes expression in C57 BL/6J mice. Our results indicated that fucoidan A3 (100 mg/kg/day) intervention significantly reduced plasma total cholesterol (~23.2%), triglyceride (~48.7%) and fat pad index. This fucoidan significantly increased the mRNA expression of low-density lipoprotein receptor (LDLR), scavenger receptor B type 1 (SR-B1), cholesterol 7 alpha-hydroxylase A1 (CYP7A1), liver X receptor (LXR) β, ATP-binding cassette transporter (ABC) A1 and sterol regulatory element-binding protein (SREBP) 1c, and decreased the expression of peroxisome proliferator-activated receptor (PPAR) γ, however, it had no effect on the expression of proprotein convertase subtilisin/kexin type 9, PPARα, LXRα, SREBP-2, ABCG1, ABCG8 and Niemann-Pick C1-like 1. These results demonstrated that this fucoidan improved lipid transfer from plasma to the liver by activating SR-B1 and LDLR, and up-regulated lipid metabolism by activating LXRβ, ABCA1 and CYP7A1. In conclusion, this fucoidan lowers lipid by enhancing RCT-related genes expression, and it can be explored as a potential candidate for prevention or treatment of lipid disorders.
Collapse
Affiliation(s)
- Zixun Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China; College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China
| | - Jiayu Yin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China; College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China
| | - Yufeng Wang
- Nanjing Well Pharmaceutical Co., Ltd., Nanjing 210042, China
| | - Jin Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Bin Xia
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China; College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China
| | - Ting Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China; College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China
| | - Xiaoqian Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China; College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China
| | - Shumei Hu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Chenfeng Ji
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China.
| | - Shoudong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
28
|
Yin J, Wang J, Li F, Yang Z, Yang X, Sun W, Xia B, Li T, Song W, Guo S. The fucoidan from the brown seaweed Ascophyllum nodosum ameliorates atherosclerosis in apolipoprotein E-deficient mice. Food Funct 2019; 10:5124-5139. [PMID: 31364648 DOI: 10.1039/c9fo00619b] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Hyperlipidemia is a major cause of atherosclerosis. Reverse cholesterol transport (RCT) is believed to attenuate hyperlipidemia and the progression of atherosclerosis. Although fucoidans are reported to have hypolipidemic effects, the underlying mechanisms are unclear. Furthermore, few reports have revealed the anti-atherosclerotic effects and the underlying mechanisms of fucoidans. This study was designed to investigate the anti-atherosclerotic effect and mechanisms of the fucoidan from seaweed A. nodosum. Our results demonstrated that the fucoidan administration ameliorated atherosclerotic lesion and lipid profiles in a dose-dependent manner in the apolipoprotein E-deficient (apoE-/-) mice fed a high-fat diet. In the apoE-/- mice liver, the fucoidan treatment significantly increased the expression of scavenger receptor B type 1 (SR-B1), peroxisome proliferator-activated receptor (PPAR) α and β, liver X receptor (LXR) α, ATP-binding cassette transporter (ABC) A1 and ABCG8; and markedly decreased the expression of PPARγ and sterol regulatory element-binding protein (SREBP) 1c, but not low-density lipoprotein receptor, proprotein convertase subtilisin/kexin type 9, cholesterol 7 alpha-hydroxylase A1, LXRβ and ABCG1. In the small intestine of the apoE-/- mice, the fucoidan treatment significantly reduced the expression of Niemann-Pick C1-like 1 (NPC1L1) and dramatically improved ABCG8 levels. These results demonstrated for the first time that the fucoidan from A. nodosum attenuated atherosclerosis by regulating RCT-related genes and proteins expression in apoE-/- mice. In summary, this fucoidan from A. nodosum may be explored as a potential compound for prevention or treatment of hyperlipidemia-induced atherosclerosis.
