1
|
Zhou J, Lei Y, Zhang S, Qin R, Liu Y, Yi D. Identification of the potential role of S1PR1 in adult moyamoya disease based on multiple bioinformatics analysis and experimental verification. FASEB J 2025; 39:e70461. [PMID: 40105149 DOI: 10.1096/fj.202401445r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/28/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Moyamoya disease (MMD) is a chronic occlusive cerebrovascular ailment with a progressively rising incidence, yet its precise etiology and pathogenesis remain elusive. Adult MMD-related datasets GSE189993 and GSE157628 were procured from the GEO database for screening of differentially expressed genes (DEGs). Weighted gene co-expression network analysis (WGCNA) was employed to unveil the most significant module associated with MMD. Least absolute shrinkage and selection operator (LASSO) logistic regression was used to identify and validate diagnostic markers for MMD. CIBERSORT and ssGSEA analyses were conducted to estimate immune cell infiltration in MMD vessels. In vitro experiments were performed to validate the biological functions of Sphingosine-1-phosphate receptor 1 (S1PR1) in mouse aortic smooth muscle cells (MOVAS). A total of 436 DEGs were identified from GSE189993, comprising 202 up-regulated genes and 234 down-regulated genes. Within the green-yellow module, 87 genes overlapped with DEGs, and 6 genes were identified by Cytoscape as key factors in the pathophysiology of MMD, namely, platelet endothelial cell adhesion molecule 1 (PECAM1), von Willebrand factor (VWF), intercellular cell adhesion molecule 1 (ICAM1), vascular endothelial growth factor C (VEGFC), tissue-type plasminogen activator (PLAT), and S1PR1. Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses also showed that key genes were mainly involved in endothelial cells and cell adhesion-related biological function. About 13 diagnostic genes were obtained by the LASSO regression algorithm, with S1PR1 emerging as a hub gene demonstrating good diagnostic performance in both the test set and validation set. Finally, we validated that overexpression of S1PR1 spurred viability, proliferation, and cell division in mouse aortic smooth muscle cells (MOVAS) and human cerebral vascular smooth muscle cells (HCVSMC) by activating the PI3K/AKT signaling pathway. S1PR1 has been identified as a hub gene for MMD. S1PR1 overexpression has been linked to enhanced cell division and proliferation in vitro, suggesting its potential as a therapeutic target for adult MMD.
Collapse
Affiliation(s)
- Jiabin Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Zhongnan Hospital, Wuhan University, Wuhan, People's Republic of China
| | - Yu Lei
- Zhongnan Hospital, Wuhan University, Wuhan, People's Republic of China
| | - Shilin Zhang
- Naval Aviation University of Chinese People's Liberation Army, Yantai, People's Republic of China
| | - Renwu Qin
- The Third People's Hospital of Yichang, The Third People's Hospital of China Three Gorges University, Yichang, People's Republic of China
| | - Yuhan Liu
- Department of Gastroenterology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, People's Republic of China
| | - Dongye Yi
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
2
|
Mohammed S, Alvarado V, Jiang YP, Velazquez FN, Alexander ME, Alvarez FA, Lambadis D, Chiappone SB, Ostermeyer-Fay AG, Zhang L, Shamseddine AA, Canals D, Snider AJ, Lin RZ, Hannun YA, Clarke CJ. A Critical Role for Neutral Sphingomyelinase-2 in Doxorubicin-induced Cardiotoxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644150. [PMID: 40166217 PMCID: PMC11957120 DOI: 10.1101/2025.03.20.644150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Although Doxorubicin (Dox) is an effective chemotherapeutic, its clinical utility is limited by a cumulative dose-dependent cardiotoxicity. While mechanisms underlying this cardiotoxicity have been investigated, strategies targeting these pathways have had marginal effects or had potential to interfere with Dox's anti-cancer activity. Sphingolipids (SL) are central to the chemotherapy response in multiple cancers, yet comparatively little is known about their role in non-transformed tissue, and actionable SL targets have not been identified. Here, we identified the SL enzyme neutral sphingomyelinase-2 (nSMase2) as a crucial downstream effector of Dox that is critical for chronic Dox-induced cardiotoxicity. In vitro studies showed that Dox treatment induces nSMase2 mRNA, protein, activity, and Cer accumulation in cardiomyocytes (CM) but not in cardiac fibroblasts. Mechanistically, nSMase2 induction was downstream of Top2B and p53, two previously identified molecular regulators of Dox-induced cardiotoxicity. In vivo studies in a chronic Dox model of cardiotoxicity found that loss of nSMase2 activity-null fro/fro mice were significantly protected from Dox-induced cardiac damage, exhibiting maintained ejection fraction, fractional shortening, and reduced left ventricle mass compared to wild-type littermates. Biologically, nSMase2 was dispensable for Dox-induced cell death but was important for Dox-induced CM senescence both in vitro and in vivo . Microarray analysis identified the dual specificity phosphatase DUSP4 as a downstream target of nSMase2 in vitro in Dox-treated CMs and in vivo in the chronic Dox-treated heart. Taken together, these results establish nSMase2 as a key component of the DNA damage response pathway in CMs and define a critical role for nSMase2 as a SL mediator of Dox-induced cardiotoxicity through effects on CM senescence. In addition to cementing a role for SLs in Dox effects in normal tissue, this study further advances nSMase2 as a target of interest for cardioprotection.
Collapse
|
3
|
Benkhoff M, Polzin A. Lipoprotection in cardiovascular diseases. Pharmacol Ther 2024; 264:108747. [PMID: 39491757 DOI: 10.1016/j.pharmthera.2024.108747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/14/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Cardioprotection is a well-established term in the scientific world. It describes the protection of various mediators on the cardiovascular system. These protective effects can also be provided by certain lipids. Since lipids are a very specific and clearly definable class of substances, we define the term lipoprotection as lipid-mediated cardioprotection. In this review, we highlight high-density lipoprotein (HDL), sphingosine-1-phosphate (S1P) and omega-3 polyunsaturated fatty acids (n-3 PUFA) as the most important lipoprotective mediators and show their beneficial impact on coronary artery disease (CAD), acute myocardial infarction (AMI) and heart failure (HF).
Collapse
Affiliation(s)
- Marcel Benkhoff
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany.
| |
Collapse
|
4
|
Alherz FA, Saleh A, Alsheikh MY, Borg HM, Kabel AM, Abd Elmaaboud MA. Shikonin mitigates cyclophosphamide-induced cardiotoxicity in mice: the role of sirtuin-1, NLRP3 inflammasome, autophagy, and apoptosis. J Pharm Pharmacol 2024; 76:1482-1496. [PMID: 39245439 DOI: 10.1093/jpp/rgae119] [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: 05/16/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVES The aim of this study was to elucidate the protective potential of shikonin (SHK) on cyclophosphamide (CP)-induced cardiotoxicity in Swiss albino mice. METHODS Mice received SHK in three different doses by oral gavage daily for 14 days and CP at 100 mg/kg, intraperitoneally once on the seventh day. On the 15th day, mice were euthanized, blood collected, and hearts were removed to estimate various biochemical and histopathological parameters. KEY FINDINGS CP significantly increased serum lactate dehydrogenase, creatine kinase-MB, troponin I and NT pro-BNP, and cardiac malondialdehyde and decreased cardiac total antioxidant capacity and Nrf2, whereas increased inflammatory markers in the cardiac tissues. CP also caused hypertrophy and fibrosis in the cardiac tissues via activation of IL6/JAK2/STAT3 while depressed SIRT1 and PI3K/p-Akt pathway with consequent increased apoptosis and dysregulation of autophagy. SHK treatment reversed these changes in a dose-dependent manner and showed a significant protective effect against CP-induced cardiotoxicity via suppressing oxidative stress, inflammation, and apoptosis with modulation of autophagy via induction of SIRT1/PI3K/p-Akt signaling. CONCLUSIONS Shikonin may be used as an adjuvant to cyclophosphamide in cancer treatment, but further research is needed to investigate its effects on cardiotoxicity in distinct animal cancer models.
