1
|
Ng M, Gao AS, Phu TA, Vu NK, Raffai RL. M2 Macrophage Exosomes Reverse Cardiac Functional Decline in Mice with Diet-Induced Myocardial Infarction by Suppressing Type 1 Interferon Signaling in Myeloid Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612924. [PMID: 39345592 PMCID: PMC11429744 DOI: 10.1101/2024.09.13.612924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Effective treatment strategies to alleviate heart failure that develops as a consequence of myocardial infarction (MI) remain an unmet need in cardiovascular medicine. In this study, we uncovered that exosomes produced by human THP-1 macrophages cultured with the cytokine IL-4 (THP1-IL4-exo), reverse cardiac functional decline in mice that develop MI as a consequence of diet-induced occlusive coronary atherosclerosis. Therapeutic benefits of THP1-IL4-exo stem from their ability to reprogram circulating Ly-6Chi monocytes into an M2-like phenotype and suppress Type 1 Interferon signaling in myeloid cells within the bone marrow, the circulation, and cardiac tissue. Collectively, these benefits suppress myelopoiesis, myeloid cell recruitment to cardiac tissue, and preserve populations of resident cardiac macrophages that together mitigate cardiac inflammation, adverse ventricular remodeling, and heart failure. Our findings introduce THP1-IL4-exo, one form of M2-macrophage exosomes, as novel therapeutics to preserve cardiac function subsequent to MI.
Collapse
Affiliation(s)
- Martin Ng
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Alex S Gao
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Tuan Anh Phu
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Ngan K Vu
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Robert L Raffai
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
2
|
Magalhães DM, Stewart NA, Mampay M, Rolle SO, Hall CM, Moeendarbary E, Flint MS, Sebastião AM, Valente CA, Dymond MK, Sheridan GK. The sphingosine 1-phosphate analogue, FTY720, modulates the lipidomic signature of the mouse hippocampus. J Neurochem 2024; 168:1113-1142. [PMID: 38339785 DOI: 10.1111/jnc.16073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
The small-molecule drug, FTY720 (fingolimod), is a synthetic sphingosine 1-phosphate (S1P) analogue currently used to treat relapsing-remitting multiple sclerosis in both adults and children. FTY720 can cross the blood-brain barrier (BBB) and, over time, accumulate in lipid-rich areas of the central nervous system (CNS) by incorporating into phospholipid membranes. FTY720 has been shown to enhance cell membrane fluidity, which can modulate the functions of glial cells and neuronal populations involved in regulating behaviour. Moreover, direct modulation of S1P receptor-mediated lipid signalling by FTY720 can impact homeostatic CNS physiology, including neurotransmitter release probability, the biophysical properties of synaptic membranes, ion channel and transmembrane receptor kinetics, and synaptic plasticity mechanisms. The aim of this study was to investigate how chronic FTY720 treatment alters the lipid composition of CNS tissue in adolescent mice at a key stage of brain maturation. We focused on the hippocampus, a brain region known to be important for learning, memory, and the processing of sensory and emotional stimuli. Using mass spectrometry-based lipidomics, we discovered that FTY720 increases the fatty acid chain length of hydroxy-phosphatidylcholine (PCOH) lipids in the mouse hippocampus. It also decreases PCOH monounsaturated fatty acids (MUFAs) and increases PCOH polyunsaturated fatty acids (PUFAs). A total of 99 lipid species were up-regulated in the mouse hippocampus following 3 weeks of oral FTY720 exposure, whereas only 3 lipid species were down-regulated. FTY720 also modulated anxiety-like behaviours in young mice but did not affect spatial learning or memory formation. Our study presents a comprehensive overview of the lipid classes and lipid species that are altered in the hippocampus following chronic FTY720 exposure and provides novel insight into cellular and molecular mechanisms that may underlie the therapeutic or adverse effects of FTY720 in the central nervous system.
Collapse
Affiliation(s)
- Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | | - Myrthe Mampay
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Sara O Rolle
- Green Templeton College, University of Oxford, Oxford, UK
| | - Chloe M Hall
- School of Applied Sciences, University of Brighton, Brighton, UK
- Department of Mechanical Engineering, University College London, London, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd, London, UK
| | - Melanie S Flint
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Marcus K Dymond
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | |
Collapse
|
3
|
Wang L, Zhang X, Ma C, Wu N. 1-Phosphate receptor agonists: A promising therapeutic avenue for ischemia-reperfusion injury management. Int Immunopharmacol 2024; 131:111835. [PMID: 38508097 DOI: 10.1016/j.intimp.2024.111835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
Ischemia-reperfusion injury (IRI) - a complex pathological condition occurring when blood supply is abruptly restored to ischemic tissues, leading to further tissue damage - poses a significant clinical challenge. Sphingosine-1-phosphate receptors (S1PRs), a specialized set of G-protein-coupled receptors comprising five subtypes (S1PR1 to S1PR5), are prominently present in various cell membranes, including those of lymphocytes, cardiac myocytes, and endothelial cells. Increasing evidence highlights the potential of targeting S1PRs for IRI therapeutic intervention. Notably, preconditioning and postconditioning strategies involving S1PR agonists like FTY720 have demonstrated efficacy in mitigating IRI. As the synthesis of a diverse array of S1PR agonists continues, with FTY720 being a prime example, the body of experimental evidence advocating for their role in IRI treatment is expanding. Despite this progress, comprehensive reviews delineating the therapeutic landscape of S1PR agonists in IRI remain limited. This review aspires to meticulously elucidate the protective roles and mechanisms of S1PR agonists in preventing and managing IRI affecting various organs, including the heart, kidney, liver, lungs, intestines, and brain, to foster novel pharmacological approaches in clinical settings.
Collapse
Affiliation(s)
- Linyuan Wang
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaowen Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Nan Wu
- The Central Laboratory of The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
4
|
Li J, Gong Y, Wang Y, Huang H, Du H, Cheng L, Ma C, Cai Y, Han H, Tao J, Li G, Cheng P. Classification of regulatory T cells and their role in myocardial ischemia-reperfusion injury. J Mol Cell Cardiol 2024; 186:94-106. [PMID: 38000204 DOI: 10.1016/j.yjmcc.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is closely related to the final infarct size in acute myocardial infarction (AMI). Therefore, reducing MIRI can effectively improve the prognosis of AMI patients. At the same time, the healing process after AMI is closely related to the local inflammatory microenvironment. Regulatory T cells (Tregs) can regulate various physiological and pathological immune inflammatory responses and play an important role in regulating the immune inflammatory response after AMI. However, different subtypes of Tregs have different effects on MIRI, and the same subtype of Tregs may also have different effects at different stages of MIRI. This article systematically reviews the classification and function of Tregs, as well as the role of various subtypes of Tregs in MIRI. A comprehensive understanding of the role of each subtype of Tregs can help design effective methods to control immune reactions, reduce MIRI, and provide new potential therapeutic options for AMI.
Collapse
Affiliation(s)
- Junlin Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Department of Cardiology, The Second People's Hospital of Neijiang, Neijiang 641100, China
| | - Yajun Gong
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yiren Wang
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Huihui Huang
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Huan Du
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lianying Cheng
- Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Cui Ma
- Department of Mathematics, Army Medical University, Chongqing 400038, China
| | - Yongxiang Cai
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Hukui Han
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jianhong Tao
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Gang Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu 610072, China.
| |
Collapse
|
5
|
Li X, Peng Z, An K, Xue M, Wang Z, Xia J, Qi Z, Shu X. Temsirolimus is a promising immunomodulatory agent for enhanced transplantation outcomes. Transpl Immunol 2023; 81:101952. [PMID: 37918580 DOI: 10.1016/j.trim.2023.101952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Identifying effective immunosuppressive strategies is critical for addressing immunological rejection following organ transplantation. This study explores the potential immunosuppressive effects and mechanisms of temsirolimus, a rapamycin derivative, in organ transplantation. METHODS A mouse cardiac allograft model was established using a cervical cannula technique with BALB/c donors and C57BL/6 recipients. Mice were administered temsirolimus intragastrically and graft survival was evaluated. Histological staining was used to assess pathological changes. The BrdU assay was used to measure splenic T cell proliferation. Flow cytometry was used to quantify regulatory T cells (Tregs), CD4+ T cells, and CD8+ T cells. ELISA and qPCR assays were used to determine Foxp3, IL-4, IFN-γ, and TGF-β expression. RESULTS Temsirolimus displayed potent immunosuppressive effects at 20 mg/kg/day, significantly inhibiting T cell proliferation (84.6%, P < 0.0001) and prolonging graft survival (median 49 days vs. 8.5 days in controls, P < 0.0001). However, median survival decreased to 34.5 days upon withdrawal. Temsirolimus also reduced splenic CD4+ and CD8+ T cells (2.85% and 2.92%, P < 0.001) and antibody levels (IgM, IgG1, IgG2) by 11.85-29.09% (P < 0.0001) and increased Tregs, Foxp3, IL-4 (P < 0.01), and TGF-β (P < 0.05), while decreasing IFN-γ (P < 0.001). CONCLUSIONS Temsirolimus exhibited potent immunosuppressive effects, emerging as a strong candidate to mitigate organ transplant rejection.