Collapse
Affiliation(s)
- Jiayu Yin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yang Z, Liu G, Wang Y, Yin J, Wang J, Xia B, Li T, Yang X, Hou P, Hu S, Song W, Guo S. Fucoidan A2 from the Brown Seaweed Ascophyllum nodosum Lowers Lipid by Improving Reverse Cholesterol Transport in C57BL/6J Mice Fed a High-Fat Diet. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:5782-5791. [PMID: 31055921 DOI: 10.1021/acs.jafc.9b01321] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Reverse cholesterol transport (RCT) is a physiological process, in which excess peripheral cholesterol is transported to the liver and further excreted into the bile and then feces. Recently, fucoidans are reported to have a lipid-lowering effect. This study was designed to investigate whether fucoidan from the brown seaweed Ascophyllum nodosum lowers lipid by modulating RCT in C57BL/6J mice fed a high-fat diet. Our results indicated that fucoidan intervention significantly reduced plasma triglyceride, total cholesterol, and fat pad index and markedly increased high-density lipoprotein cholesterol in a dose-dependent manner. In the liver, fucoidan significantly increased the expression of peroxisome proliferator-activated receptor (PPAR)α, PPARγ, liver X receptor (LXR)β, adenosine triphosphate (ATP) binding cassette (ABC)A1, ABCG8, low-density lipoprotein receptor (LDLR), scavenger receptor B type 1 (SR-B1), and cholesterol 7-α-hydroxylase A1 (CYP7A1) and decreased the triglyceride level and expression of proprotein convertase subtilisin/kexin type 9 (PCSK9) and PPARβ but had no effect on LXRα, ABCG1, and ABCG5. In the small intestine, the fucoidan treatment significantly reduced the expression of Niemann-Pick C1-like 1 (NPC1L1) and improved ABCG5 and ABCG8. These results demonstrated that fucoidan can improve lipid transfer from plasma to the liver by activating SR-B1 and LDLR and inactivating PCSK9 and upregulate lipid metabolism by activating PPARα, LXRβ, ABC transporters, and CYP7A1. In the small intestine, this fucoidan can decrease cholesterol absorption and increase cholesterol excretion by activating NPC1L1 and ABCG5 and ABCG8, respectively. In conclusion, fucoidan from A. nodosum may lower lipids by modulating RCT-related protein expression and can be explored as a potential compound for prevention or treatment of hyperlipidemia-related diseases.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- Animals
- Ascophyllum/chemistry
- Biological Transport/drug effects
- Cholesterol/metabolism
- Cholesterol 7-alpha-Hydroxylase/genetics
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Diet, High-Fat/adverse effects
- Humans
- Hyperlipidemias/drug therapy
- Hyperlipidemias/etiology
- Hyperlipidemias/genetics
- Hyperlipidemias/metabolism
- Hypolipidemic Agents/administration & dosage
- Lipid Metabolism/drug effects
- Liver X Receptors/genetics
- Liver X Receptors/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Plant Extracts/administration & dosage
- Polysaccharides/administration & dosage
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Receptors, Scavenger/genetics
- Receptors, Scavenger/metabolism
- Seaweed/chemistry
Collapse
Affiliation(s)
- Zixun Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Guanjun Liu
- Weihai Municipal Hospital , Weihai , Shandong 264200 , People's Republic of China
| | - Yufeng Wang
- Nanjing Well Pharmaceutical Company, Limited Nanjing , Jiangsu 210042 , People's Republic of China
| | - Jiayu Yin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Jin Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Bin Xia
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Ting Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Xiaoqian Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Pengbo Hou
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Shumei Hu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Weiguo Song
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| | - Shoudong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy , Weifang Medical University , Weifang , Shandong 261053 , People's Republic of China
| |
Collapse
|
30
|
Hou P, Hu S, Wang J, Yang Z, Yin J, Zhou G, Guo S. Exogenous supplement of N-acetylneuraminic acid improves macrophage reverse cholesterol transport in apolipoprotein E-deficient mice. Lipids Health Dis 2019; 18:24. [PMID: 30678697 PMCID: PMC6346517 DOI: 10.1186/s12944-019-0971-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/08/2019] [Indexed: 12/30/2022] Open
Abstract
Background N-acetylneuraminic acid (NANA) is the major form of sialic acid in mammals, and the plasma NANA level is increased in patients with cardiovascular diseases. Exogenous supplement of NANA has been demonstrated to reduce hyperlipidaemia and the formation of atherosclerotic lesions; however, the underlying mechanisms have not yet been clarified. The aim of this study is to investigate whether exogenous supplement of NANA improves reverse cholesterol transprot (RCT) in vivo. Methods Apolipoprotein E-deficient mice fed a high-fat diet were used to investigate the effect of NANA on RCT by [3H]-cholesterol-loaded macrophages, and the underlying mechanism was further investigated by various molecular techniques using fenofibrate as a positive control. Results Our novel results demonstrated that exogenous supplement of NANA significantly improved [3H]-cholesterol transfer from [3H]-cholesterol-loaded macrophages to the plasma (an increase of > 42.9%), liver (an increase of 35.8%), and finally to the feces (an increase of 50.4% from 0 to 24 h) for excretion in apolipoprotein E-deficient mice fed a high-fat diet. In addition, NANA up regulated the protein expression of ATP-binding cassette (ABC) G1 and peroxisome proliferator-activated receptor α (PPARα), but not the protein expression of ABCA1and scavenger receptor B type 1 in the liver. Therefore, the underlying mechanism of NANA in improving RCT may be partially due to the elevated protein levels of PPARα and ABCG1. Conclusion Exogenous supplement of NANA improves RCT in apolipoprotein E-deficient mice fed a high-fat diet mainly by improving the protein expression of PPARα and ABCG1. These results are helpful in explaining the lipid-lowering effect of NANA.