Collapse
Affiliation(s)
- Fatemah A Alherz
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mona Y Alsheikh
- Pharmacy Practice Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah 22254-2265, Saudi Arabia
| | - Hany M Borg
- Physiology Department, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt
| | - Ahmed M Kabel
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | | |
Collapse
|
5
|
Phan F, Bourron O, Foufelle F, Le Stunff H, Hajduch E. Sphingosine-1-phosphate signalling in the heart: exploring emerging perspectives in cardiopathology. FEBS Lett 2024; 598:2641-2655. [PMID: 38965662 DOI: 10.1002/1873-3468.14973] [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: 03/18/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
Cardiometabolic disorders contribute to the global burden of cardiovascular diseases. Emerging sphingolipid metabolites like sphingosine-1-phosphate (S1P) and its receptors, S1PRs, present a dynamic signalling axis significantly impacting cardiac homeostasis. S1P's intricate mechanisms extend to its transportation in the bloodstream by two specific carriers: high-density lipoprotein particles and albumin. This intricate transport system ensures the accessibility of S1P to distant target tissues, influencing several physiological processes critical for cardiovascular health. This review delves into the diverse functions of S1P and S1PRs in both physiological and pathophysiological conditions of the heart. Emphasis is placed on their diverse roles in modulating cardiac health, spanning from cardiac contractility, angiogenesis, inflammation, atherosclerosis and myocardial infarction. The intricate interplays involving S1P and its receptors are analysed concerning different cardiac cell types, shedding light on their respective roles in different heart diseases. We also review the therapeutic applications of targeting S1P/S1PRs in cardiac diseases, considering existing drugs like Fingolimod, as well as the prospects and challenges in developing novel therapies that selectively modulate S1PRs.
Collapse
Affiliation(s)
- Franck Phan
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (APHP), La Pitié-Salpêtrière-Charles Foix University Hospital, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Olivier Bourron
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Diabetology Department, Assistance Publique-Hôpitaux de Paris (APHP), La Pitié-Salpêtrière-Charles Foix University Hospital, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Fabienne Foufelle
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Hervé Le Stunff
- Institut des Neurosciences Paris-Saclay, CNRS UMR 9197, Université Paris-Saclay, France
| | - Eric Hajduch
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| |
Collapse
|
6
|
Xu X, Li S, Wang T, Zhen P, Wei Q, Yu F, Tong J. Mitigation of myocardial ischemia/reperfusion-induced chronic heart failure via Shexiang Baoxin Pill-mediated regulation of the S1PR1 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155390. [PMID: 38569296 DOI: 10.1016/j.phymed.2024.155390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/12/2024] [Accepted: 01/24/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Well-defined and effective pharmacological interventions for clinical management of myocardial ischemia/reperfusion (MI/R) injury are currently unavailable. Shexiang Baoxin Pill (SBP), a traditional Chinese medicine Previous research on SBP has been confined to single-target treatments for MI/R injury, lacking a comprehensive examination of various aspects of MI/R injury and a thorough exploration of its underlying mechanisms. PURPOSE This study aimed to investigate the therapeutic potential of SBP for MI/R injury and its preventive effects on consequent chronic heart failure (CHF). Furthermore, we elucidated the specific mechanisms involved, contributing valuable insights into the potential pharmacological interventions for the clinical treatment of MI/R injury. METHODS We conducted a comprehensive identification of SBP components using high-performance liquid chromatography. Subsequently, we performed a network pharmacology analysis based on the identification results, elucidating the key genes influenced by SBP. Thereafter, through bioinformatics analysis of the key genes and validation through mRNA and protein assays, we ultimately determined the centralized upstream targets. Lastly, we conducted in vitro experiments using myocardial and endothelial cells to elucidate and validate potential underlying mechanisms. RESULTS SBP can effectively mitigate cell apoptosis, oxidative stress, and inflammation, as well as promote vascular regeneration following MI/R, resulting in improved cardiac function and reduced CHF risk. Mechanistically, SBP treatment upregulates sphingosine-1-phosphate receptor 1 (S1PR1) expression and activates the S1PR1 signaling pathway, thereby regulating the expression of key molecules, including phosphorylated Protein Kinase B (AKT), phosphorylated signal transducer and activator of transcription 3, epidermal growth factor receptor, vascular endothelial growth factor A, tumor necrosis factor-α, and p53. CONCLUSION This study elucidated the protective role of SBP in MI/R injury and its potential to reduce the risk of CHF. Furthermore, by integrating downstream effector proteins affected by SBP, this research identified the upstream effector protein S1PR1, enhancing our understanding of the pharmacological characteristics and mechanisms of action of SBP. The significance of this study lies in providing compelling evidence for the use of SBP as a traditional Chinese medicine for MI/R injury and consequent CHF prevention.
Collapse
Affiliation(s)
- Xuan Xu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing 210096, PR China; School of medicine, Southeast University, Nanjing 210096, PR China
| | - Shengnan Li
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing 210096, PR China; School of medicine, Southeast University, Nanjing 210096, PR China
| | - Tao Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, PR China
| | - Penghao Zhen
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing 210096, PR China; School of medicine, Southeast University, Nanjing 210096, PR China
| | - Qin Wei
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing 210096, PR China
| | - Fuchao Yu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing 210096, PR China.
| | - Jiayi Tong
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, 87 Dingjiaqiao, Nanjing 210096, PR China; School of medicine, Southeast University, Nanjing 210096, PR China.
| |
Collapse
|
7
|
Zhu N, Li T, Bai Y, Sun J, Guo J, Yuan H, Shan Z. Targeting myocardial inflammation: investigating the therapeutic potential of atrial natriuretic peptide in atrial fibrosis. Mol Biol Rep 2024; 51:506. [PMID: 38622341 PMCID: PMC11018689 DOI: 10.1007/s11033-024-09393-w] [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: 12/18/2023] [Accepted: 02/28/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Atrial Fibrillation (AF), a prevalent arrhythmic condition, is intricately associated with atrial fibrosis, a major pathological contributor. Central to the development of atrial fibrosis is myocardial inflammation. This study focuses on Atrial Natriuretic Peptide (ANP) and its role in mitigating atrial fibrosis, aiming to elucidate the specific mechanisms by which ANP exerts its effects, with an emphasis on fibroblast dynamics. METHODS AND RESULTS The study involved forty Sprague-Dawley rats, divided into four groups: control, Angiotensin II (Ang II), Ang II + ANP, and ANP only. The administration of 1 µg/kg/min Ang II was given to Ang II and Ang II + ANP groups, while both Ang II + ANP and ANP groups received 0.1 µg/kg/min ANP intravenously for a duration of 14 days. Cardiac fibroblasts were used for in vitro validation of the proposed mechanisms. The study observed that rats in the Ang II and Ang II + ANP groups showed an increase in blood pressure and a decrease in body weight, more pronounced in the Ang II group. Diastolic dysfunction, a characteristic of the Ang II group, was alleviated by ANP. Additionally, ANP significantly reduced Ang II-induced atrial fibrosis, myofibroblast proliferation, collagen overexpression, macrophage infiltration, and the elevated expression of Interleukin 6 (IL-6) and Tenascin-C (TN-C). Transcriptomic sequencing indicated enhanced PI3K/Akt signaling in the Ang II group. Furthermore, in vitro studies showed that ANP, along with the PI3K inhibitor LY294002, effectively reduced PI3K/Akt pathway activation and the expression of TN-C, collagen-I, and collagen-III, which were induced by Ang II. CONCLUSIONS The study demonstrates ANP's potential in inhibiting myocardial inflammation and reducing atrial fibrosis. Notably, ANP's effect in countering atrial fibrosis seems to be mediated through the suppression of the Ang II-induced PI3K/Akt-Tenascin-C signaling pathway. These insights enhance our understanding of AF pathogenesis and position ANP as a potential therapeutic agent for treating atrial fibrosis.
Collapse
Affiliation(s)
- Nana Zhu
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Tianlun Li
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Yili Bai
- Southern Medical Branch, Chinese PLA General Hospital, Beijing, China
| | - Jiao Sun
- Graduate School, Medical School of Chinese PLA, Beijing, China
| | - Jianping Guo
- Department of Cardiovascular Medicine, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hongtao Yuan
- Department of Cardiovascular Medicine, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhaoliang Shan
- Department of Cardiovascular Medicine, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
8
|
Liu J, Liu X, Luo Y, Huang F, Xie Y, Zheng S, Jia B, Xiao Z. Sphingolipids: drivers of cardiac fibrosis and atrial fibrillation. J Mol Med (Berl) 2024; 102:149-165. [PMID: 38015241 PMCID: PMC10858135 DOI: 10.1007/s00109-023-02391-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/13/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023]
Abstract
Sphingolipids (SLs) are vital constituents of the plasma membrane of animal cells and concurrently regulate numerous cellular processes. An escalating number of research have evinced that SLs assume a crucial part in the progression of tissue fibrosis, a condition for which no efficacious cure exists as of now. Cardiac fibrosis, and in particular, atrial fibrosis, is a key factor in the emergence of atrial fibrillation (AF). AF has become one of the most widespread cardiac arrhythmias globally, with its incidence continuing to mount, thereby propelling it to the status of a major public health concern. This review expounds on the structure and biosynthesis pathways of several pivotal SLs, the pathophysiological mechanisms of AF, and the function of SLs in cardiac fibrosis. Delving into the influence of sphingolipid levels in the alleviation of cardiac fibrosis offers innovative therapeutic strategies to address cardiac fibrosis and AF.