Collapse
Affiliation(s)
- Xianguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zuojie Peng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke An
- Department of Physiology, Xuzhou Medical University, Xuzhou 221009, China
| | - Mengjiao Xue
- Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenzhen Wang
- Department of Pharmacy, Zhoukou Central Hospital, Zhoukou 466000, China
| | - Junjie Xia
- Organ Transplantation Institute, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen 361100, China.
| | - Zhongquan Qi
- Medical College of Guangxi University, Guangxi University, Nanning 530004, China.
| | - Xiaogang Shu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
Staršíchová A. SR-B1-/-ApoE-R61h/h Mice Mimic Human Coronary Heart Disease. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07475-8. [PMID: 37273155 DOI: 10.1007/s10557-023-07475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2023] [Indexed: 06/06/2023]
Abstract
Cardiovascular diseases are the leading cause of death in the modern world. Atherosclerosis underlies the majority of these pathologies and may result in sudden life-threatening events such as myocardial infarction or stroke. Current concepts consider a rupture (resp. erosion) of "unstable/vulnerable" atherosclerotic plaques as a primary cause leading to thrombus formation and subsequent occlusion of the artery lumen finally triggering an acute clinical event. We and others described SR-B1-/-ApoE-R61h/h mice mimicking clinical coronary heart disease in all major aspects: from coronary atherosclerosis through vulnerable plaque ruptures leading to thrombus formation/coronary artery occlusion, finally resulting in myocardial infarction/ischemia. SR-B1-/-ApoE-R61h/h mouse provides a valuable model to study vulnerable/occlusive plaques, to evaluate bioactive compounds as well as new anti-inflammatory and "anti-rupture" drugs, and to test new technologies in experimental cardiovascular medicine. This review summarizes and discuss our knowledge about SR-B1-/-ApoE-R61h/h mouse model based on recent publications and experimental observations from the lab.
Collapse
Affiliation(s)
- Andrea Staršíchová
- Graduate School Cell Dynamics and Disease, University of Muenster, Muenster, Germany.
- European Institute for Molecular Imaging, University of Muenster, Muenster, Germany.
- Novogenia Covid GmbH, Eugendorf, Austria.
| |
Collapse
|
7
|
Soldozy S, Dalzell C, Skaff A, Ali Y, Norat P, Yagmurlu K, Park MS, Kalani MYS. Reperfusion injury in acute ischemic stroke: Tackling the irony of revascularization. Clin Neurol Neurosurg 2023; 225:107574. [PMID: 36696846 DOI: 10.1016/j.clineuro.2022.107574] [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: 04/07/2022] [Revised: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023]
Abstract
Reperfusion injury is an unfortunate consequence of restoring blood flow to tissue after a period of ischemia. This phenomenon can occur in any organ, although it has been best studied in cardiac cells. Based on cardiovascular studies, neuroprotective strategies have been developed. The molecular biology of reperfusion injury remains to be fully elucidated involving several mechanisms, however these mechanisms all converge on a similar final common pathway: blood brain barrier disruption. This results in an inflammatory cascade that ultimately leads to a loss of cerebral autoregulation and clinical worsening. In this article, the authors present an overview of these mechanisms and the current strategies being employed to minimize injury after restoration of blood flow to compromised cerebral territories.
Collapse
Affiliation(s)
- Sauson Soldozy
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA; Department of Neurosurgery, Westchester Medical Center, Valhalla, NY, USA
| | - Christina Dalzell
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Anthony Skaff
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Yusuf Ali
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Pedro Norat
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Kaan Yagmurlu
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - Min S Park
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, USA
| | - M Yashar S Kalani
- Department of Surgery, University of Oklahoma, and St. John's Neuroscience Institute, Tulsa, OK, USA.
| |
Collapse
|
8
|
Immunomodulatory drug fingolimod (FTY720) restricts the growth of opportunistic yeast Candida albicans in vitro and in a mouse candidiasis model. PLoS One 2022; 17:e0278488. [PMID: 36477491 PMCID: PMC9728862 DOI: 10.1371/journal.pone.0278488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Fingolimod (FTY720) is a drug derived from the fungicidal compound myriocin. As it was unclear whether FTY720 has antifungal effects as well, we aimed to characterize its effect on Candida albicans in vitro and in a mouse candidiasis model. First, antifungal susceptibility testing was performed in vitro. Then, a randomized, six-arm, parallel, open-label trial was conducted on 48 mice receiving oral FTY720 (0.3 mg/kg/day), intraperitoneal C. albicans inoculation, or placebo with different combinations and chorological patterns. The outcome measures of the trial included serum concentrations of interleukin-10 and interferon-gamma, absolute lymphocyte counts, and fungal burden values in the mice's livers, kidneys, and vaginas. Broth microdilution assay revealed FTY720's minimum inhibitory concentration (MIC99) to be 0.25 mg/mL for C. albicans. The infected mice treated with FTY720 showed lower fungal burden values than the ones not treated with FTY720 (p<0.05). As expected, the mice treated with FTY720 showed a less-inflammatory immune profile compared to the ones not treated with FTY720. We hypothesize that FTY720 synergizes the host's innate immune functions by inducing the production of reactive oxygen species. Further studies are warranted to unveil the mechanistic explanations of our observations and clarify further aspects of repurposing FTY720 for clinical antifungal usage.
Collapse
|
9
|
Milford EM, Meital L, Kuballa A, Reade MC, Russell FD. Fingolimod does not prevent syndecan-4 shedding from the endothelial glycocalyx in a cultured human umbilical vein endothelial cell model of vascular injury. Intensive Care Med Exp 2022; 10:34. [PMID: 35980492 PMCID: PMC9388705 DOI: 10.1186/s40635-022-00462-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/10/2022] [Indexed: 11/27/2022] Open
Abstract
Background Shedding of the endothelial glycocalyx (EG) is associated with poor outcomes in a range of conditions including sepsis. Fresh frozen plasma (FFP) restores the damaged EG to baseline thickness, however the mechanism for this effect is unknown, and some components of FFP have adverse effects unrelated to the EG. There is some limited evidence that sphingosine-1-phosphate (S1P) within FFP restores the EG by activating the endothelial cell S1P receptor 1 (S1PR1). However, there are disadvantages to using S1P clinically as an EG restorative therapy. A potential alternative is the S1PR agonist fingolimod (FTY720). The aim of this study was to assess whether FTY720 prevents EG shedding in injured cultured human umbilical vein endothelial cells. Methods Shedding of the EG was induced in cultured human umbilical vein endothelial cells (HUVECs) by exposure to adrenaline, TNF-α and H2O2. The cells were then assigned to one of six conditions for 4 h: uninjured and untreated, injured and untreated, injured and treated with FTY720 with and without the S1PR1 inhibitor W146, and injured and treated with 25% FFP with and without W146. Syndecan-4, a component of the EG, was measured in cell supernatants, and syndecan-4 and thrombomodulin mRNA expression was quantitated in cell lysates. Results The injury resulted in a 2.1-fold increase in syndecan-4 (p < 0.001), consistent with EG shedding. Syndecan-4 and thrombomodulin mRNA expression was increased (p < 0.001) and decreased (p < 0.05), respectively, by the injury. Syndecan-4 shedding was not affected by treatment with FTY720, whereas FFP attenuated syndecan-4 shedding back to baseline levels in the injured cells and this was unaffected by W146. Neither treatment affected syndecan-4 or thrombomodulin mRNA expression. Conclusions FTY720 did not prevent syndecan-4 shedding from the EG in the HUVEC model of endothelial injury, suggesting that activation of S1PR does not prevent EG damage. FFP prevented syndecan-4 shedding from the EG via a mechanism that was independent of S1PR1 and upregulation of SDC-4 production. Further studies to examine whether FTY720 or another S1PR agonist might have EG-protective effects under different conditions are warranted, as are investigations seeking the mechanism of EG protection conferred by FFP in this experimental model.
Collapse
Affiliation(s)
- Elissa M Milford
- Faculty of Medicine, University of Queensland, Herston, QLD, Australia. .,Intensive Care Unit, Royal Brisbane and Women's Hospital, Butterfield St., Herston, QLD, Australia.
| | - Lara Meital
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia.,Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Anna Kuballa
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia.,Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Michael C Reade
- Faculty of Medicine, University of Queensland, Herston, QLD, Australia.,Intensive Care Unit, Royal Brisbane and Women's Hospital, Butterfield St., Herston, QLD, Australia.,Joint Health Command, Australian Defence Force, Canberra, ACT, Australia
| | - Fraser D Russell
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia.,Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, QLD, Australia
| |
Collapse
|
10
|
Artru F, McPhail MJW, Triantafyllou E, Trovato FM. Lipids in Liver Failure Syndromes: A Focus on Eicosanoids, Specialized Pro-Resolving Lipid Mediators and Lysophospholipids. Front Immunol 2022; 13:867261. [PMID: 35432367 PMCID: PMC9008479 DOI: 10.3389/fimmu.2022.867261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/08/2022] [Indexed: 12/30/2022] Open
Abstract
Lipids are organic compounds insoluble in water with a variety of metabolic and non-metabolic functions. They not only represent an efficient energy substrate but can also act as key inflammatory and anti-inflammatory molecules as part of a network of soluble mediators at the interface of metabolism and the immune system. The role of endogenous bioactive lipid mediators has been demonstrated in several inflammatory diseases (rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, cancer). The liver is unique in providing balanced immunotolerance to the exposure of bacterial components from the gut transiting through the portal vein and the lymphatic system. This balance is abruptly deranged in liver failure syndromes such as acute liver failure and acute-on-chronic liver failure. In these syndromes, researchers have recently focused on bioactive lipid mediators by global metabonomic profiling and uncovered the pivotal role of these mediators in the immune dysfunction observed in liver failure syndromes explaining the high occurrence of sepsis and subsequent organ failure. Among endogenous bioactive lipids, the mechanistic actions of three classes (eicosanoids, pro-resolving lipid mediators and lysophospholipids) in the pathophysiological modulation of liver failure syndromes will be the topic of this narrative review. Furthermore, the therapeutic potential of lipid-immune pathways will be described.