Collapse
Affiliation(s)
- Pengbo Hou
- Research Center on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin, 150076, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Shumei Hu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Jin Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Zixun Yang
- Research Center on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin, 150076, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Jiayu Yin
- Research Center on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin, 150076, China.,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | | | - Shoudong Guo
- Research Center on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin, 150076, China. .,Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China. .,Taishan Medical University, Taian, 271000, China.
| |
Collapse
|
31
|
Yafasova A, Mandrup CM, Egelund J, Nyberg M, Stallknecht B, Hellsten Y, Nielsen LB, Christoffersen C. Effect of menopause and exercise training on plasma apolipoprotein M and sphingosine-1-phosphate. J Appl Physiol (1985) 2019; 126:214-220. [DOI: 10.1152/japplphysiol.00527.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The axis of apolipoprotein M (apoM) and sphingosine-1-phosphate (S1P) is of importance to plasma lipid levels, endothelial function, and development of atherosclerosis. Menopause is accompanied by dyslipidemia and an increased risk of atherosclerosis, which can be lowered by exercise training. The aim of this study was to explore if effects of menopause and training are paralleled by changes in the apoM/S1P axis. Healthy, late premenopausal [ n = 38, age 49.2 (SD 2)] and recent postmenopausal [ n = 37, age 53.3 (SD 3)] women from the Copenhagen Women Study participated in a 3-mo, aerobic high-intensity exercise intervention. Before training, plasma apoM was higher in postmenopausal [1.08 µmol/l (SD 0.2)] compared with premenopausal [0.82 µmol/l (SD 0.2)] women ( P < 0.0001). Plasma S1P was similar in the two groups [0.44 µmol/l (SD 0.1) and 0.46 µmol/l (SD 0.1), respectively]. Thus, the pretraining S1P/apoM ratio was 26% lower in postmenopausal than premenopausal women ( P < 0.0001). After the training program, plasma apoM increased from 0.82 µmol/l (SD 0.2) to 0.90 µmol/l (SD 0.3) in premenopausal women and from 1.08 µmol/l (SD 0.2) to 1.16 µmol/l (SD 0.3) in postmenopausal women ( P < 0.05). Plasma S1P increased from 0.44 µmol/l (SD 0.1) to 0.47 µmol/l (SD 0.1) in premenopausal women and from 0.46 µmol/l (SD 0.1) to 0.48 µmol/l (SD 0.1) in postmenopausal women ( P < 0.05). The results suggest that menopause is accompanied by higher plasma apoM but not S1P concentrations and that exercise training increases plasma apoM and S1P in healthy middle-aged women irrespective of menopausal status. NEW & NOTEWORTHY The apolipoprotein M/sphingosine-1-phosphate (apoM/S1P) complex is involved in maintaining a healthy endothelial barrier function. Our study is the first, to our knowledge, to show how menopause affects the apoM/S1P axis. The results suggest that menopause is accompanied by higher plasma apoM but not S1P concentrations. Second, to our knowledge the study is also the first to show that exercise training increases both apoM/S1P in women irrespective of menopausal status.