Collapse
Affiliation(s)
- Junjie Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ximao Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yucheng Luo
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangze Huang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Xie
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Al-Kuraishy HM, Batiha GES, Al-Gareeb AI, Al-Harcan NAH, Welson NN. Receptor-dependent effects of sphingosine-1-phosphate (S1P) in COVID-19: the black side of the moon. Mol Cell Biochem 2023; 478:2271-2279. [PMID: 36652045 PMCID: PMC9848039 DOI: 10.1007/s11010-023-04658-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023]
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection leads to hyper-inflammation and amplified immune response in severe cases that may progress to cytokine storm and multi-organ injuries like acute respiratory distress syndrome and acute lung injury. In addition to pro-inflammatory cytokines, different mediators are involved in SARS-CoV-2 pathogenesis and infection, such as sphingosine-1-phosphate (S1P). S1P is a bioactive lipid found at a high level in plasma, and it is synthesized from sphingomyelin by the action of sphingosine kinase. It is involved in inflammation, immunity, angiogenesis, vascular permeability, and lymphocyte trafficking through G-protein coupled S1P receptors. Reduction of the circulating S1P level correlates with COVID-19 severity. S1P binding to sphingosine-1-phosphate receptor 1 (S1PR1) elicits endothelial protection and anti-inflammatory effects during SARS-CoV-2 infection, by limiting excessive INF-α response and hindering mitogen-activated protein kinase and nuclear factor kappa B action. However, binding to S1PR2 opposes the effect of S1PR1 with vascular inflammation, endothelial permeability, and dysfunction as the concomitant outcome. This binding also promotes nod-like receptor pyrin 3 (NLRP3) inflammasome activation, causing liver inflammation and fibrogenesis. Thus, higher expression of macrophage S1PR2 contributes to the activation of the NLRP3 inflammasome and the release of pro-inflammatory cytokines. In conclusion, S1PR1 agonists and S1PR2 antagonists might effectively manage COVID-19 and its severe effects. Further studies are recommended to elucidate the potential conflict in the effects of S1P in COVID-19.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Nasser A Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Baghdad, Iraq
| | - Nermeen N Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt.
| |
Collapse
|
10
|
Bermick J, Watson S, Lueschow S, McElroy SJ. The fetal response to maternal inflammation is dependent upon maternal IL-6 in a murine model. Cytokine 2023; 167:156210. [PMID: 37130487 PMCID: PMC10225346 DOI: 10.1016/j.cyto.2023.156210] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND The induction of maternal inflammation in mice leads to fetal injury that is believed to be IL-6 dependent. The fetal inflammatory response, defined by elevated fetal or amniotic fluid IL-6, has been described as a potential mechanism for subsequent fetal injury. The role of maternal IL-6 production and signaling in the fetal IL-6 response is currently unclear. METHODS Genetic and anti-IL-6 antibody strategies were used to systematically block the maternal IL-6 response during inflammation. Chorioamnionitis was induced using intraperitoneal injection of lipopolysaccharide (LPS) at mid gestation (E14.5) and late gestation (E18.5). This model was used in pregnant C57Bl/6 dams, IL6-/- dams, C57Bl/6 dams treated with anti-IL-6 (blocks both classical and trans-signaling) or anti-gp130 antibodies (blocks trans-signaling only) and IL6+/- dams. Six hours following LPS injection, maternal serum, placental tissue, amniotic fluid and fetal tissue or serum were collected. A bead-based multiplex assay was used to evaluate levels of IL-6, KC, IL-1β, TNF, IL-10, IL-22, IFN-γ, IL-13 and IL-17A. RESULTS Chorioamnionitis in C57Bl/6 dams was characterized by elevated maternal serum levels of IL-6, KC and IL-22 with litter loss during mid gestation. The fetal response to maternal inflammation in C57Bl/6 mice was primarily characterized by elevated levels of IL-6, KC and IL-22 in the placenta, amniotic fluid and fetus during both mid and late gestation. A global IL-6 knockout (IL6-/-) eradicated the maternal, placental, amniotic fluid and fetal IL-6 response to LPS during mid and late gestation and improved litter survival, while minimally influencing the KC or IL-22 responses. Blocking maternal classical IL-6 signaling in C57Bl/6 dams at the time of LPS exposure diminished the maternal, placental, amniotic fluid and fetal IL-6 response during mid and late gestation, while blocking maternal IL-6 trans-signaling only affected fetal IL-6 expression. To evaluate whether maternal IL-6 was crossing the placenta and reaching the fetus, IL-6+/- dams were utilized in the chorioamnionitis model. IL-6+/- dams mounted a systemic inflammatory response following injection with LPS, characterized by elevated IL-6, KC and IL-22. IL-6-/- pups born to IL6+/- dams had decreased amniotic fluid levels of IL-6 and undetectable levels of fetal IL-6 compared to IL-6+/+ littermate controls. CONCLUSION The fetal response to systemic maternal inflammation is dependent upon maternal IL-6 signaling, but maternal IL-6 is not crossing the placenta and reaching the fetus at detectable levels.
Collapse
Affiliation(s)
- Jennifer Bermick
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, IA, USA; Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA.
| | - Sarah Watson
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, USA; Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Kaiser Permanente, Roseville, CA, USA
| | - Shiloh Lueschow
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, IA, USA; Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA
| | - Steven J McElroy
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, IA, USA; Division of Neonatology, Department of Pediatrics, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
11
|
Wang N, Li JY, Zeng B, Chen GL. Sphingosine-1-Phosphate Signaling in Cardiovascular Diseases. Biomolecules 2023; 13:biom13050818. [PMID: 37238688 DOI: 10.3390/biom13050818] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is an important sphingolipid molecule involved in regulating cardiovascular functions in physiological and pathological conditions by binding and activating the three G protein-coupled receptors (S1PR1, S1PR2, and S1PR3) expressed in endothelial and smooth muscle cells, as well as cardiomyocytes and fibroblasts. It exerts its actions through various downstream signaling pathways mediating cell proliferation, migration, differentiation, and apoptosis. S1P is essential for the development of the cardiovascular system, and abnormal S1P content in the circulation is involved in the pathogenesis of cardiovascular disorders. This article reviews the effects of S1P on cardiovascular function and signaling mechanisms in different cell types in the heart and blood vessels under diseased conditions. Finally, we look forward to more clinical findings with approved S1PR modulators and the development of S1P-based therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Na Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Jing-Yi Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Gui-Lan Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
12
|
Polzin A, Dannenberg L, Benkhoff M, Barcik M, Keul P, Ayhan A, Weske S, Ahlbrecht S, Trojovsky K, Helten C, Haberkorn S, Flögel U, Zeus T, Müller T, Gräler MH, Kelm M, Levkau B. Sphingosine-1-phosphate improves outcome of no-reflow acute myocardial infarction via sphingosine-1-phosphate receptor 1. ESC Heart Fail 2022; 10:334-341. [PMID: 36217778 PMCID: PMC9871711 DOI: 10.1002/ehf2.14176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/03/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
AIMS Therapeutic options targeting post-ischaemic cardiac remodelling are sparse. The bioactive sphingolipid sphingosine-1-phosphate (S1P) reduces ischaemia/reperfusion injury. However, its impact on post-ischaemic remodelling independently of its infarct size (IS)-reducing effect is yet unknown and was addressed in this study. METHODS AND RESULTS Acute myocardial infarction (AMI) in mice was induced by permanent ligation of the left anterior descending artery (LAD). C57Bl6 were treated with the S1P lyase inhibitor 4-deoxypyridoxine (DOP) starting 7 days prior to AMI to increase endogenous S1P concentrations. Cardiac function and myocardial healing were assessed by cardiovascular magnetic resonance imaging (cMRI), murine echocardiography, histomorphology, and gene expression analysis. DOP effects were investigated in cardiomyocyte-specific S1P receptor 1 deficient (S1PR1 Cardio Cre+) and Cre- control mice and S1P concentrations measured by LC-MS/MS. IS and cardiac function did not differ between control and DOP-treated groups on day one after LAD-ligation despite fourfold increase in plasma S1P. In contrast, cardiac function was clearly improved and myocardial scar size reduced, respectively, on Day 21 in DOP-treated mice. The latter also exhibited smaller cardiomyocyte size and reduced embryonic gene expression. The benefit of DOP treatment was abolished in S1PR1 Cardio Cre+. CONCLUSIONS S1P improves cardiac function and myocardial healing post AMI independently of initial infarct size and accomplishes this via the cardiomyocyte S1PR1. Hence, in addition to its beneficial effects on I/R injury, S1PR1 may be a promising target in post-infarction myocardial remodelling as adjunctive therapy to revascularization as well as in patients not eligible for standard interventional procedures.