Collapse
Affiliation(s)
- Florent Artru
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Mark J W McPhail
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | | |
Collapse
|
11
|
Pig and Mouse Models of Hyperlipidemia and Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:379-411. [PMID: 35237978 DOI: 10.1007/978-1-0716-1924-7_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is a chronic inflammatory disorder that is the underlying cause of most cardiovascular disease. Resident cells of the artery wall and cells of the immune system participate in atherogenesis. This process is influenced by plasma lipoproteins, genetics, and the hemodynamics of the blood flow in the artery. A variety of animal models have been used to study the pathophysiology and mechanisms that contribute to atherosclerotic lesion formation. No model is ideal as each has its own advantages and limitations with respect to manipulation of the atherogenic process and modeling human atherosclerosis and lipoprotein profile. In this chapter we will discuss pig and mouse models of experimental atherosclerosis. The similarity of pig lipoprotein metabolism and the pathophysiology of the lesions in these animals with that of humans is a major advantage. While a few genetically engineered pig models have been generated, the ease of genetic manipulation in mice and the relatively short time frame for the development of atherosclerosis has made them the most extensively used model. Newer approaches to induce hypercholesterolemia in mice have been developed that do not require germline modifications. These approaches will facilitate studies on atherogenic mechanisms.
Collapse
|
12
|
Cellular mechanisms and recommended drug-based therapeutic options in diabetic cardiomyopathy. Pharmacol Ther 2021; 228:107920. [PMID: 34171330 DOI: 10.1016/j.pharmthera.2021.107920] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 12/18/2022]
Abstract
Diabetes mellitus (DM) is associated with a specific cardiac phenotype characterized by structural and functional alterations. This so-called diabetic cardiomyopathy (DM CM) is clinically relevant as patients with DM show high incidence of heart failure. Mechanistically, several parameters interact on the cardiomyocyte level leading to increased inflammation, apoptosis, reactive oxygen species and altered calcium signaling. This in turn provokes functional myocardial changes that might inter alia play into the worsened clinical outcome in DM patients. Therefore, efficient therapeutic options are urgently needed. This review focuses on mechanistic effects of currently recommended antidiabetic treatment and heart failure therapy for DM CM.
Collapse
|
13
|
Song JH, Kim GT, Park KH, Park WJ, Park TS. Bioactive Sphingolipids as Major Regulators of Coronary Artery Disease. Biomol Ther (Seoul) 2021; 29:373-383. [PMID: 33903284 PMCID: PMC8255146 DOI: 10.4062/biomolther.2020.218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is the deposition of plaque in the main arteries. It is an inflammatory condition involving the accumulation of macrophages and various lipids (low-density lipoprotein [LDL] cholesterol, ceramide, S1P). Moreover, endothelial cells, macrophages, leukocytes, and smooth muscle cells are the major players in the atherogenic process. Sphingolipids are now emerging as important regulators in various pathophysiological processes, including the atherogenic process. Various sphingolipids exist, such as the ceramides, ceramide-1-phosphate, sphingosine, sphinganine, sphingosine-1-phosphate (S1P), sphingomyelin, and hundreds of glycosphingolipids. Among these, ceramides, glycosphingolipids, and S1P play important roles in the atherogenic processes. The atherosclerotic plaque consists of higher amounts of ceramide, glycosphingolipids, and sphingomyelin. The inhibition of the de novo ceramide biosynthesis reduces the development of atherosclerosis. S1P regulates atherogenesis via binding to the S1P receptor (S1PR). Among the five S1PRs (S1PR1-5), S1PR1 and S1PR3 mainly exert anti-atherosclerotic properties. This review mainly focuses on the effects of ceramide and S1P via the S1PR in the development of atherosclerosis. Moreover, it discusses the recent findings and potential therapeutic implications in atherosclerosis.
Collapse
Affiliation(s)
- Jae-Hwi Song
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Kyung-Ho Park
- Department of Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Woo-Jae Park
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| |
Collapse
|
14
|
Cruz JDO, Silva AO, Ribeiro JM, Luizon MR, Ceron CS. Epigenetic Regulation of the N-Terminal Truncated Isoform of Matrix Metalloproteinase-2 (NTT-MMP-2) and Its Presence in Renal and Cardiac Diseases. Front Genet 2021; 12:637148. [PMID: 33732288 PMCID: PMC7959838 DOI: 10.3389/fgene.2021.637148] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/26/2021] [Indexed: 12/19/2022] Open
Abstract
Several clinical and experimental studies have documented a compelling and critical role for the full-length matrix metalloproteinase-2 (FL-MMP-2) in ischemic renal injury, progressive renal fibrosis, and diabetic nephropathy. A novel N-terminal truncated isoform of MMP-2 (NTT-MMP-2) was recently discovered, which is induced by hypoxia and oxidative stress by the activation of a latent promoter located in the first intron of the MMP2 gene. This NTT-MMP-2 isoform is enzymatically active but remains intracellular in or near the mitochondria. In this perspective article, we first present the findings about the discovery of the NTT-MMP-2 isoform, and its functional and structural differences as compared with the FL-MMP-2 isoform. Based on publicly available epigenomics data from the Encyclopedia of DNA Elements (ENCODE) project, we provide insights into the epigenetic regulation of the latent promoter located in the first intron of the MMP2 gene, which support the activation of the NTT-MMP-2 isoform. We then focus on its functional assessment by covering the alterations found in the kidney of transgenic mice expressing the NTT-MMP-2 isoform. Next, we highlight recent findings regarding the presence of the NTT-MMP-2 isoform in renal dysfunction, in kidney and cardiac diseases, including damage observed in aging, acute ischemia-reperfusion injury (IRI), chronic kidney disease, diabetic nephropathy, and human renal transplants with delayed graft function. Finally, we briefly discuss how our insights may guide further experimental and clinical studies that are needed to elucidate the underlying mechanisms and the role of the NTT-MMP-2 isoform in renal dysfunction, which may help to establish it as a potential therapeutic target in kidney diseases.
Collapse
Affiliation(s)
- Juliana de Oliveira Cruz
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alessandra O Silva
- Department of Food and Drugs, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Jessyca M Ribeiro
- Department of Food and Drugs, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Marcelo R Luizon
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carla S Ceron
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
15
|
Qian Y, Gao C, Zhao X, Song Y, Luo H, An S, Huang J, Zhang J, Jiang R. Fingolimod Attenuates Lung Injury and Cardiac Dysfunction after Traumatic Brain Injury. J Neurotrauma 2020; 37:2131-2140. [PMID: 32434456 DOI: 10.1089/neu.2019.6951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Acute lung injury (ALI) and cardiac dysfunction are common in traumatic brain injury (TBI) patients and always indicate poor outcomes. Inflammatory responses play important roles in TBI-induced cardiac and pulmonary damage. Fingolimod, an immunomodulatory agent, alleviates brain edema, restores the integrity of the blood-brain barrier (BBB), and improves functional deficits by inhibiting multiple inflammatory responses. Fingolimod (1 mg/kg) was injected intraperitoneally at 2 h after the controlled cortical impact (CCI) model was established in adult male mice. The concentration of inflammatory cytokines in the lung and heart after TBI was measured with a cytokine array. The lung wet/dry weight ratio and Evans blue dye leakage were used to quantify pulmonary edema and capillary leakage. Immunofluorescence, electron microscopy, and echocardiographic examination were used to assess the pathology and functional deficits in hearts. We found that TBI caused significant heart and lung damage. The administration of fingolimod significantly reduced the elevated inflammatory cytokine production, neutrophil infiltration, the leakage of protein in bronchoalveolar lavage fluid (BALF), and the wet/dry weight ratio in lung tissue at 3 days after TBI. In addition, fingolimod treatment also alleviated the inflammatory response in the heart; decreased cardiac apoptosis, fibrosis, and histological microstructural changes; and improved cardiac function from 3 days after TBI and maintained it for 30 days after TBI as measured by echocardiography. These results suggest that TBI resulted in significant cardiac and pulmonary damage accompanied by significant inflammatory responses in heart and lung tissue. Fingolimod treatment reduced the inflammatory response and alleviated TBI-induced lung and heart injury.