Collapse
Affiliation(s)
- Adelina Yafasova
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Camilla M. Mandrup
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jon Egelund
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Michael Nyberg
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Bente Stallknecht
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Lars B. Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg Hospital, Copenhagen, Denmark
| |
Collapse
|
32
|
Biological function of SPNS2: From zebrafish to human. Mol Immunol 2018; 103:55-62. [DOI: 10.1016/j.molimm.2018.08.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 01/01/2023]
|
33
|
Sukocheva OA. Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming. Int J Mol Sci 2018; 19:420. [PMID: 29385066 PMCID: PMC5855642 DOI: 10.3390/ijms19020420] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 02/05/2023] Open
Abstract
Sphingolipids, sphingolipid metabolizing enzymes, and their receptors network are being recognized as part of the signaling mechanisms, which govern breast cancer cell growth, migration, and survival during chemotherapy treatment. Approximately 70% of breast cancers are estrogen receptor (ER) positive and, thus, rely on estrogen signaling. Estrogen activates an intracellular network composed of many cytoplasmic and nuclear mediators. Some estrogen effects can be mediated by sphingolipids. Estrogen activates sphingosine kinase 1 (SphK1) and amplifies the intracellular concentration of sphingosine-1-phosphate (S1P) in breast cancer cells during stimulation of proliferation and survival. Specifically, Estrogen activates S1P receptors (S1PR) and induces growth factor receptor transactivation. SphK, S1P, and S1PR expression are causally associated with endocrine resistance and progression to advanced tumor stages in ER-positive breast cancers in vivo. Recently, the network of SphK/S1PR was shown to promote the development of ER-negative cancers and breast cancer stem cells, as well as stimulating angiogenesis. Novel findings confirm and broaden our knowledge about the cross-talk between sphingolipids and estrogen network in normal and malignant cells. Current S1PRs therapeutic inhibition was indicated as a promising chemotherapy approach in non-responsive and advanced malignancies. Considering that sphingolipid signaling has a prominent role in terminally differentiated cells, the impact should be considered when designing specific SphK/S1PR inhibitors. This study analyzes the dynamic of the transformation of sphingolipid axis during a transition from normal to pathological condition on the level of the whole organism. The sphingolipid-based mediation and facilitation of global effects of estrogen were critically accented as a bridging mechanism that should be explored in cancer prevention.
Collapse
Affiliation(s)
- Olga A Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| |
Collapse
|
34
|
Yin J, Guo YM, Chen P, Xiao H, Wang XH, DiSanto ME, Zhang XH. Testosterone regulates the expression and functional activity of sphingosine-1-phosphate receptors in the rat corpus cavernosum. J Cell Mol Med 2017; 22:1507-1516. [PMID: 29266713 PMCID: PMC5824404 DOI: 10.1111/jcmm.13416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 09/12/2017] [Indexed: 01/29/2023] Open
Abstract
The bioactive lipid sphingosine‐1‐phosphate (S1P) regulates smooth muscle (SM) contractility predominantly via three G protein‐coupled receptors. The S1P1 receptor is associated with nitric oxide (NO)‐mediated SM relaxation, while S1P2 & S1P3 receptors are linked to SM contraction via activation of the Rho‐kinase pathway. This study is to determine testosterone (T) modulating the expression and functional activity of S1P receptors in corpus cavernosum (CC). Adult male Sprague‐Dawley rats were randomly divided into three groups: sham‐operated controls, surgical castration and T supplemented group. Serum S1P levels were detected by high‐performance liquid chromatography. The expression of S1P1‐3 receptors and sphingosine kinases was detected by real‐time RT‐PCR. In vitro organ bath contractility and in vivo intracavernous pressure (ICP) measurement were also performed. T deprivation significantly decreased ICP rise. Meanwhile, surgical castration induced a significant increase in serum S1P level and the expression of S1P2‐3 receptors by twofold (P < 0.05) but a decrease in the expression of S1P1 receptor. Castration also augmented exogenous phenylephrine (PE), S1P, S1P1,3 receptor agonist FTY720‐P contractility and S1P2‐specific antagonist JTE013 relaxation effect. T supplemented could restore the aforementioned changes. We provide novel data that castration increased serum S1P concentration and up‐regulated the expression of S1P2‐3 receptors in CC. Consistently, agonizing S1P receptors induced CCSM contraction and antagonizing mediated relaxation were augmented. This provides the first clear evidence that S1P system dysregulation may contribute to hypogonadism‐related erectile dysfunction (ED), and S1P receptors may be expected as a potential target for treating ED.