Collapse
Affiliation(s)
- Amin Polzin
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany,Division of Cardiology, Pulmonary Diseases and Vascular MedicineUniversity Hospital of DuesseldorfDüsseldorfGermany
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany,Division of Cardiology, Pulmonary Diseases and Vascular MedicineUniversity Hospital of DuesseldorfDüsseldorfGermany
| | - Marcel Benkhoff
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany
| | - Maike Barcik
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany
| | - Petra Keul
- Institute of Molecular Medicine III, University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Aysel Ayhan
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany
| | - Sarah Weske
- Institute of Molecular Medicine III, University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Samantha Ahlbrecht
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany
| | - Kajetan Trojovsky
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany
| | - Carolin Helten
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany,Division of Cardiology, Pulmonary Diseases and Vascular MedicineUniversity Hospital of DuesseldorfDüsseldorfGermany
| | - Sebastian Haberkorn
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany,Division of Cardiology, Pulmonary Diseases and Vascular MedicineUniversity Hospital of DuesseldorfDüsseldorfGermany
| | - Ulrich Flögel
- Department of Molecular CardiologyHeinrich Heine UniversityDüsseldorfGermany
| | - Tobias Zeus
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany,Division of Cardiology, Pulmonary Diseases and Vascular MedicineUniversity Hospital of DuesseldorfDüsseldorfGermany
| | - Tina Müller
- Department of Anesthesiology and Intensive CareUniversity Hospital JenaJenaGermany
| | - Markus H. Gräler
- Department of Anesthesiology and Intensive CareUniversity Hospital JenaJenaGermany
| | - Malte Kelm
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of DuesseldorfHeinrich Heine University of DuesseldorfMoorenstr. 540225DüsseldorfGermany,Division of Cardiology, Pulmonary Diseases and Vascular MedicineUniversity Hospital of DuesseldorfDüsseldorfGermany
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
| |
Collapse
|
13
|
Zhang H, Ren L, Shivnaraine RV. Targeting GPCRs to treat cardiac fibrosis. Front Cardiovasc Med 2022; 9:1011176. [PMID: 36277752 PMCID: PMC9582444 DOI: 10.3389/fcvm.2022.1011176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiac fibrosis occurs ubiquitously in ischemic heart failure, genetic cardiomyopathies, diabetes mellitus, and aging. It triggers myocardial stiffness, which impairs cardiac function, ultimately progressing to end-stage heart failure and increased mortality. Although several targets for anti-fibrotic therapies have been identified, including TGF-β and receptor tyrosine kinase, there is currently no FDA-approved drug specifically targeting cardiac fibrosis. G protein-coupled receptors (GPCRs) are integral, multipass membrane-bound receptors that exhibit diverse and cell-specific expression, offering novel and unrealized therapeutic targets for cardiac fibrosis. This review highlights the emerging roles of several GPCRs and briefly explores their downstream pathways that are crucial in cardiac fibrosis. We will not only provide an overview of the GPCRs expressed on cardiac fibroblasts that are directly involved in myofibroblast activation but also describe those GPCRs which contribute to cardiac fibrosis via indirect crosstalk mechanisms. We also discuss the challenges of identifying novel effective therapies for cardiac fibrosis and offer strategies to circumvent these challenges.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,*Correspondence: Hao Zhang
| | - Lu Ren
- Department of Medicine, Division of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | | |
Collapse
|
14
|
Tanaka S, Zheng S, Kharel Y, Fritzemeier RG, Huang T, Foster D, Poudel N, Goggins E, Yamaoka Y, Rudnicka KP, Lipsey JE, Radel HV, Ryuh SM, Inoue T, Yao J, Rosin DL, Schwab SR, Santos WL, Lynch KR, Okusa MD. Sphingosine 1-phosphate signaling in perivascular cells enhances inflammation and fibrosis in the kidney. Sci Transl Med 2022; 14:eabj2681. [PMID: 35976996 PMCID: PMC9873476 DOI: 10.1126/scitranslmed.abj2681] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Chronic kidney disease (CKD), characterized by sustained inflammation and progressive fibrosis, is highly prevalent and can eventually progress to end-stage kidney disease. However, current treatments to slow CKD progression are limited. Sphingosine 1-phosphate (S1P), a product of sphingolipid catabolism, is a pleiotropic mediator involved in many cellular functions, and drugs targeting S1P signaling have previously been studied particularly for autoimmune diseases. The primary mechanism of most of these drugs is functional antagonism of S1P receptor-1 (S1P1) expressed on lymphocytes and the resultant immunosuppressive effect. Here, we documented the role of local S1P signaling in perivascular cells in the progression of kidney fibrosis using primary kidney perivascular cells and several conditional mouse models. S1P was predominantly produced by sphingosine kinase 2 in kidney perivascular cells and exported via spinster homolog 2 (Spns2). It bound to S1P1 expressed in perivascular cells to enhance production of proinflammatory cytokines/chemokines upon injury, leading to immune cell infiltration and subsequent fibrosis. A small-molecule Spns2 inhibitor blocked S1P transport, resulting in suppression of inflammatory signaling in human and mouse kidney perivascular cells in vitro and amelioration of kidney fibrosis in mice. Our study provides insight into the regulation of inflammation and fibrosis by S1P and demonstrates the potential of Spns2 inhibition as a treatment for CKD and potentially other inflammatory and fibrotic diseases that avoids the adverse events associated with systemic modulation of S1P receptors.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA.,Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Shuqiu Zheng
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Russell G. Fritzemeier
- Department of Chemistry and Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Tao Huang
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Daniel Foster
- Department of Chemistry and Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Nabin Poudel
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Eibhlin Goggins
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Yusuke Yamaoka
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Kinga P. Rudnicka
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Jonathan E. Lipsey
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Hope V. Radel
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Sophia M. Ryuh
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Tsuyoshi Inoue
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Junlan Yao
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Diane L. Rosin
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Susan R. Schwab
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, NY, New York 10016, USA
| | - Webster L. Santos
- Department of Chemistry and Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Mark D. Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia 22903, USA.,Corresponding author.
| |
Collapse
|
15
|
Yan H, Zhao H, Yi SW, Zhuang H, Wang DW, Jiang JG, Shen GF. Sphingosine-1-Phosphate Protects Against the Development of Cardiac Remodeling via Sphingosine Kinase 2 and the S1PR2/ERK Pathway. Curr Med Sci 2022; 42:702-710. [PMID: 35963947 DOI: 10.1007/s11596-022-2600-x] [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: 09/19/2020] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Cardiac remodeling is a common pathological change in various cardiovascular diseases and can ultimately result in heart failure. Thus, there is an urgent need for more effective strategies to aid in cardiac protection. Our previous work found that sphingosine-1-phosphate (S1P) could ameliorate cardiac hypertrophy. In this study, we aimed to investigate whether S1P could prevent cardiac fibrosis and the associated mechanisms in cardiac remodeling. METHODS Eight-week-old male C57BL/6 mice were randomly divided into a sham, transverse aortic constriction (TAC) or a TAC+S1P treatment group. RESULTS We found that S1P treatment improved cardiac function in TAC mice and that the cardiac fibrosis ratio in the TAC+S1P group was significantly lower and was accompanied by a decrease in α-smooth muscle actin (α-SMA) and collagen type I (COL I) expression compared with the TAC group. We also found that one of the key S1P enzymes, sphingosine kinase 2 (SphK2), which was mainly distributed in cytoblasts, was downregulated in the cardiac remodeling case and recovered after S1P treatment in vivo and in vitro. In addition, our in vitro results showed that S1P treatment activated extracellular regulated protein kinases (ERK) phosphorylation mainly through the S1P receptor 2 (S1PR2) and spurred p-ERK transposition from the cytoplasm to cytoblast in H9c2 cells exposed to phenylephrine. CONCLUSION These findings suggest that SphK2 and the S1PR2/ERK pathway may participate in the anti-remodeling effect of S1P on the heart. This work therefore uncovers a novel potential therapy for the prevention of cardiac remodeling.
Collapse
Affiliation(s)
- Hui Yan
- Wuhan No. 4 Hospital, Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hu Zhao
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shao-Wei Yi
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhuang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dao-Wen Wang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Gang Jiang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gui-Fen Shen
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
16
|
Spampinato SF, Sortino MA, Salomone S. Sphingosine-1-phosphate and Sphingosine-1-phosphate receptors in the cardiovascular system: pharmacology and clinical implications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:95-139. [PMID: 35659378 DOI: 10.1016/bs.apha.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a lipid that binds and activates five distinct receptor subtypes, S1P1, S1P2, S1P3, S1P4, S1P5, widely expressed in different cells, tissues and organs. In the cardiovascular system these receptors have been extensively studied, but no drug acting on them has been approved so far for treating cardiovascular diseases. In contrast, a number of S1P receptor agonists are approved as immunomodulators, mainly for multiple sclerosis, because of their action on lymphocyte trafficking. This chapter summarizes the available information on S1P receptors in the cardiovascular system and discusses their potential for treating cardiovascular conditions and/or their role on the clinical pharmacology of drugs so far approved for non-cardiovascular conditions. Basic research has recently produced data useful to understand the molecular pharmacology of S1P and S1P receptors, regarding biased agonism, S1P storage, release and vehiculation and chaperoning by lipoproteins, paracrine actions, intracellular non-receptorial S1P actions. On the other hand, the approval of fingolimod and newer generation S1P receptor ligands as immunomodulators, provides information on a number of clinical observations on the impact of these drugs on cardiovascular system which need to be integrated with preclinical data. S1P receptors are potential targets for prevention and treatment of major cardiovascular conditions, including hypertension, myocardial infarction, heart failure and stroke.