Collapse
Affiliation(s)
- Yu Qian
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Chuang Gao
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Yiming Song
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongliang Luo
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuo An
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhao Huang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Rongcai Jiang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
16
|
Red Wine Grape Pomace Attenuates Atherosclerosis and Myocardial Damage and Increases Survival in Association with Improved Plasma Antioxidant Activity in a Murine Model of Lethal Ischemic Heart Disease. Nutrients 2019; 11:nu11092135. [PMID: 31500172 PMCID: PMC6770693 DOI: 10.3390/nu11092135] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/29/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
A healthy dietary pattern and high quality nutrient intake reduce atherosclerotic cardiovascular disease risk. Red wine grape pomace (RWGP)—a rich natural source of dietary fiber and antioxidants—appears to be a potential functional food ingredient. The impact of a dietary supplementation with RWGP flour was evaluated in atherogenic diet-fed SR-B1 KO/ApoER61h/h mice, a model of lethal ischemic heart disease. SR-B1 KO/ApoER61h/h mice were fed with atherogenic (high fat, cholesterol, and cholic acid, HFC) diet supplemented with: (a) 20% chow (HFC-Control), (b) 20% RWGP flour (HFC-RWGP), or (c) 10% chow/10% oat fiber (HFC-Fiber); and survival time was evaluated. In addition, SR-B1 KO/ApoER61h/h mice were fed for 7 or 14 days with HFC-Control or HFC-RWGP diets and plasma lipid levels, inflammation, oxidative damage, and antioxidant activity were measured. Atherosclerosis and myocardial damage were assessed by histology and magnetic resonance imaging, respectively. Supplementation with RWGP reduced premature death, changed TNF-α and IL-10 levels, and increased plasma antioxidant activity. Moreover, decreased atheromatous aortic and brachiocephalic plaque sizes and attenuated myocardial infarction and dysfunction were also observed. These results suggest that RWGP flour intake may be used as a non-pharmacological therapeutic approach, contributing to decreased progression of atherosclerosis, reduced coronary heart disease, and improved cardiovascular outcomes.
Collapse
|
17
|
Ahmed N. Cardioprotective mechanism of FTY720 in ischemia reperfusion injury. J Basic Clin Physiol Pharmacol 2019; 30:jbcpp-2019-0063. [PMID: 31469655 DOI: 10.1515/jbcpp-2019-0063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/06/2019] [Indexed: 12/17/2022]
Abstract
Cardioprotection is a very challenging area in the field of cardiovascular sciences. Myocardial damage accounts for nearly 50% of injury due to reperfusion, yet there is no effective strategy to prevent this to reduce the burden of heart failure. During last couple of decades, by combining genetic and bimolecular studies, many new drugs have been developed to treat hypertension, heart failure, and cancer. The use of percutaneous coronary intervention has reduced the mortality and morbidity of acute coronary syndrome dramatically. However, there is no standard therapy available that can mitigate cardiac reperfusion injury, which contributes to up to half of myocardial infarcts. Literature shows that the activation of sphingosine receptors, which are G protein-coupled receptors, induces cardioprotection both in vitro and in vivo. The exact mechanism of this protection is not clear yet. In this review, we discuss the mechanism of ischemia reperfusion injury and the role of the FDA-approved sphingosine 1 phosphate drug fingolimod in cardioprotection.
Collapse
Affiliation(s)
- Naseer Ahmed
- The Aga Khan University, Medical College, Karachi, Pakistan, Phone: +92 21 3486 4465
| |
Collapse
|
18
|
Wang J, Duan Y, Sluijter JPG, Xiao J. Lymphocytic subsets play distinct roles in heart diseases. Am J Cancer Res 2019; 9:4030-4046. [PMID: 31281530 PMCID: PMC6592175 DOI: 10.7150/thno.33112] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022] Open
Abstract
Heart diseases are one of the leading causes of death for humans in the world. Increasing evidence has shown that myocardial injury induced innate and adaptive immune responses upon early cellular damage but also during chronic phases post-injury. The immune cells can not only aggravate the injury but also play an essential role in the induction of wound healing responses, which means they play a complex role throughout the acute inflammatory response and reparative response after cardiac injury. This review will summarize the current experimental and clinical evidence of lymphocytes, one of the major types of immune cells, participate in heart diseases and try to explain the possible role of these immune cells following cardiac injury.
Collapse
|
19
|
Jia J, Guo X, Feng L, Yin X, Zhu L, Li J, Yu D, Fang Y, Jiang Z, Yu M, Xia H, Shi L, Ju L, Zhang M, Xiao Y, Lu CA, Shi W, Zhang X, Lou J. Genome-wide profiling reveals novel microRNAs in hand-spinning-specific chrysotile exposure. Epigenomics 2019; 11:511-525. [DOI: 10.2217/epi-2018-0143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: We aimed to explore miRNA expression profiles in hand-spinning chrysotile exposed workers and their potential influencing factors. Methods: miRNA array technique was applied to screen differentially expressed miRNAs between plasma samples from three exposed workers and three controls. Then, seven selected miRNAs were validated in 143 workers and 100 controls, and the potential influencing factors were revealed by multiple linear regression. Finally, the expression levels of those seven miRNAs were evaluated in human mesothelial cells (Met-5A) that were exposed to chrysotile at 5 μg·cm-2 for 8, 24 and 48 h, respectively. Results & conclusion: Hand-spinning chrysotile exposure can result in differential expression of miRNAs. Several of those miRNAs have positive correlations with asbestos exposure.
Collapse
Affiliation(s)
- Junlin Jia
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Xinnian Guo
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Lingfang Feng
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Xianhong Yin
- Jiading District Center for Disease Control & Prevention, Shanghai, PR China
| | - Lijin Zhu
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Jinhao Li
- Department of Molecular Environmental Biology, College of Natural Resources, University of California, Berkeley, CA 94720, USA
| | - Dandan Yu
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Yuan Fang
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Zhaoqiang Jiang
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Min Yu
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Hailing Xia
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Li Shi
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Li Ju
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Min Zhang
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Yun Xiao
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Chensheng A Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Wei Shi
- Department of Surgery, Children’s Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA
| | - Xing Zhang
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| | - Jianlin Lou
- Department of Pneumoconiosis, Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, PR China
| |
Collapse
|
20
|
Abstract
The liver is the central organ involved in lipid metabolism and the gastrointestinal (GI) tract is responsible for nutrient absorption and partitioning. Obesity, dyslipidemia and metabolic disorders are of increasing public health concern worldwide, and novel therapeutics that target both the liver and the GI tract (gut-liver axis) are much needed. In addition to aiding fat digestion, bile acids act as important signaling molecules that regulate lipid, glucose and energy metabolism via activating nuclear receptor, G protein-coupled receptors (GPCRs), Takeda G protein receptor 5 (TGR5) and sphingosine-1-phosphate receptor 2 (S1PR2). Sphingosine-1-phosphate (S1P) is synthesized by two sphingosine kinase isoforms and is a potent signaling molecule that plays a critical role in various diseases such as fatty liver, inflammatory bowel disease (IBD) and colorectal cancer. In this review, we will focus on recent findings related to the role of S1P-mediated signaling pathways in the gut-liver axis.
Collapse
Affiliation(s)
- Eric K. Kwong
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA,McGuire VA Medical Center, Richmond, VA, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA,McGuire VA Medical Center, Richmond, VA, USA,Corresponding author. Department of Microbiology and Immunology, Virginia Commonwealth University, McGuire Veterans Affairs Medical Center, Richmond, VA, USA. (H. Zhou)
| |
Collapse
|
21
|
Apolipoprotein E and Atherosclerosis: From Lipoprotein Metabolism to MicroRNA Control of Inflammation. J Cardiovasc Dev Dis 2018; 5:jcdd5020030. [PMID: 29789495 PMCID: PMC6023389 DOI: 10.3390/jcdd5020030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 12/26/2022] Open
Abstract
Apolipoprotein (apo) E stands out among plasma apolipoproteins through its unprecedented ability to protect against atherosclerosis. Although best recognized for its ability to mediate plasma lipoprotein clearance in the liver and protect against macrophage foam cell formation, our recent understanding of the influence that apoE can exert to control atherosclerosis has significantly widened. Among apoE’s newfound athero-protective properties include an ability to control exaggerated hematopoiesis, blood monocyte activation and aortic stiffening in mice with hyperlipidemia. Mechanisms responsible for these exciting new properties extend beyond apoE’s ability to prevent cellular lipid excess. Rather, new findings have revealed a role for apoE in regulating microRNA-controlled cellular signaling in cells of the immune system and vascular wall. Remarkably, infusions of apoE-responsive microRNA mimics were shown to substitute for apoE in protecting against systemic and vascular inflammation to suppress atherosclerosis in mice with hyperlipidemia. Finally, more recent evidence suggests that apoE may control the release of microvesicles that could modulate cellular signaling, inflammation and atherosclerosis at a distance. These exciting new findings position apoE within the emerging field of intercellular communication that could introduce new approaches to control atherosclerosis cardiovascular disease.