Collapse
Affiliation(s)
- Jing Yin
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu-Ming Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - He Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xing-Huan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Michael E DiSanto
- Surgery and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Xin-Hua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
35
|
Yin J, Guo YM, Chen P, Xiao H, Wang XH, DiSanto ME, Zhang XH. Testosterone regulates the expression and functional activity of sphingosine-1-phosphate receptors in the rat corpus cavernosum. J Cell Mol Med 2017. [DOI: 10.1111/jcmm.13416 29266713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Jing Yin
- Department of Rehabilitation; Zhongnan Hospital of Wuhan University; Wuhan China
| | - Yu-ming Guo
- Department of Urology; Zhongnan Hospital of Wuhan University; Wuhan China
| | - Ping Chen
- Department of Urology; Zhongnan Hospital of Wuhan University; Wuhan China
| | - He Xiao
- Department of Urology; Zhongnan Hospital of Wuhan University; Wuhan China
| | - Xing-huan Wang
- Department of Urology; Zhongnan Hospital of Wuhan University; Wuhan China
| | - Michael E DiSanto
- Surgery and Biomedical Sciences; Cooper Medical School of Rowan University; Camden NJ USA
| | - Xin-hua Zhang
- Department of Urology; Zhongnan Hospital of Wuhan University; Wuhan China
| |
Collapse
|
36
|
Ramanathan R, Raza A, Sturgill J, Lyon D, Young J, Hait NC, Takabe K. Paradoxical Association of Postoperative Plasma Sphingosine-1-Phosphate with Breast Cancer Aggressiveness and Chemotherapy. Mediators Inflamm 2017; 2017:5984819. [PMID: 29147072 PMCID: PMC5632905 DOI: 10.1155/2017/5984819] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/24/2017] [Accepted: 08/08/2017] [Indexed: 12/11/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator that has been shown to serve an important regulatory function in breast cancer progression. This study analyzes plasma S1P levels in breast cancer patients undergoing adjuvant therapy as compared to healthy control volunteers. 452 plasma S1P samples among 158 breast cancer patients, along with 20 healthy control volunteers, were analyzed. Mean S1P levels did not significantly differ between cancer patients and controls. Smoking was associated with higher S1P levels in cancer patients. Baseline S1P levels had weak inverse correlation with levels of the inflammatory mediator interleukin- (IL-) 17 and CCL-2 and positive correlation with tumor necrosis factor alpha (TNF-α). Midpoint S1P levels during adjuvant therapy were lower than baseline, with near return to baseline after completion, indicating a relationship between chemotherapy and circulating S1P. While stage of disease did not correlate with plasma S1P levels, they were lower among patients with Her2-enriched and triple-negative breast cancer as compared to luminal-type breast cancer. Plasma S1P levels are paradoxically suppressed in aggressive breast cancer and during adjuvant chemotherapy, which raises the possibility that postoperative plasma S1P levels do not reflect S1P secretion from resected breast cancer.
Collapse
Affiliation(s)
- Rajesh Ramanathan
- Department of Surgery, Virginia Commonwealth University Medical Center, 1200 E. Broad St., Richmond, VA, USA
| | - Ali Raza
- Lincoln Medical and Mental Health Center, Cancer Center, Room 9-69, Bronx, NY, USA
| | - Jamie Sturgill
- Department of Family and Community Health Nursing, Virginia Commonwealth University, 1100 E. Leigh St., Richmond, VA, USA
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Debra Lyon
- University of Florida, College of Nursing, Gainesville, FL, USA
| | - Jessica Young
- Breast Surgery, Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Nitai C. Hait
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
- Breast Surgery, Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Kazuaki Takabe
- Department of Surgery, Virginia Commonwealth University Medical Center, 1200 E. Broad St., Richmond, VA, USA
- Breast Surgery, Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
37
|
Cui Y, Hou P, Li F, Liu Q, Qin S, Zhou G, Xu X, Si Y, Guo S. Quercetin improves macrophage reverse cholesterol transport in apolipoprotein E-deficient mice fed a high-fat diet. Lipids Health Dis 2017; 16:9. [PMID: 28088205 PMCID: PMC5237507 DOI: 10.1186/s12944-016-0393-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/16/2016] [Indexed: 01/20/2023] Open
Abstract
Background Quercetin, one of the most widely distributed flavonoids in plants, has been demonstrated to reduce hyperlipidaemia and atherosclerotic lesion formation. Reverse cholesterol transport (RCT) plays a crucial role in exporting cholesterol from peripheral cells, which is one mechanism utilized in the prevention and treatment of atherosclerosis. The aim of this study is to investigate whether quercetin reduces lipid accumulation by improving RCT in vivo. Methods Apolipoprotein E-deficient mice fed a high-fat diet were used to investigate the effect of quercetin on RCT by an isotope tracing method, and the underlying mechanisms were clarified by molecular techniques. Results These novel results demonstrated that quercetin significantly improved [3H]-cholesterol transfer from [3H]-cholesterol-loaded macrophages to the plasma (approximately 34% increase), liver (30% increase), and bile (50% increase) and finally to the feces (approximately 40% increase) for excretion in apolipoprotein E-deficient mice fed a high-fat diet. Furthermore, quercetin markedly increased the cholesterol accepting ability of plasma and high-density lipoprotein (HDL) and dramatically decreased the content of malondialdehyde in plasma and oxidized phosphocholine carried by HDL. Therefore, the underlying mechanisms of quercetin in improving RCT may be partially due to the elevated cholesterol accepting ability of HDL, the increased expression levels of proteins related to RCT, such as ATP-binding cassettes (ABC) A1 and G1, and the improved antioxidant activity of HDL. Conclusion Quercetin accelerates RCT in an atherosclerosis model, which is helpful in clarifying the lipid-lowering effect of quercetin.