Collapse
Affiliation(s)
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Science, University of Catania, Catania, Italy.
| |
Collapse
|
17
|
Advanced Glycation End Product Blocker Drugs Have a Great Potential to Prevent Diabetic Cardiomyopathy in an Animal Model of Diabetes Mellitus Type-2. Cardiovasc Ther 2022; 2022:7014680. [PMID: 35414826 PMCID: PMC8977315 DOI: 10.1155/2022/7014680] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 02/10/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Sphingosine 1 phosphate (S1P) is a product of the sphingosine kinase 1 (SphK1) enzyme. Increased S1P can lead to tissue fibrosis that is also one of the pathways for developing diabetic cardiomyopathy. Advanced glycation end products (AGEs) increase S1P in cells. The study is aimed at using aminoguanidine (AG) as an AGEs blocker drug to prevent diabetic cardiomyopathy. Materials and methods. 210 rats were enrolled in the study. Diabetes mellitus type-2 was induced, and rats were divided into AG treated diabetic and nondiabetic groups. The heart histology was assessed with Masson's trichrome and hematoxylin-eosin staining. Cardiac function was measured with transthoracic echocardiography. S1P level and SphK1 gene expression were measured by western-blot and RT-qPCR, respectively. Results Results showed that S1P level increases in diabetes, and its augmentation in cardiac tissue with K6PC-5 leads to cardiac fibrosis. 50 and 200 mg/kg of AG prevented cardiac fibrosis, but 100 mg/kg had no significant preventive effect. AG suppressed the SphK1 gene expression and reduced the fibrotic effect of S1P. AG preserved cardiac function by keeping ejection fraction and fractional shortening within the normal range in diabetic rats. Conclusion AG has a suppressor effect on SphK1 gene expression besides its AGEs blocker role. AG is a potential drug to use in diabetic patients for preventing the development of diabetic cardiomyopathy. Other drugs that have AGEs or S1P blocker effects are a good choice for diabetic cardiomyopathy prevention.
Collapse
|
18
|
Wang J, Goren I, Yang B, Lin S, Li J, Elias M, Fiocchi C, Rieder F. Review article: the sphingosine 1 phosphate/sphingosine 1 phosphate receptor axis - a unique therapeutic target in inflammatory bowel disease. Aliment Pharmacol Ther 2022; 55:277-291. [PMID: 34932238 PMCID: PMC8766911 DOI: 10.1111/apt.16741] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/03/2021] [Accepted: 12/09/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Ozanimod, a high selective sphingosine 1 phosphate (S1P) receptor (S1PR) 1/5 modulator was approved by the Food and Drug Administration for the treatment of adult patients with moderately to severely active ulcerative colitis. Additional S1PR modulators are being tested in clinical development programmes for both ulcerative colitis and Crohn's disease. AIM To provide an overview of advances in understanding S1PRs biology and summarise preclinical and clinical investigations of S1P receptor modulators in chronic inflammatory disease with special emphasis on inflammatory bowel diseases (IBD). METHODS We performed a narrative review using PubMed and ClinicalTrials.gov. RESULTS Through S1PRs, S1P regulates multiple cellular processes, including proliferation, migration, survival, and vascular barrier integrity. The S1PRs function of regulating lymphocyte trafficking is well known, but new functions of S1PRs expand our knowledge of S1PRs biology. Several S1PR modulators are in clinical development for both ulcerative colitis and Crohn's disease and have shown promise in phase II and III studies with ozanimod now being approved for ulcerative colitis. CONCLUSIONS S1P receptor modulators constitute a novel, promising, safe, and convenient strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Jie Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang 453003, Henan Province, China,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Idan Goren
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel, Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Bo Yang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Sinan Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiannan Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Michael Elias
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute; Cleveland Clinic Foundation, Cleveland, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute; Cleveland Clinic Foundation, Cleveland, USA
| |
Collapse
|
19
|
Li F, Li SS, Chen H, Zhao JZ, Hao J, Liu JM, Zu XG, Cui W. miR-320 accelerates chronic heart failure with cardiac fibrosis through activation of the IL6/STAT3 axis. Aging (Albany NY) 2021; 13:22516-22527. [PMID: 34582362 PMCID: PMC8507257 DOI: 10.18632/aging.203562] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 08/24/2021] [Indexed: 12/12/2022]
Abstract
Cardiac fibrosis could induce abnormal cardiac function and become a novel target for cardiac hypertrophy and chronic heart failure. MiRNA-320 is a crucial miRNA in cardiovascular disease, but it is poorly understood whether it plays a role in cardiac fibrosis pathogenesis. We aimed to identify the specific underlying mechanism of miR-320 in cardiac fibrosis and hypertrophic pathogenesis. In our study, the GEO datasets revealed that STAT3 was significantly highly expressed in cardiomyocyte lines. MiR-320 activation and STAT3 signaling pathways were statistically significantly connected. Furthermore, miR-320 was highly associated with cardiac fibrosis and hypertrophic disease. Interstitial fibrosis was observed in the mice subjected to TAC surgery, markedly enhanced in miR-320 mimics. Mechanistically, we revealed that miR-320 mimics aggravated the pressure overload and induced cardiac hypertrophy and fibrosis via the IL6/STAT3/PTEN axis. MiR-320 mimics accelerated cardiac hypertrophy and cardiac fibrosis via the IL6/STAT3/PTEN axis. These results suggest that targeting miR-320 may represent a potential therapeutic strategy for cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Fang Li
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Shan-Shan Li
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Hui Chen
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Jian-Zhi Zhao
- Department of Biochemistry and Molecular Biology, The Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Jie Hao
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Jin-Ming Liu
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Xiu-Guang Zu
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Wei Cui
- Department of Cardiology, The Second Hospital of Hebei Medical University and Hebei Institute of Cardiovascular Research, Shijiazhuang, Hebei 050011, PR China
| |
Collapse
|
20
|
Chen Z, Zhou D, Zhang X, Wu Q, Wu G. Diagnostic biomarkers and potential drug targets for coronary artery disease as revealed by systematic analysis of lncRNA characteristics. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1243. [PMID: 34532380 PMCID: PMC8421950 DOI: 10.21037/atm-21-3276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/02/2021] [Indexed: 12/27/2022]
Abstract
Background The expression profile of lncRNAs in coronary artery disease (CAD) patients has not yet been fully explored. Therefore, the current study aimed to investigate lncRNA-based prognostic biomarkers for CAD. Methods The expression profiles of lncRNA and messenger RNA (mRNA) were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed lncRNA (DElncRNAs) and DEmRNAs were identified from CAD and normal samples, and weighted gene co-expression network analysis (WGCNA) was conducted. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to investigate the principal functions of significantly dysregulated genes. The potential drugs of new CAD-specific genes were identified by network distance method. Receiver operating characteristic (ROC) was used to verify the classification performance of genes. Results A total of 512 differentially expressed genes (DEGs) and 308 DElncRNAs were identified from GSE113079 dataset to classify CAD samples. Through WGCNA co-expression analysis, 24 co-expression modules were obtained. A total of 187 DElncRNAs and 253 DEGs were determined from 7 modules correlated with CAD. Functional enrichment analysis showed that these DEGs were mainly related to inflammatory and immune-related pathways. Furthermore, 36 regulatory pairs of significantly shared micro RNAs (miRNAs) were identified as dysregulated lncRNA-mRNA (LRM-CAD), which contained 11 lncRNAs and 33 genes. Compared with a single lncRNA or gene, LRM-CAD showed stronger classification performance [average area under the curve (AUC) =0.958]. We screened 3 potential therapeutic drugs, DB09105, DB12371, and DB12612, a by binding drug-target gene interaction network. Molecular docking verified that the S1PR1 gene bound relatively closely to DB12371 and DB12612. The ROC analysis on external data sets showed that S1PR1, AC012640.4, and S1PR1-AC012640.4 could effectively distinguish CAD samples from control samples. Conclusions We provided a transcriptome overview of abnormally expressed lncRNAs in CAD patients and identified novel biomarkers for diagnosing CAD.