Collapse
|
22
|
Inflammation following acute myocardial infarction: Multiple players, dynamic roles, and novel therapeutic opportunities. Pharmacol Ther 2018; 186:73-87. [PMID: 29330085 PMCID: PMC5981007 DOI: 10.1016/j.pharmthera.2018.01.001] [Citation(s) in RCA: 529] [Impact Index Per Article: 88.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute myocardial infarction (AMI) and the heart failure that often follows, are major causes of death and disability worldwide. As such, new therapies are required to limit myocardial infarct (MI) size, prevent adverse left ventricular (LV) remodeling, and reduce the onset of heart failure following AMI. The inflammatory response to AMI, plays a critical role in determining MI size, and a persistent pro-inflammatory reaction can contribute to adverse post-MI LV remodeling, making inflammation an important therapeutic target for improving outcomes following AMI. In this article, we provide an overview of the multiple players (and their dynamic roles) involved in the complex inflammatory response to AMI and subsequent LV remodeling, and highlight future opportunities for targeting inflammation as a therapeutic strategy for limiting MI size, preventing adverse LV remodeling, and reducing heart failure in AMI patients.
Collapse
|
23
|
Update on the Protective Role of Regulatory T Cells in Myocardial Infarction: A Promising Therapy to Repair the Heart. J Cardiovasc Pharmacol 2017; 68:401-413. [PMID: 27941502 DOI: 10.1097/fjc.0000000000000436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial infarction (MI) remains one of the leading causes of heart failure development and death worldwide. To date, interventional and pharmacological therapies are effective in reducing the onset of heart failure and promoting survival. However, progressive maladaptive remodeling post-MI persists in a large fraction of patients resulting in poor prognosis. Immune cell responses and an inflammatory environment largely contribute to adverse cardiac remodeling post-MI. CD4FOXP3 regulatory T cells (Tregs) are known for their immunosuppressive capacity and have been successfully implemented in multiple preclinical studies of permanent and ischemia-reperfusion MI. In this review, we highlight the important cardioprotective role of Tregs at the cardiac tissue, cellular, and molecular level, as well as the most prominent pharmacological venues that could be used to exploit Tregs as a novel therapeutic intervention to lessen myocardial injury post-MI.
Collapse
|
24
|
Kwong EK, Li X, Hylemon PB, Zhou H. Sphingosine Kinases/Sphingosine 1-Phosphate Signaling in Hepatic Lipid Metabolism. ACTA ACUST UNITED AC 2017; 3:176-183. [PMID: 29130028 DOI: 10.1007/s40495-017-0093-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ever-increasing prevalence of metabolic diseases such as dyslipidemia and diabetes in the western world continues to be of great public health concern. Biologically active sphingolipids, such as sphingosine 1-phosphate (S1P) and ceramide, are important regulators of lipid metabolism. S1P not only directly functions as an active intracellular mediator, but also activates multiple signaling pathways via five transmembrane G-protein coupled receptors (GPCRs), S1PR1-5. S1P is exclusively formed by sphingosine kinases (SphKs). Two isoforms of SphKs, SphK1 and SphK2, have been identified. Recent identification of the conjugated bile acid-induced activation of S1PR2 as a key regulator of SphK2 opened new directions for both the sphingolipid and bile acid research fields. The role of SphKs/S1P-mediated signaling pathways in health and various human diseases has been extensively reviewed elsewhere. This review focuses on recent findings related to SphKs/S1P-medaited signaling pathways in regulating hepatic lipid metabolism.
Collapse
Affiliation(s)
- Eric K Kwong
- Department of Microbiology and Immunology, Medical College of Virginia Campus, McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Xiaojiaoyang Li
- Department of Microbiology and Immunology, Medical College of Virginia Campus, McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Phillip B Hylemon
- Department of Microbiology and Immunology, Medical College of Virginia Campus, McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Huiping Zhou
- Department of Microbiology and Immunology, Medical College of Virginia Campus, McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, 23298
| |
Collapse
|
25
|
Zhao Z, Wang J, Huo Z, Wang Z, Mei Q. FTY720 elevates smooth muscle contraction of aorta and blood pressure in rats via ERK activation. Pharmacol Res Perspect 2017; 5:e00308. [PMID: 28480040 PMCID: PMC5415948 DOI: 10.1002/prp2.308] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/07/2017] [Indexed: 01/04/2023] Open
Abstract
Sphingosine 1‐phosphate (S1P) is an important signaling sphingolipid involved in the pathogenesis of various cardio cerebral vascular diseases such as ischemic stroke. In particular, the S1P mimetic FTY720 is protective for brain against ischemic conditions. However, whether and how FTY720 can modulate vascular tone and blood pressure remains to be determined. We showed that FTY720 (1 mg/kg) enhanced the contractile response of rat thoracic aortic rings induced by high potassium and phenylephrine, respectively. This enhancement involves the activation of extracellular signal‐regulated kinase (ERK) since ERK phosphorylation was also enhanced and application of PD98059 (10 μmol/L), an inhibitor of ERK activation abrogated the aforementioned enhanced response by FTY720. In parallel, FTY720 (1 mg/kg) led to a modest elevation of blood pressure in rats, effects also being prevented by PD98059. In contrast, FTY720 decreased the high potassium‐induced contractile response in basilarartery preparations from rabbits, an effect blocked by PD98059. Together, FTY720‐induced an enhanced response of artery contractility in aorta and in arterial pressure involving ERK activation, with an attenuation in basilarartery contractility. This action property of FTY720 would be endowed with a potential of facilitating more blood flow perfusion to the brain and improving blood supply to the ischemic brain region and could be useful as an adjuvant in the treatment of ischemic stroke in the clinics.
Collapse
Affiliation(s)
- Zhen Zhao
- State Key Laboratory of New Drug & Pharmaceutical Process Shanghai Institute of Pharmaceutical Industry Shanghai 200437 China
| | - Jinxin Wang
- State Key Laboratory of New Drug & Pharmaceutical Process Shanghai Institute of Pharmaceutical Industry Shanghai 200437 China
| | - Zhijun Huo
- State Key Laboratory of New Drug & Pharmaceutical Process Shanghai Institute of Pharmaceutical Industry Shanghai 200437 China
| | - Zhiyong Wang
- State Key Laboratory of New Drug & Pharmaceutical Process Shanghai Institute of Pharmaceutical Industry Shanghai 200437 China
| | - Qibing Mei
- State Key Laboratory of New Drug & Pharmaceutical Process Shanghai Institute of Pharmaceutical Industry Shanghai 200437 China
| |
Collapse
|
26
|
Grilo GA, Shaver PR, de Castro Brás LE. Mechanisms of cardioprotection via modulation of the immune response. Curr Opin Pharmacol 2017; 33:6-11. [PMID: 28388508 PMCID: PMC11034833 DOI: 10.1016/j.coph.2017.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 03/08/2017] [Indexed: 10/19/2022]
Abstract
Both morbidity and mortality as a result of cardiovascular disease remain significant worldwide and account for approximately 31% of annual deaths in the US. Current research is focused on novel therapeutic strategies to protect the heart during and after ischemic events and from subsequent adverse myocardial remodeling. After cardiac insult, the immune system is activated and plays an essential role in the beginning, development, and resolution of the healing cascade. Uncontrolled inflammatory responses can cause chronic disease and exacerbate progression to heart failure and therefore, constitute a major area of focus of cardiac therapies. In the present overview, we share novel insights and promising therapeutic cardioprotective strategies that target the immune response.
Collapse
Affiliation(s)
- Gabriel A Grilo
- The Brody School of Medicine, Department of Physiology, East Carolina University, 600 Moye Blvd, Greenville NC 27834, USA
| | - Patti R Shaver
- The Brody School of Medicine, Department of Physiology, East Carolina University, 600 Moye Blvd, Greenville NC 27834, USA
| | - Lisandra E de Castro Brás
- The Brody School of Medicine, Department of Physiology, East Carolina University, 600 Moye Blvd, Greenville NC 27834, USA.
| |
Collapse
|
27
|
van Vuuren D, Marais E, Genade S, Lochner A. The differential effects of FTY720 on functional recovery and infarct size following myocardial ischaemia/reperfusion. Cardiovasc J Afr 2017; 27:375-386. [PMID: 27966000 PMCID: PMC5408499 DOI: 10.5830/cvja-2016-039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/30/2016] [Indexed: 01/08/2023] Open
Abstract
AIM The aim of this study was to evaluate the effects of the sphingosine analogue, FTY720 (Fingolimod), on the outcomes of myocardial ischaemia/reperfusion (I/R) injury. METHODS Two concentrations of FTY720 (1 or 2.5 µM were administered either prior to (PreFTY), or following (PostFTY) 20 minutes' global (GI) or 35 minutes' regional ischaemia (RI) in the isolated, perfused, working rat heart. Functional recovery during reperfusion was assessed following both models of ischaemia, while infarct size (IFS) was determined following RI. RESULTS FTY720 at 1 µM exerted no effect on functional recovery, while 2.5 µM significantly impaired aortic output (AO) recovery when administered prior to GI (% recovery: control: 33.88 ± 6.12% vs PreFTY: 0%, n = 6-10; p < 0.001), as well as before and after RI ( % recovery: control: 27.86 ± 13.22% vs PreFTY: 0.62% ; p < 0.05; and PostFTY: 2.08%; p = 0.0585, n = 6). FTY720 at 1 µM administered during reperfusion reduced IFS (% of area at risk (AAR): control: 39.89 ± 3.93% vs PostFTY: 26.56 ± 4.32%, n = 6-8; p < 0.05), while 2.5 µM FTY720 reduced IFS irrespective of the time of administration ( % of AAR: control: 39.89 ± 3.93% vs PreFTY: 29.97 ± 1.03% ; and PostFTY: 30.45 ± 2.16%, n = 6; p < 0.05). CONCLUSION FTY720 exerted divergent outcomes on function and tissue survival depending on the concentration administered, as well as the timing of administration.