Collapse
Affiliation(s)
- Yingjie Cui
- Key Laboratory of Atherosclerosis in Universities of Shandong Province, Institute of Atherosclerosis, Taishan Medical University, 2# Yingsheng East Road, Taian, Shandong Province, 271000, China
| | - Pengbo Hou
- Key Laboratory of Atherosclerosis in Universities of Shandong Province, Institute of Atherosclerosis, Taishan Medical University, 2# Yingsheng East Road, Taian, Shandong Province, 271000, China.,Research Center on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin, 150076, China
| | - Fahui Li
- Department of Chemistry and Chemical Engineering, Weifang University, Weifang, 261061, China
| | - Qinghua Liu
- Affiliated Hospital of Taishan Medical University, Taian, 271000, China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong Province, Institute of Atherosclerosis, Taishan Medical University, 2# Yingsheng East Road, Taian, Shandong Province, 271000, China.
| | - Guanghai Zhou
- Key Laboratory of Atherosclerosis in Universities of Shandong Province, Institute of Atherosclerosis, Taishan Medical University, 2# Yingsheng East Road, Taian, Shandong Province, 271000, China
| | - Xuelian Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yanhong Si
- Key Laboratory of Atherosclerosis in Universities of Shandong Province, Institute of Atherosclerosis, Taishan Medical University, 2# Yingsheng East Road, Taian, Shandong Province, 271000, China
| | - Shoudong Guo
- Key Laboratory of Atherosclerosis in Universities of Shandong Province, Institute of Atherosclerosis, Taishan Medical University, 2# Yingsheng East Road, Taian, Shandong Province, 271000, China.
| |
Collapse
|
38
|
Tian H, Liu Q, Qin S, Zong C, Zhang Y, Yao S, Yang N, Guan T, Guo S. Synthesis and cardiovascular protective effects of quercetin 7-O-sialic acid. J Cell Mol Med 2017; 21:107-120. [PMID: 27511707 PMCID: PMC5192943 DOI: 10.1111/jcmm.12943] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 07/04/2016] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress and inflammation play important roles in the pathogenesis of cardiovascular disease (CVD). Oxidative stress-induced desialylation is considered to be a primary step in atherogenic modification, and therefore, the attenuation of oxidative stress and/or inflammatory reactions may ameliorate CVD. In this study, quercetin 7-O-sialic acid (QA) was synthesized aiming to put together the cardiovascular protective effect of quercetin and the recently reported anti-oxidant and anti-atherosclerosis functions of N-acetylneuraminic acid. The biological efficacy of QA was evaluated in vitro in various cellular models. The results demonstrated that 50 μM QA could effectively protect human umbilical vein endothelial cells (HUVEC, EA.hy926) against hydrogen peroxide- or oxidized low-density lipoprotein-induced oxidative damage by reducing the production of reactive oxygen species. QA attenuated hydrogen peroxide-induced desialylation of HUVEC and lipoproteins. QA decreased lipopolysaccharide-induced secretion of tumour necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1), and it significantly reduced the expression of intercellular adhesion molecule-1, vascular cell adhesion molecule-1, TNF-α and MCP-1. Furthermore, QA effectively promoted cholesterol efflux from Raw 264.7 macrophages to apolipoprotein A-1 and high-density lipoprotein by up-regulating ATP-binding cassette transporter A1 and G1, respectively. Results indicated that the novel compound QA exhibited a better capacity than quercetin for anti-oxidation, anti-inflammation, cholesterol efflux promotion and biomolecule protection against desialylation and therefore could be a candidate compound for the prevention or treatment of CVD.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Qingchao Liu
- Department of Pharmaceutical EngineeringNorthwest UniversityXi'anChina
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Chuanlong Zong
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Ying Zhang
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Nana Yang
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Tao Guan
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Shoudong Guo