Collapse
Affiliation(s)
- Ziqi Chen
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Dawang Zhou
- Department of Emergency, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaocong Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qian Wu
- Department of Gerontology, Guangzhou First People's Hospital of South China University of Technology, Guangzhou, China
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| |
Collapse
|
21
|
Jiménez-Uribe AP, Gómez-Sierra T, Aparicio-Trejo OE, Orozco-Ibarra M, Pedraza-Chaverri J. Backstage players of fibrosis: NOX4, mTOR, HDAC, and S1P; companions of TGF-β. Cell Signal 2021; 87:110123. [PMID: 34438016 DOI: 10.1016/j.cellsig.2021.110123] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022]
Abstract
The fibrotic process could be easily defined as a pathological excess of extracellular matrix deposition, leading to disruption of tissue architecture and eventually loss of function; however, this process involves a complex network of several signal transduction pathways. Virtually almost all organs could be affected by fibrosis, the most affected are the liver, lung, skin, kidney, heart, and eyes; in all of them, the transforming growth factor-beta (TGF-β) has a central role. The canonical and non-canonical signal pathways of TGF-β impact the fibrotic process at the cellular and molecular levels, inducing the epithelial-mesenchymal transition (EMT) and the induction of profibrotic gene expression with the consequent increase in proteins such as alpha-smooth actin (α-SMA), fibronectin, collagen, and other extracellular matrix proteins. Recently, it has been reported that some molecules that have not been typically associated with the fibrotic process, such as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4), mammalian target of rapamycin (mTOR), histone deacetylases (HDAC), and sphingosine-1 phosphate (S1P); are critical in its development. In this review, we describe and discuss the role of these new players of fibrosis and the convergence with TGF-β signaling pathways, unveiling new insights into the panorama of fibrosis that could be useful for future therapeutic targets.
Collapse
Affiliation(s)
| | - Tania Gómez-Sierra
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| | - Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - Marisol Orozco-Ibarra
- Laboratorio de Neurobiología Molecular y Celular, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, Av. Insurgentes Sur # 3877, La Fama, Alcaldía Tlalpan, CP 14269 Ciudad de México, Mexico
| | - José Pedraza-Chaverri
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico.
| |
Collapse
|
22
|
Shakartalla SB, Alhumaidi RB, Shammout ODA, Al Shareef ZM, Ashmawy NS, Soliman SSM. Dyslipidemia in breast cancer patients increases the risk of SAR-CoV-2 infection. INFECTION GENETICS AND EVOLUTION 2021; 92:104883. [PMID: 33905884 PMCID: PMC8079327 DOI: 10.1016/j.meegid.2021.104883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/17/2021] [Accepted: 04/22/2021] [Indexed: 12/27/2022]
Abstract
Breast cancer (BC) is the most diagnosed and second leading cause of death among women worldwide. Elevated levels of lipids have been reported in BC patients. On the other hand, lipids play an important role in coronavirus infections including the newly emerged disease caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and designated COVID-19 by WHO. Cancer patients including BC have been reported to be at higher risk of SARS-CoV-2 infection, which is mostly attributed to the chronic immunosuppressive status of cancer patients along with the use of cytotoxic drugs. Here in this review, we highlighted the role of dyslipidemia associated with BC patients in the incidence and severity of SARS-CoV-2 infection. Elevated levels of lipids namely phospholipids, cholesterol, sphingolipids, and eicosanoids in the serum of BC patients and their re-localization to the alveolar spaces can increase susceptibility and/or severity due to SARA-CoV-2 infection. Therefore, manipulation of dyslipidemia in BC patients should be recommended as prophylactic and therapy against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sarra B Shakartalla
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; Faculty of Pharmacy, University of Gezira, P.O.Box. 21111, Wadmedani, Sudan
| | - Razan B Alhumaidi
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Ola D A Shammout
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Zainab M Al Shareef
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Naglaa S Ashmawy
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; Faculty of Pharmacy, Department of Pharmacognosy, Ain Shams University, 11566-Abbassia, Cairo, Egypt
| | - Sameh S M Soliman
- Research Institute for Medical and Health sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
23
|
Jozefczuk E, Szczepaniak P, Guzik TJ, Siedlinski M. Silencing of Sphingosine kinase 1 Affects Maturation Pathways in Mouse Neonatal Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22073616. [PMID: 33807180 PMCID: PMC8037404 DOI: 10.3390/ijms22073616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/18/2022] Open
Abstract
Sphingosine kinase-1 (Sphk1) and its product, sphingosine-1-phosphate (S1P) are important regulators of cardiac growth and function. Numerous studies have reported that Sphk1/S1P signaling is essential for embryonic cardiac development and promotes pathological cardiac hypertrophy in adulthood. However, no studies have addressed the role of Sphk1 in postnatal cardiomyocyte (CM) development so far. The present study aimed to assess the molecular mechanism(s) by which Sphk1 silencing might influence CMs development and hypertrophy in vitro. Neonatal mouse CMs were transfected with siRNA against Sphk1 or negative control, and subsequently treated with 1 µM angiotensin II (AngII) or a control buffer for 24 h. The results of RNASeq analysis revealed that diminished expression of Sphk1 significantly accelerated neonatal CM maturation by inhibiting cell proliferation and inducing developmental pathways in the stress (AngII-induced) conditions. Importantly, similar effects were observed in the control conditions. Enhanced maturation of Sphk1-lacking CMs was further confirmed by the upregulation of the physiological hypertrophy-related signaling pathway involving Akt and downstream glycogen synthase kinase 3 beta (Gsk3β) downregulation. In summary, we demonstrated that the Sphk1 silencing in neonatal mouse CMs facilitated their postnatal maturation in both physiological and stress conditions.
Collapse
Affiliation(s)
- Ewelina Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
| | - Tomasz Jan Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | - Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
- Correspondence:
| |
Collapse
|
24
|
Alborghetti M, Bellucci G, Gentile A, Calderoni C, Nicoletti F, Capra R, Salvetti M, Centonze D. Drugs used in the treatment of multiple sclerosis during COVID-19 pandemic: a critical viewpoint. Curr Neuropharmacol 2021; 20:107-125. [PMID: 33784961 PMCID: PMC9199540 DOI: 10.2174/1570159x19666210330094017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 11/22/2022] Open
Abstract
Since COVID-19 has emerged as a word public health problem, attention has been focused on how immune-suppressive drugs used for the treatment of autoimmune disorders influence the risk for SARS-CoV-2 infection and the development of acute respiratory distress syndrome (ARDS). Here, we discuss the disease-modifying agents approved for the treatment of multiple sclerosis (MS) within this context. Interferon (IFN)-β1a and -1b, which display antiviral activity, could be protective in the early stage of COVID-19 infection, although SARS-CoV-2 may have developed resistance to IFNs. However, in the hyperinflammation stage, IFNs may become detrimental by facilitating macrophage invasion in the lung and other organs. Glatiramer acetate and its analogues should not interfere with the development of COVID-19 and may be considered safe. Teriflunomide, a first-line oral drug used in the treatment of relapsing-remitting MS (RRMS), may display antiviral activity by depleting cellular nucleotides necessary for viral replication. The other first-line drug, dimethyl fumarate, may afford protection against SARS-CoV-2 by activating the Nrf-2 pathway and reinforcing the cellular defenses against oxidative stress. Concern has been raised regarding the use of second-line treatments for MS during the COVID-19 pandemic. However, this concern is not always justified. For example, fingolimod might be highly beneficial during the hyperinflammatory stage of COVID-19 for a number of mechanisms, including the reinforcement of the endothelial barrier. Caution is suggested for the use of natalizumab, cladribine, alemtuzumab, and ocrelizumab, although MS disease recurrence after discontinuation of these drugs may overcome a potential risk for COVID-19 infection.