Collapse
Affiliation(s)
- Derick van Vuuren
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa.
| | - Erna Marais
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Sonia Genade
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Amanda Lochner
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
28
|
Immunosuppression With FTY720 Reverses Cardiac Dysfunction in Hypomorphic ApoE Mice Deficient in SR-BI Expression That Survive Myocardial Infarction Caused by Coronary Atherosclerosis. J Cardiovasc Pharmacol 2016; 67:47-56. [PMID: 26322923 DOI: 10.1097/fjc.0000000000000312] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AIMS We recently reported that immunosuppression with FTY720 improves cardiac function and extends longevity in Hypomorphic ApoE mice deficient in scavenger receptor Type-BI expression, also known as the HypoE/SR-BI(–/–) mouse model of diet-induced coronary atherosclerosis and myocardial infarction (MI). In this study, we tested the impact of FTY720 on cardiac dysfunction in HypoE/SR-BI(–/–) mice that survive MI and subsequently develop chronic heart failure. METHODS/RESULTS HypoE/SR-BI(–/–) mice were bred to Mx1-Cre transgenic mice, and offspring were fed a high-fat diet (HFD) for 3.5 weeks to provoke hyperlipidemia, coronary atherosclerosis, and recurrent MIs. In contrast to our previous study, hyperlipidemia was rapidly reversed by inducible Cre-mediated gene repair of the HypoE allele and switching mice to a normal chow diet. Mice that survived the period of HFD were subsequently given oral FTY720 in drinking water or not, and left ventricular (LV) function was monitored using serial echocardiography for up to 15 weeks. In untreated mice, LV performance progressively deteriorated. Although FTY720 treatment did not initially prevent a decline of heart function among mice 6 weeks after Cre-mediated gene repair, it almost completely restored normal LV function in these mice by 15 weeks. Reversal of heart failure did not result from reduced atherosclerosis as the burden of aortic and coronary atherosclerosis actually increased to similar levels in both groups of mice. Rather, FTY720 caused systemic immunosuppression as assessed by reduced numbers of circulating T and B lymphocytes. In contrast, FTY720 did not enhance the loss of T cells or macrophages that accumulated in the heart during the HFD feeding period, but it did enhance the loss of B cells soon after plasma lipid lowering. Moreover, FTY720 potently reduced the expression of matrix metalloproteinase-2 and genes involved in innate immunity-associated inflammation in the heart. CONCLUSIONS Our data demonstrate that immunosuppression with FTY720 prevents postinfarction myocardial remodeling and chronic heart failure.
Collapse
|
29
|
The CXCL10/CXCR3 Axis and Cardiac Inflammation: Implications for Immunotherapy to Treat Infectious and Noninfectious Diseases of the Heart. J Immunol Res 2016; 2016:4396368. [PMID: 27795961 PMCID: PMC5066021 DOI: 10.1155/2016/4396368] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/16/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidence reveals involvement of T lymphocytes and adaptive immunity in the chronic inflammation associated with infectious and noninfectious diseases of the heart, including coronary artery disease, Kawasaki disease, myocarditis, dilated cardiomyopathies, Chagas, hypertensive left ventricular (LV) hypertrophy, and nonischemic heart failure. Chemokine CXCL10 is elevated in cardiovascular diseases, along with increased cardiac infiltration of proinflammatory Th1 and cytotoxic T cells. CXCL10 is a chemoattractant for these T cells and polarizing factor for the proinflammatory phenotype. Thus, targeting the CXCL10 receptor CXCR3 is a promising therapeutic approach to treating cardiac inflammation. Due to biased signaling CXCR3 also couples to anti-inflammatory signaling and immunosuppressive regulatory T cell formation when activated by CXCL11. Numbers and functionality of regulatory T cells are reduced in patients with cardiac inflammation, supporting the utility of biased agonists or biologicals to simultaneously block the pro-inflammatory and activate the anti-inflammatory actions of CXCR3. Other immunotherapy strategies to boost regulatory T cell actions include intravenous immunoglobulin (IVIG) therapy, adoptive transfer, immunoadsorption, and low-dose interleukin-2/interleukin-2 antibody complexes. Pharmacological approaches include sphingosine 1-phosphate receptor 1 agonists and vitamin D supplementation. A combined strategy of switching CXCR3 signaling from pro- to anti-inflammatory and improving Treg functionality is predicted to synergistically lessen adverse cardiac remodeling.
Collapse
|
30
|
Juif PE, Kraehenbuehl S, Dingemanse J. Clinical pharmacology, efficacy, and safety aspects of sphingosine-1-phosphate receptor modulators. Expert Opin Drug Metab Toxicol 2016; 12:879-95. [PMID: 27249325 DOI: 10.1080/17425255.2016.1196188] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Sphingosine-1-phosphate (S1P) receptor modulators, of which one has received marketing approval and several others are in clinical development, display promising potential in the treatment of a spectrum of autoimmune diseases. AREAS COVERED Administration of S1P1 receptor modulators leads to functional receptor antagonism triggering sustained inhibition of the egress of lymphocytes from lymphoid organs. First-dose administration is associated with transient cardiovascular effects. We compiled and discussed available pharmacokinetic, pharmacodynamic, and safety data of selective and non-selective S1P receptor modulators that were investigated in recent years. EXPERT OPINION The safety profile of S1P receptor modulators is considered better than other classes of immunomodulators and was further improved by the development of up-titration regimens to mitigate first-dose effects. S1P receptor modulators display similar pharmacodynamic effects but have very different pharmacokinetic profiles. Drugs with a rapid elimination are of interest in case of opportunistic infections or pregnancy, whereas the need of re-initiation of up-titration in case of treatment interruption can present a challenge.
Collapse
Affiliation(s)
- Pierre-Eric Juif
- a Department of Clinical Pharmacology , Actelion Pharmaceuticals Ltd , Allschwil , Switzerland
| | - Stephan Kraehenbuehl
- b Department of Clinical Pharmacology and Toxicology , Universitätsspital Basel , Basel , Switzerland
| | - Jasper Dingemanse
- a Department of Clinical Pharmacology , Actelion Pharmaceuticals Ltd , Allschwil , Switzerland
| |
Collapse
|
31
|
Keul P, van Borren MMGJ, Ghanem A, Müller FU, Baartscheer A, Verkerk AO, Stümpel F, Schulte JS, Hamdani N, Linke WA, van Loenen P, Matus M, Schmitz W, Stypmann J, Tiemann K, Ravesloot JH, Alewijnse AE, Hermann S, Spijkers LJA, Hiller KH, Herr D, Heusch G, Schäfers M, Peters SLM, Chun J, Levkau B. Sphingosine-1-Phosphate Receptor 1 Regulates Cardiac Function by Modulating Ca2+ Sensitivity and Na+/H+ Exchange and Mediates Protection by Ischemic Preconditioning. J Am Heart Assoc 2016; 5:JAHA.116.003393. [PMID: 27207969 PMCID: PMC4889204 DOI: 10.1161/jaha.116.003393] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background Sphingosine‐1‐phosphate plays vital roles in cardiomyocyte physiology, myocardial ischemia–reperfusion injury, and ischemic preconditioning. The function of the cardiomyocyte sphingosine‐1‐phosphate receptor 1 (S1P1) in vivo is unknown. Methods and Results Cardiomyocyte‐restricted deletion of S1P1 in mice (S1P1αMHCCre) resulted in progressive cardiomyopathy, compromised response to dobutamine, and premature death. Isolated cardiomyocytes from S1P1αMHCCre mice revealed reduced diastolic and systolic Ca2+ concentrations that were secondary to reduced intracellular Na+ and caused by suppressed activity of the sarcolemmal Na+/H+ exchanger NHE‐1 in the absence of S1P1. This scenario was successfully reproduced in wild‐type cardiomyocytes by pharmacological inhibition of S1P1 or sphingosine kinases. Furthermore, Sarcomere shortening of S1P1αMHCCre cardiomyocytes was intact, but sarcomere relaxation was attenuated and Ca2+ sensitivity increased, respectively. This went along with reduced phosphorylation of regulatory myofilament proteins such as myosin light chain 2, myosin‐binding protein C, and troponin I. In addition, S1P1 mediated the inhibitory effect of exogenous sphingosine‐1‐phosphate on β‐adrenergic–induced cardiomyocyte contractility by inhibiting the adenylate cyclase. Furthermore, ischemic precondtioning was abolished in S1P1αMHCCre mice and was accompanied by defective Akt activation during preconditioning. Conclusions Tonic S1P1 signaling by endogenous sphingosine‐1‐phosphate contributes to intracellular Ca2+ homeostasis by maintaining basal NHE‐1 activity and controls simultaneously myofibril Ca2+ sensitivity through its inhibitory effect on adenylate cyclase. Cardioprotection by ischemic precondtioning depends on intact S1P1 signaling. These key findings on S1P1 functions in cardiac physiology may offer novel therapeutic approaches to cardiac diseases.