- Key Laboratory of Atherosclerosis in Universities of Shandong ProvinceInstitute of AtherosclerosisTaishan Medical UniversityTaianChina
| |
Collapse
|
39
|
Duarte AC, Hrynchak MV, Gonçalves I, Quintela T, Santos CRA. Sex Hormone Decline and Amyloid β Synthesis, Transport and Clearance in the Brain. J Neuroendocrinol 2016; 28. [PMID: 27632792 DOI: 10.1111/jne.12432] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Sex hormones (SH) are essential regulators of the central nervous system. The decline in SH levels along with ageing may contribute to compromised neuroprotection and set the grounds for neurodegeneration and cognitive impairments. In Alzheimer's disease, besides other pathological features, there is an imbalance between amyloid β (Aβ) production and clearance, leading to its accumulation in the brain of older subjects. Aβ accumulation is a primary cause for brain inflammation and degeneration, as well as concomitant cognitive decline. There is mounting evidence that SH modulate Aβ production, transport and clearance. Importantly, SH regulate most of the molecules involved in the amyloidogenic pathway, their transport across brain barriers for elimination, and their degradation in the brain interstitial fluid. This review brings together data on the regulation of Aβ production, metabolism, degradation and clearance by SH.
Collapse
Affiliation(s)
- A C Duarte
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - M V Hrynchak
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - I Gonçalves
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - T Quintela
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - C R A Santos
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
40
|
Guo S, Tian H, Dong R, Yang N, Zhang Y, Yao S, Li Y, Zhou Y, Si Y, Qin S. Exogenous supplement of N-acetylneuraminic acid ameliorates atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis 2016; 251:183-191. [DOI: 10.1016/j.atherosclerosis.2016.05.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/13/2016] [Accepted: 05/18/2016] [Indexed: 12/14/2022]
|
41
|
Huang C, Yuan P, Wu J, Huang J. Estrogen regulates excitatory amino acid carrier 1 (EAAC1) expression through sphingosine kinase 1 (SphK1) transacting FGFR-mediated ERK signaling in rat C6 astroglial cells. Neuroscience 2016; 319:9-22. [PMID: 26804240 DOI: 10.1016/j.neuroscience.2016.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/04/2016] [Accepted: 01/12/2016] [Indexed: 12/28/2022]
Abstract
Excitatory amino acid carrier 1 (EAAC1) is one important subtype of the excitatory amino acid transporters (EAATs), and its absence can increase the vulnerability to oxidative stress in neural tissue. Enhanced expression of EAAC1 can provide neuroprotection in multiple disorders, including ischemia and multiple sclerosis. However, the mechanism regulating EAAC1 expression is not fully understood. Using rat C6 astroglial cells, which specifically express EAAC1, we found that 17β-estradiol (E2) and (±)-1-[(3aR(∗),4S(∗),9bS(∗))-4-(6-bromo-1,3-benzodioxol-5-yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone (G1), an agonist of the G-protein-coupled estrogen receptor (GPR30), strongly increased EAAC1 protein levels and protected cells from hydrogen peroxide (H2O2) toxicity. We further found that E2/G1 activated sphingosine kinase 1 (SphK1) via GPR30, resulting in the transcription of fibroblast growth factor 2 (FGF2), which stimulated its receptor (FGFR) and led to the phosphorylation of FGFR substrate 2α (FRS2α). This triggered downstream ERK1/2 signaling for the expression of EAAC1. Both the knockdown of FGF2 by siRNA and the pharmacological suppression of the FGFR-ERK cascade abolished the E2/G1 effect on EAAC1 expression. Overall, our work characterizes a signaling pathway by which E2 transactivates FGFR-ERK to induce EAAC1 expression in an FGF2-dependent manner. This occurs through SphK1 activation via GPR30 and leads to a resistance to H2O2 toxicity. This signal transduction pathway may provide novel insights into our understanding of the neuroprotective effects of E2 and may reveal new therapeutic targets or drugs for regulating the oxidative toxicity effects of various neurological diseases.