Collapse
Affiliation(s)
- Marika Alborghetti
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS), University Sapienza of Rome. Italy
| | - Gianmarco Bellucci
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS), University Sapienza of Rome. Italy
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome. Italy
| | - Chiara Calderoni
- Departments of Physiology and Pharmacology, University Sapienza of Rome. Italy
| | | | - Ruggero Capra
- Multiple Sclerosis Center, ASST Ospedali Civili, Brescia. Italy
| | - Marco Salvetti
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS),University Sapienza of Rome. Italy
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome. Italy
| |
Collapse
|
25
|
Magaye RR, Savira F, Hua Y, Xiong X, Huang L, Reid C, Flynn BL, Kaye D, Liew D, Wang BH. Attenuating PI3K/Akt- mTOR pathway reduces dihydrosphingosine 1 phosphate mediated collagen synthesis and hypertrophy in primary cardiac cells. Int J Biochem Cell Biol 2021; 134:105952. [PMID: 33609744 DOI: 10.1016/j.biocel.2021.105952] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022]
Abstract
Cardiac fibrosis and myocyte hypertrophy play contributory roles in the progression of diseases such as heart Failure (HF) through what is collectively termed cardiac remodelling. The phosphoinositide 3- kinase (PI3K), protein kinase B (Akt) and mammalian target for rapamycin (mTOR) signalling pathway (PI3K/Akt- mTOR) is an important pathway in protein synthesis, cell growth, cell proliferation, and lipid metabolism. The sphingolipid, dihydrosphingosine 1 phosphate (dhS1P) has been shown to bind to high density lipids in plasma. Unlike its analog, spingosine 1 phosphate (S1P), the role of dhS1P in cardiac fibrosis is still being deciphered. This study was conducted to investigate the effect of dhS1P on PI3K/Akt signalling in primary cardiac fibroblasts and myocytes. Our findings demonstrate that inhibiting PI3K reduced collagen synthesis in neonatal cardiac fibroblasts (NCFs), and hypertrophy in neonatal cardiac myocytes (NCMs) induced by dhS1P, in vitro. Reduced activation of the PI3K/Akt- mTOR signalling pathway led to impaired translation of fibrotic proteins such as collagen 1 (Coll1) and transforming growth factor β (TGFβ) and inhibited the transcription and translation of tissue inhibitor of matrix metalloproteinase 1 (TIMP1). PI3K inhibition also affected the gene expression of S1P receptors and enzymes such as the dihydroceramide delta 4 desaturase (DEGS1) and sphingosine kinase 1 (SK1) in the de novo sphingolipid pathway. While in myocytes, PI3K inhibition reduced myocyte hypertrophy induced by dhS1P by reducing skeletal muscle α- actin (αSKA) mRNA expression, and protein translation due to increased glycogen synthase kinase 3β (GSK3β) mRNA expression. Our findings show a relationship between the PI3K/Akt- mTOR signalling cascade and exogenous dhS1P induced collagen synthesis and myocyte hypertrophy in primary neonatal cardiac cells.
Collapse
Affiliation(s)
- Ruth R Magaye
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Feby Savira
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Xiong
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; Shanghai Institute of Heart Failure, Research Centre for Translational Medicine, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai 200120, China
| | - Li Huang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia; School of Public Health School, Curtin University, Perth, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - David Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia
| | - Bing H Wang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in Therapeutics, Melbourne, Australia.
| |
Collapse
|
26
|
Bernacchioni C, Ciarmela P, Vannuzzi V, Greco S, Vannuccini S, Malentacchi F, Pellegrino P, Capezzuoli T, Sorbi F, Cencetti F, Bruni P, Donati C, Petraglia F. Sphingosine 1-phosphate signaling in uterine fibroids: implication in activin A pro-fibrotic effect. Fertil Steril 2021; 115:1576-1585. [PMID: 33500141 DOI: 10.1016/j.fertnstert.2020.12.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To explore the link between sphingosine 1-phosphate (S1P) signaling and leiomyoma and the possible S1P cross-talk with the fibrotic effect of activin A. DESIGN Case-control laboratory study. SETTING University institute and university hospital. PATIENT(S) Patients with uterine fibroids (n = 26). INTERVENTIONS(S) Tissue specimens of leiomyoma and normal myometrium were obtained from patients undergoing myomectomy or total hysterectomy. MAIN OUTCOME MEASURE(S) Expression of mRNA levels of the enzyme involved in S1P metabolism, S1P receptors, and S1P transporter Spns2 was evaluated in matched leiomyoma/myometrium specimens and cell populations. The effects of inhibition of S1P metabolism and signaling was evaluated on activin A-induced fibrotic action in leiomyoma cell lines. RESULT(S) The expression of the enzymes responsible for S1P formation, sphingosine kinase (SK) 1 and 2, and S1P2, S1P3, and S1P5 receptors was significantly augmented in leiomyomas compared with adjacent myometrium. In leiomyoma cells, but not in myometrial cells, activin A increased mRNA expression levels of SK1, SK2, and S1P2. The profibrotic action of activin A was abolished when SK1/2 were inhibited or S1P2/3 were blocked. Finally, S1P augmented by itself mRNA levels of fibrotic markers (fibronectin, collagen 1A1) and activin A in leiomyomas but not in myometrial cells. CONCLUSION(S) This study shows that S1P signaling is dysregulated in uterine fibroids and involved in activin A-induced fibrosis, opening new perspectives for uterine fibroid treatment.
Collapse
Affiliation(s)
- Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Pasquapina Ciarmela
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Valentina Vannuzzi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Stefania Greco
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Silvia Vannuccini
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Francesca Malentacchi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Pamela Pellegrino
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Tommaso Capezzuoli
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Flavia Sorbi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy.
| | - Felice Petraglia
- Department of Experimental and Clinical Biomedical Sciences "M. Serio," University of Florence, Florence, Italy
| |
Collapse
|
27
|
Cirillo F, Piccoli M, Ghiroldi A, Monasky MM, Rota P, La Rocca P, Tarantino A, D'Imperio S, Signorelli P, Pappone C, Anastasia L. The antithetic role of ceramide and sphingosine-1-phosphate in cardiac dysfunction. J Cell Physiol 2021; 236:4857-4873. [PMID: 33432663 DOI: 10.1002/jcp.30235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/27/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally and the number of cardiovascular patients, which is estimated to be over 30 million in 2018, represent a challenging issue for the healthcare systems worldwide. Therefore, the identification of novel molecular targets to develop new treatments is an ongoing challenge for the scientific community. In this context, sphingolipids (SLs) have been progressively recognized as potent bioactive compounds that play crucial roles in the modulation of several key biological processes, such as proliferation, differentiation, and apoptosis. Furthermore, SLs involvement in cardiac physiology and pathophysiology attracted much attention, since these molecules could be crucial in the development of CVDs. Among SLs, ceramide and sphingosine-1-phosphate (S1P) represent the most studied bioactive lipid mediators, which are characterized by opposing activities in the regulation of the fate of cardiac cells. In particular, maintaining the balance of the so-called ceramide/S1P rheostat emerged as an important novel therapeutical target to counteract CVDs. Thus, this review aims at critically summarizing the current knowledge about the antithetic roles of ceramide and S1P in cardiomyocytes dysfunctions, highlighting how the modulation of their metabolism through specific molecules, such as myriocin and FTY720, could represent a novel and interesting therapeutic approach to improve the management of CVDs.
Collapse
Affiliation(s)
- Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
| | | | - Paola Rota
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Paolo La Rocca
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Adriana Tarantino
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Sara D'Imperio
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy
| | - Paola Signorelli
- Department of Health Sciences, Biochemistry and Molecular Biology Laboratory, University of Milan, Milan, Italy
| | - Carlo Pappone
- Department of Arrhythmology, IRCCS Policlinico San Donato, Milan, Italy.,Faculty of Medicine and Surgery, University of Vita-Salute San Raffaele, Milan, Italy
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy.,Faculty of Medicine and Surgery, University of Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
28
|
Zhao LK, Zhao YB, Zhang PX. High-throughput metabolomics discovers metabolite biomarkers and insights the protective mechanism of schisandrin B on myocardial injury rats. J Sep Sci 2020; 44:717-725. [PMID: 33247873 DOI: 10.1002/jssc.202000875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Schisandrin B has been proved to possess anti-inflammatory and anti-endoplasmic effects, could improve cardiac function, inhibit apoptosis, and reduce inflammation after ischemic injury. However, the detailed metabolic mechanism and potential pathways of Schisandrin B effects on myocardial injury are unclear. Metabolomics could yield in-depth mechanistic insights and explore the potential therapeutic effect of natural products. In this study, the preparation of doxorubicin-induced myocardial injury rat model for evaluation of Schisandrin B on viral myocarditis sequelae related pathological changes and its mechanism. The metabolite profiling of myocardial injury rats was performed through ultra-high performance liquid chromatography combined with mass spectrometry combined with pattern recognition approaches and pathway analysis. A total of 15 metabolites (nine in positive ion mode and six in negative ion mode) were considered as potential biomarkers of myocardial injury, and these metabolites may correlate with the regulation of Schisandrin B treatment. A total of six metabolic pathways are closely related to Schisandrin B treatment, including glycerophospholipid metabolism, sphingolipid metabolism, purine metabolism, etc. This study revealed the potential biomarkers and metabolic network pathways of myocardial injury, and illuminated the protective mechanism of Schisandrin B on myocardial injury.