Collapse
Affiliation(s)
- Petra Keul
- Institute for Pathophysiology, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | | | - Alexander Ghanem
- Department of Cardiology, Universitätsklinikum Bonn, Bonn, Germany
| | | | | | - Arie O Verkerk
- Heart Failure Research Center, AMC, University of Amsterdam, The Netherlands
| | - Frank Stümpel
- Institute for Pharmakology und Toxikology, Münster, Germany
| | | | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Wolfgang A Linke
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Pieter van Loenen
- Department of Pharmacology & Pharmacotherapy, AMC, University of Amsterdam, The Netherlands
| | - Marek Matus
- Institute for Pharmakology und Toxikology, Münster, Germany Department of Pharmacology and Toxicology, Comenius University, Bratislava, Slovakia
| | | | - Jörg Stypmann
- Medizinische Klinik und Poliklinik C, Universitätsklinikum Münster, Münster, Germany
| | - Klaus Tiemann
- Medizinische Klinik und Poliklinik C, Universitätsklinikum Münster, Münster, Germany
| | | | - Astrid E Alewijnse
- Department of Pharmacology & Pharmacotherapy, AMC, University of Amsterdam, The Netherlands
| | - Sven Hermann
- European Institute for Molecular Imaging, Münster, Germany
| | - Léon J A Spijkers
- Department of Pharmacology & Pharmacotherapy, AMC, University of Amsterdam, The Netherlands
| | - Karl-Heinz Hiller
- MRB Forschungszentrum Magnet-Resonanz-Bayern e.V., Würzburg, Germany
| | - Deron Herr
- Department of Molecular Biology, Scripps Research Institute, La Jolla, CA
| | - Gerd Heusch
- Institute for Pathophysiology, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | | | - Stephan L M Peters
- Department of Pharmacology & Pharmacotherapy, AMC, University of Amsterdam, The Netherlands
| | - Jerold Chun
- Department of Molecular Biology, Scripps Research Institute, La Jolla, CA
| | - Bodo Levkau
- Institute for Pathophysiology, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| |
Collapse
|
32
|
Abstract
Hypertriglyceridemic pancreatitis (HTGP) is often encountered clinically as a common form of recurrent acute pancreatitis (AP). It is important to evaluate the management of severe hypertriglyceridemia (HTG) or anti-inflammation in the prophylaxis of HTGP in the clinic. FTY720 (2-amino-2[2-(4-octylphenyl) ethyl]-1, 3-propanediol) is a new anti-inflammatory agent with low toxicity and reported to ameliorate lung injury with pancreatitis in rat. We evaluated its protective affection on AP induced by seven hourly intraperitoneal injection of cerulein in apolipoprotein CIII transgenic mice with severe HTG. FTY720 at 1.5 mg/kg was administered by gastric lavage daily for 3 days before induction of AP. The effects of FTY720 to protect against HTGP were assessed by serum amylase, pancreatic pathological scores, immunostaining, and the expression of inflammatory cytokine genes. As a result, injection of cerulein resulted in more severe pathological changes of AP and higher monocyte chemoattractant protein 1 expression in the pancreas in transgenic than in nontransgenic mice. FTY720 pretreatment improved the pathological severity of AP and decreased the expression of monocyte chemoattractant protein 1 in the pancreas significantly, especially near fourfold reduction in transgenic mice. However, FTY720 did not affect plasma triglyceride levels, and other inflammatory factors and plasma amylase were not correlated with the extent of pancreatic damage in AP with or without FTY720 administration. In summary, our study in a new model, apolipoprotein CIII transgenic mice, demonstrated that HTG mice are susceptible to induction of AP. Prophylactic treatment of FTY720 can significantly attenuate cerulein-induced AP and hence warrant further investigation of sphingosine-1-phosphate receptors agonist for potential clinical application in recurrent attacks of HTGP.
Collapse
|
33
|
Hermann S, Kuhlmann MT, Starsichova A, Eligehausen S, Schäfers K, Stypmann J, Tiemann K, Levkau B, Schäfers M. Imaging Reveals the Connection Between Spontaneous Coronary Plaque Ruptures, Atherothrombosis, and Myocardial Infarctions in HypoE/SRBI-/- Mice. J Nucl Med 2016; 57:1420-7. [PMID: 27127225 DOI: 10.2967/jnumed.115.171132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/21/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED The hyperlipidemic mouse model HypoE/SRBI(-/-) has been shown to develop occlusive coronary atherosclerosis followed by myocardial infarctions and premature deaths in response to high-fat, high-cholesterol diet (HFC). However, the causal connection between myocardial infarctions and atherosclerotic plaque rupture events in the coronary arteries has not been investigated so far. The objective of this study was to assess whether diet-induced coronary plaque ruptures trigger atherothrombotic occlusions, resulting in myocardial infarctions in HFC-fed HypoE/SRBI(-/-) mice. METHODS HypoE/SRBI(-/-) mice were characterized with respect to the individual dynamics of myocardial infarctions and features of infarct-related coronary atherosclerosis by serial noninvasive molecular and functional imaging, histopathology, and a pharmaceutical intervention. Detailed histologic analysis of whole mouse hearts was performed when spontaneously occurring acute myocardial infarctions were diagnosed by imaging. RESULTS Using the imaging-triggered approach, we discovered thrombi in 32 (10.8%) of all 296 atherosclerotic coronary plaques in 14 HFC-fed HypoE/SRBI(-/-) mice. These thrombi typically were found in arteries presenting with inflammatory plaque phenotypes. Acetylsalicylic acid treatment did not attenuate the development of atherosclerotic coronary plaques but profoundly reduced the incidence of premature deaths, the number of thrombi (7 in 249 plaques), and also the degree of inflammation in the culprit lesions. CONCLUSION HFC-induced ruptures of coronary plaques trigger atherothrombosis, vessel occlusions, myocardial infarctions, and sudden death in these mice. Thus, the HypoE/SRBI(-/-) mouse model mimics major features of human coronary heart disease and might therefore be a valuable model for the investigation of molecular and cellular parameters driving plaque rupture-related events and the development of new interventional approaches.
Collapse
Affiliation(s)
- Sven Hermann
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany DFG EXC 1003 Cluster of Excellence 'Cells in Motion', University of Münster, Münster, Germany
| | - Michael T Kuhlmann
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Andrea Starsichova
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Sarah Eligehausen
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Klaus Schäfers
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany DFG EXC 1003 Cluster of Excellence 'Cells in Motion', University of Münster, Münster, Germany
| | - Jörg Stypmann
- Division of Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Klaus Tiemann
- Division of Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Bodo Levkau
- Institute of Pathophysiology, University Duisburg-Essen, Essen, Germany; and
| | - Michael Schäfers
- European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany DFG EXC 1003 Cluster of Excellence 'Cells in Motion', University of Münster, Münster, Germany Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
34
|
Santos-Gallego CG, Vahl TP, Goliasch G, Picatoste B, Arias T, Ishikawa K, Njerve IU, Sanz J, Narula J, Sengupta PP, Hajjar RJ, Fuster V, Badimon JJ. Sphingosine-1-Phosphate Receptor Agonist Fingolimod Increases Myocardial Salvage and Decreases Adverse Postinfarction Left Ventricular Remodeling in a Porcine Model of Ischemia/Reperfusion. Circulation 2016; 133:954-66. [PMID: 26826180 DOI: 10.1161/circulationaha.115.012427] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 01/08/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Fingolimod, a sphingosine-1-phosphate receptor agonist, is used for the treatment of multiple sclerosis and exerts antiapoptotic properties. We hypothesized that sphingosine-1-phosphate receptor activation with fingolimod during acute myocardial infarction (MI) inhibits apoptosis, leading to increased myocardial salvage, reduced infarct size, and mitigated left ventricular (LV) remodeling in a porcine model of ischemia/reperfusion. METHODS AND RESULTS Ischemia/reperfusion was induced in pigs by balloon occlusion of the left anterior descending artery, followed by reperfusion. Animals randomly received fingolimod or saline (control). In short-term experiments, fingolimod treatment activated the cardioprotective reperfusion injury salvage kinase and survivor activating factor enhancement pathways in the infarct border zone 24 hours after MI, leading to decreased cardiomyocyte apoptosis and reduced myocardial oxidative stress. These effects were abolished by specific inhibitors of both pathways, demonstrating that fingolimod-induced cardioprotection was mediated by reperfusion injury salvage kinase and survivor activating factor enhancement pathways. In long-term experiments, fingolimod significantly improved myocardial salvage, reduced infarct size, and improved systolic LV function measured by cardiac magnetic resonance 1 week and 1 month after MI. Importantly, fingolimod mitigated the development of adverse post-MI LV remodeling 1 month after MI. Specifically, fingolimod treatment led to a significant reduction in LV mass, LV dilatation, and neurohormonal activation, and it preserved LV geometry. Furthermore, fingolimod decreased interstitial fibrosis, cardiomyocyte hypertrophy, and chronic activation of Akt and extracellular receptor kinase 1/2 in the remote noninfarcted myocardium. CONCLUSIONS Sphingosine-1-phosphate receptor activation with fingolimod during acute MI reduced infarct size via the reperfusion injury salvage kinase and survivor activating factor enhancement pathways, improved systolic LV function, and mitigated post-MI LV remodeling. Our data strongly support a cardioprotective role for sphingosine-1-phosphate receptor activation during MI.