Collapse
Affiliation(s)
- C Huang
- College of Life Science, Wuhan University, Wuhan 430072, PR China
| | - P Yuan
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - J Wu
- College of Life Science, Wuhan University, Wuhan 430072, PR China
| | - J Huang
- College of Life Science, Wuhan University, Wuhan 430072, PR China.
| |
Collapse
|
42
|
Sukocheva O, Wadham C, Gamble J, Xia P. Sphingosine-1-phosphate receptor 1 transmits estrogens' effects in endothelial cells. Steroids 2015; 104:237-245. [PMID: 26476183 DOI: 10.1016/j.steroids.2015.10.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 09/29/2015] [Accepted: 10/11/2015] [Indexed: 02/08/2023]
Abstract
We have previously reported that the steroid hormone estrogens stimulate activation of sphingosine kinase 1 (SphK1) and sphingosine-1-phosphate (S1P) receptors in breast cancer cells. Both estrogens and S1P are potent biological modulators of endothelial function in vasculature able to activate multiple effectors, including endothelial nitric oxide synthase (eNOS). In this study we report that treatment of endothelial cells (ECs) with 17β-estradiol (E2) resulted in a rapid, transient, and dose-dependent increase in SphK activity and increased S1P production. The effect was not reproduced by the inactive E2 analogue 17α-E2. Expression of the dominant-negative mutant SphK1(G82D) or transfection of SphK1-targeted siRNA in ECs caused not only a defect in SphK activation by E2, but also a significant inhibition of E2-induced activation of Akt/eNOS. Furthermore, E2 treatment induced internalization of plasma membrane S1P1 receptor, accompanied with an increase in the amount of cytosolic S1P1. By down-regulating S1P1 receptor expression, the S1P1-specific antisense oligonucleotides significantly inhibited E2-induced activation of Akt/eNOS in ECs. E2-induced EC migration and tube formation were also inhibited by S1P1 down-regulation. Thus, the findings indicate an important role of the SphK1/S1P1 pathway in mediating estrogen signaling and its actions in vasculature.
Collapse
Affiliation(s)
- Olga Sukocheva
- School of Health Sciences, Flinders University, SA, Australia.
| | | | | | - Pu Xia
- Department of Endocrinology, Zhongsan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
43
|
Książek M, Chacińska M, Chabowski A, Baranowski M. Sources, metabolism, and regulation of circulating sphingosine-1-phosphate. J Lipid Res 2015; 56:1271-81. [PMID: 26014962 PMCID: PMC4479332 DOI: 10.1194/jlr.r059543] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/12/2015] [Indexed: 12/16/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that acts either as an intracellular messenger or as a ligand for its membrane receptors. S1P is a normal constituent of blood, where it is found both in plasma and blood cells. Compared with other cell types, sphingolipid metabolism in erythrocytes and platelets has unique features that allow the erythrocytes and platelets to accumulate S1P. In plasma, S1P is bound mainly to HDLs and albumin. Of note, metabolism and biological activity of S1P is to a large extent affected by the type of its carrier. Plasma S1P is characterized by a short half-life, indicating rapid clearance by degradative enzymes and the presence of high-capacity sources involved in maintaining its high concentration. These sources include blood cells, vascular endothelium, and hepatocytes. However, the extent to which each of these contributes to the plasma pool of S1P is a matter of debate. Circulating S1P plays a significant physiological role. It was found to be the key regulator of lymphocyte trafficking, endothelial barrier function, and vascular tone. The purpose of this review is to summarize the present state of knowledge on the metabolism, transport, and origin of plasma S1P, and to discuss the mechanisms regulating its homeostasis in blood.
Collapse
Affiliation(s)
- Monika Książek
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Marta Chacińska
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Marcin Baranowski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
44
|
Rhee SH, Zhang P, Hunter K, Mama ST, Caraballo R, Holzberg AS, Seftel RH, Seftel AD, Echols KT, DiSanto ME. Pelvic organ prolapse is associated with alteration of sphingosine-1-phosphate/Rho-kinase signalling pathway in human vaginal wall. J OBSTET GYNAECOL 2015; 35:726-32. [DOI: 10.3109/01443615.2015.1004527] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|