Collapse
Affiliation(s)
- Carlos G Santos-Gallego
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.).
| | - Torsten P Vahl
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Georg Goliasch
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Belen Picatoste
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Teresa Arias
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Kiyotake Ishikawa
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Ida U Njerve
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Javier Sanz
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Jagat Narula
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Partho P Sengupta
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Roger J Hajjar
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Valentin Fuster
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| | - Juan J Badimon
- From Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, New York, NY (C.G.S.-G., T.P.V., G.G., B.P., T.A., K.I., I.U.N., J.S., J.N., P.P.S., R.J.H., V.F., J.J.B.); Columbia University Medical Center, New York Presbyterian Hospital, NY (T.P.V.); Department of Cardiology, Medical University of Vienna, Austria (G.G.); Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Norway (I.U.N.); and Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (T.A., V.F.)
| |
Collapse
|
35
|
Wang YP, Xie Y, Ma H, Su SA, Wang YD, Wang JA, Xiang MX. Regulatory T lymphocytes in myocardial infarction: A promising new therapeutic target. Int J Cardiol 2016; 203:923-8. [DOI: 10.1016/j.ijcard.2015.11.078] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 10/21/2015] [Accepted: 11/08/2015] [Indexed: 12/31/2022]
|
36
|
miR-223 Inhibits Lipid Deposition and Inflammation by Suppressing Toll-Like Receptor 4 Signaling in Macrophages. Int J Mol Sci 2015; 16:24965-82. [PMID: 26492242 PMCID: PMC4632784 DOI: 10.3390/ijms161024965] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 09/04/2015] [Accepted: 09/20/2015] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis and its complications rank as the leading cause of death with the hallmarks of lipid deposition and inflammatory response. MicroRNAs (miRNAs) have recently garnered increasing interests in cardiovascular disease. In this study, we investigated the function of miR-223 and the underlying mechanism in atherosclerosis. In the atherosclerotic ApoE−/− mice models, an obvious increase of miR-223 was observed in aortic atherosclerotic lesions. In lipopolysaccharide (LPS) activated macrophages, its expression was decreased. The miR-223 overexpression significantly attenuated macrophage foam cell formation, lipid accumulation and pro-inflammatory cytokine production, which were reversed by anti-miR-223 inhibitor transfection. Mechanism assay corroborated that miR-223 negatively regulated the activation of the toll-like receptor 4 (TLR4)-nuclear factor-κB (NF-κB) pathway. Pretreatment with a specific inhibitor of NF-κB (pyrrolidinedithiocarbamate, PDTC) strikingly abrogated miR-223 silence-induced lipid deposition and inflammatory cytokine production. Furthermore, PI3K/AKT was activated by miR-223 up-regulation. Pretreatment with PI3K/AKT inhibitor LY294002 strikingly ameliorated the inhibitory effects of miR-223 on the activation of TLR4 and p65, concomitant with the increase in lipid deposition and inflammatory cytokine production. Together, these data indicate that miR-223 up-regulation might abrogate the development of atherosclerosis by blocking TLR4 signaling through activation of the PI3K/AKT pathway, and provides a promising therapeutic avenue for the treatment of atherosclerosis.
Collapse
|
37
|
Goltz D, Huss S, Ramadori E, Büttner R, Diehl L, Meyer R. Immunomodulation by splenectomy or by FTY720 protects the heart against ischemia reperfusion injury. Clin Exp Pharmacol Physiol 2015. [DOI: 10.1111/1440-1681.12465] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- D Goltz
- Institute of Pathology; University of Bonn; Bonn Germany
| | - S Huss
- Institute of Pathology; University of Cologne; Cologne Germany
| | - E Ramadori
- Institute of Pathology; University of Bonn; Bonn Germany
| | - R Büttner
- Institute of Pathology; University of Cologne; Cologne Germany
| | - L Diehl
- Experimental Immunology and Hepatology; Centre for Experimental Medicine; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - R Meyer
- Institute of Physiology II; University of Bonn; Bonn Germany
| |
Collapse
|
38
|
Li HQ, Zhang Q, Chen L, Yin CS, Chen P, Tang J, Rong R, Li TT, Hu LQ. Captopril inhibits maturation of dendritic cells and maintains their tolerogenic property in atherosclerotic rats. Int Immunopharmacol 2015; 28:715-23. [DOI: 10.1016/j.intimp.2015.05.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 05/11/2015] [Accepted: 05/31/2015] [Indexed: 10/23/2022]
|
39
|
Yu XH, Zhang J, Zheng XL, Yang YH, Tang CK. Interferon-γ in foam cell formation and progression of atherosclerosis. Clin Chim Acta 2015; 441:33-43. [DOI: 10.1016/j.cca.2014.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 11/28/2014] [Accepted: 12/05/2014] [Indexed: 10/24/2022]
|
40
|
Wang M, Lu L, Liu Y, Gu G, Tao R. FTY720 attenuates hypoxia-reoxygenation-induced apoptosis in cardiomyocytes. Exp Mol Pathol 2014; 97:218-24. [PMID: 25034934 DOI: 10.1016/j.yexmp.2014.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 07/12/2014] [Indexed: 12/29/2022]
Abstract
FTY720, sphingosine 1 phosphate (S1P) receptor agonist, is a potent immunosuppressive agent. Numerous studies have documented a relationship between S1P and cardioprotection. We therefore hypothesized that a S1P analogue FTY720 would attenuate hypoxia/reoxygenation (H/R) induced cadiomyocyte apoptosis. H9C2 cardiomyocytes were employed to establish an in vitro model of H/R. Cells were treated or not with different doses of FTY720. Cell viability was measured by flow cytometry and TUNEL staining. Western blot was used to analyze downstream signaling pathway. We observed that FTY720 inhibits the expression of cleaved caspase-3 and activates both AKT and ERK1/2 pathways. AKT pathway can be blocked by MEK kinase inhibitor PD98059. ERK1/2 pathway can be blocked by the phosphoinositide-3 kinase inhibitor wortmannin. AKT and ERK1/2 activation can also be inhibited by S1P1/3 receptor antagonist VPC23019, Gi antagonist PTX. The protein levels of TNF-α and IL1ß were upregulated during hypoxia/reoxygenation and were attenuated by FTY720. We conclude that FTY720, via its cargo of S1P, can protect cardiomyocytes against hypoxia/reoxygenation injury. This effect is achieved by inhibiting caspase-3 expression, inflammatory cytokine levels and activating AKT and ERK1/2 signaling pathways. The prosurvival signal activation is dependent on S1P1, 3 subtype receptors and Gi protein.
Collapse
Affiliation(s)
- Min Wang
- Department of Cardiology, Rui Jin Hospital, Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Lu
- Department of Cardiology, Rui Jin Hospital, Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yehong Liu
- Department of Cardiology, Rui Jin Hospital, Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Gang Gu
- Department of Cardiology, Rui Jin Hospital, Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Rong Tao
- Department of Cardiology, Rui Jin Hospital, Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
41
|
Karliner JS. Arsonists and firefighters: the perpetual inflammatory civil war for survival. Circulation 2014; 129:2368-70. [PMID: 24710031 DOI: 10.1161/circulationaha.114.010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Joel S Karliner
- From the Cardiology Section, VA Medical Center and the Department of Medicine and the Cardiovascular Research Institute, University of California, San Francisco, CA.
| |
Collapse
|
42
|
Potì F, Simoni M, Nofer JR. Atheroprotective role of high-density lipoprotein (HDL)-associated sphingosine-1-phosphate (S1P). Cardiovasc Res 2014; 103:395-404. [PMID: 24891400 DOI: 10.1093/cvr/cvu136] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Numerous epidemiological studies documented an inverse relationship between plasma high-density lipoprotein (HDL) cholesterol levels and the extent of atherosclerotic disease. However, clinical interventions targeting HDL cholesterol failed to show clinical benefits with respect to cardiovascular risk reduction, suggesting that HDL components distinct from cholesterol may account for anti-atherogenic effects attributed to this lipoprotein. Sphingosine-1-phosphate (S1P)-a lysosphingolipid exerting its biological activity via binding to specific G protein-coupled receptors and regulating a wide array of biological responses in a variety of different organs and tissues including the cardiovascular system-has been identified as an integral constituent of HDL particles. In the present review, we discuss current evidence from epidemiological studies, experimental approaches in vitro, and animal models of atherosclerosis, suggesting that S1P contributes to atheroprotective effects exerted by HDL particles.
Collapse
Affiliation(s)
- Francesco Potì
- Department of Biomedical, Metabolic and Neural Sciences-Endocrinology Section, University of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences-Endocrinology Section, University of Modena and Reggio Emilia, Modena, Italy
| | - Jerzy-Roch Nofer
- Department of Biomedical, Metabolic and Neural Sciences-Endocrinology Section, University of Modena and Reggio Emilia, Modena, Italy Center for Laboratory Medicine, University Hospital Münster, Albert-Schweizer-Campus 1, Geb. A1, Münster D-48149, Germany
| |
Collapse
|