Collapse
Affiliation(s)
- Ling-Kun Zhao
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, P. R. China
| | - Yun-Bo Zhao
- First Affiliated Hospital, Jiamusi University, Jiamusi, Heilongjiang, P. R. China
| | - Peng-Xia Zhang
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, P. R. China
| |
Collapse
|
29
|
Donati C, Cencetti F, Bernacchioni C, Vannuzzi V, Bruni P. Role of sphingosine 1-phosphate signalling in tissue fibrosis. Cell Signal 2020; 78:109861. [PMID: 33253915 DOI: 10.1016/j.cellsig.2020.109861] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Fibrosis is characterized by the excessive accumulation of extracellular matrix components, leading to loss of tissue function in affected organs. Although the majority of fibrotic diseases have different origins, they have in common a persistent inflammatory stimulus and lymphocyte-monocyte interactions that determine the production of numerous fibrogenic cytokines. Treatment to contrast fibrosis is urgently needed, since some fibrotic diseases lead to systemic fibrosis and represent a major cause of death. In this article, the role of the bioactive sphingolipid sphingosine 1-phosphate (S1P) and its signalling pathway in the fibrosis of different tissue contexts is extensively reviewed, highlighting that it may represent an innovative and promising pharmacological therapeutic target for treating this devastating multifaceted disease. In multiple tissues S1P influences different aspects of fibrosis modulating the recruitment of inflammatory cells, as well as cell proliferation, migration and transdifferentiation into myofibroblasts, the cell type mainly involved in fibrosis development. Moreover, at the level of fibrotic lesions, S1P metabolism is profoundly influenced by multiple cross-talk with profibrotic mediators, such as transforming growth factor β, thus finely regulating the development of fibrosis. This article is part of a Special Issue entitled "Physiological and pathological roles of bioactive sphingolipids".
Collapse
Affiliation(s)
- Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy.
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| | - Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| | - Valentina Vannuzzi
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, viale GB Morgagni 50, 50134 Florence, Italy
| |
Collapse
|
30
|
Wolska-Gawron K, Bartosińska J, Rusek M, Kowal M, Raczkiewicz D, Krasowska D. Circulating miRNA-181b-5p, miRNA-223-3p, miRNA-210-3p, let 7i-5p, miRNA-21-5p and miRNA-29a-3p in patients with localized scleroderma as potential biomarkers. Sci Rep 2020; 10:20218. [PMID: 33214624 PMCID: PMC7678876 DOI: 10.1038/s41598-020-76995-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Localized scleroderma (LoSc) is a rare disease manifested by an inflammation and sclerosis of the skin. The latest studies focused on glycoprotein Krebs von den Lungen-6, surfactant protein-D, chemokine ligand 18 and dipeptidylpeptidase 4 as potential biomarkers of skin fibrosis in systemic scleroderma. Our study aimed to identify 6 miRNAs with elevated or decreased levels in 38 LoSc patients (31 females, 7 males) compared to healthy volunteers (HVs) and to correlate the selected miRNAs' serum levels with the severity and the clinical symptoms of LoSc and some laboratory parameters with the selected miRNAs' serum levels. The serum levels of miRNAs, i.e. miRNA-181b-5p, miRNA-223-3p, miRNA-21-5p, let 7i-5p, miRNA-29a-3p and miRNA-210-3p were significantly increased in the LoSc patients compared to the HVs. The level of let-7i increase in the female LoSc patients correlated negatively with BSA (r = - 0.355, p = 0.049) and mLoSSI (r = - 0.432, p = 0.015). Moreover, the female patients with inactive LoSc had significantly higher level of let-7i (2.68-fold on average) in comparison to those with active disease (p = 0.045). The exact role of those molecules has not been revealed in LoSc and a long-term longitudinal research is pivotal to confirm their prognostic value.
Collapse
Affiliation(s)
- Katarzyna Wolska-Gawron
- Department of Dermatology, Venerology and Paediatric Dermatology, The Medical University of Lublin, 20-081 Lublin 11 Staszica St, Lublin, Poland.
| | - Joanna Bartosińska
- Department of Cosmetology and Aesthetic Medicine, The Medical University of Lublin, Lublin, Poland
| | - Marta Rusek
- Department of Dermatology, Venerology and Paediatric Dermatology, The Medical University of Lublin, 20-081 Lublin 11 Staszica St, Lublin, Poland
- Department of Pathophysiology, The Medical University of Lublin, Lublin, Poland
| | - Małgorzata Kowal
- Department of Dermatology, Venerology and Paediatric Dermatology, The Medical University of Lublin, 20-081 Lublin 11 Staszica St, Lublin, Poland
| | - Dorota Raczkiewicz
- SGH Warsaw School of Economics, Collegium of Economic Analysis, Institute of Statistics and Demography, Warsaw, Poland
| | - Dorota Krasowska
- Department of Dermatology, Venerology and Paediatric Dermatology, The Medical University of Lublin, 20-081 Lublin 11 Staszica St, Lublin, Poland
| |
Collapse
|
31
|
Prakash H, Upadhyay D, Bandapalli OR, Jain A, Kleuser B. Host sphingolipids: Perspective immune adjuvant for controlling SARS-CoV-2 infection for managing COVID-19 disease. Prostaglandins Other Lipid Mediat 2020; 152:106504. [PMID: 33147503 PMCID: PMC7605809 DOI: 10.1016/j.prostaglandins.2020.106504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 01/12/2023]
Abstract
That Sphingolipid derivatives are promising drug candidates for the management of novel COVID-19 disease. C-1P based tailoring of Th1 effector immunity for the eradication of infection is a translationally viable approach and deserves immediate attention. That C-1P would promote the killing of infected cells and resolve infection in moderate to severely infected cases. Ceramide derivatives can be exploited as drug candidates for controlling SARS-CoV-2 against novel COVID-19 disease.
Sphingolipids are potent bioactive agents involved in the pathogenesis of various respiratory bacterial infections. To date, several sphingolipid derivatives are known, but S1P (Sphingosine-1-phosphate) and Ceramide are the best-studied sphingolipid derivatives in the context of human diseases. These are membrane-bound lipids that influence host-pathogen interactions. Based on these features, we believe that sphingolipids might control SARS-CoV-2 infection in the host. SARS-CoV-2 utilizes the ACE-II receptor (Angiotensin-converting enzyme II receptor) on epithelial cells for its entry and replication. Activation of the ACE-II receptor is indirectly associated with the activation of S1P Receptor 1 signaling which is associated with IL-6 driven fibrosis. This is expected to promote pathological responses during SARS-CoV-2 infection in COVID-19 cases. Given this, mitigating S1P signaling by application of either S1P Lyase (SPL) or S1P analog (Fingolimod / FTY720) seems to be potential approach for controlling these pathological outcomes. However, due to the immunosuppressive nature of FTY720, it can modulate hyper-inflammatory responses and only provide symptomatic relief, which may not be sufficient for controlling the novel COVID-19 infection. Since Th1 effector immune responses are essential for the clearance of infection, we believe that other sphingolipid derivatives like Cermaide-1 Phosphate with antiviral potential and adjuvant immune potential can potentially control SARS-CoV-2 infection in the host by its ability in enhancing autophagy and antigen presentation by DC to promote T cell response which can be helpful in controlling SARS-CoV-2 infection in novel COVID-19 patients.
Collapse
Affiliation(s)
- Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University, Noida, India.
| | - Dilip Upadhyay
- Amity Institute of Virology and Immunology, Amity University, Noida, India
| | | | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, India
| | - Burkhard Kleuser
- Institute of Nutritional Science, Department of Nutritional Toxicology, University of Potsdam Nuthetal, Germany
| |
Collapse
|
32
|
Toor D, Jain A, Kalhan S, Manocha H, Sharma VK, Jain P, Tripathi V, Prakash H. Tempering Macrophage Plasticity for Controlling SARS-CoV-2 Infection for Managing COVID-19 Disease. Front Pharmacol 2020; 11:570698. [PMID: 33178021 PMCID: PMC7596271 DOI: 10.3389/fphar.2020.570698] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Affiliation(s)
- Devinder Toor
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Noida, India
| | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, India
| | - Shivani Kalhan
- Department of Pathology, Government Institute of Medical Sciences, Greater Noida, India
| | - Harmesh Manocha
- Department of Microbiology, Government Institute of Medical Sciences, Greater Noida, India
| | - Vivek Kumar Sharma
- Department of Physiology, Government Institute of Medical Sciences, Greater Noida, India
| | - Payal Jain
- Department of Medicine, Government Institute of Medical Sciences, Greater Noida, India
| | - Vishwas Tripathi
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
33
|
Meacci E, Garcia-Gil M, Pierucci F. SARS-CoV-2 Infection: A Role for S1P/S1P Receptor Signaling in the Nervous System? Int J Mol Sci 2020; 21:E6773. [PMID: 32942748 PMCID: PMC7556035 DOI: 10.3390/ijms21186773] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/07/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023] Open
Abstract
The recent coronavirus disease (COVID-19) is still spreading worldwide. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the virus responsible for COVID-19, binds to its receptor angiotensin-converting enzyme 2 (ACE2), and replicates within the cells of the nasal cavity, then spreads along the airway tracts, causing mild clinical manifestations, and, in a majority of patients, a persisting loss of smell. In some individuals, SARS-CoV-2 reaches and infects several organs, including the lung, leading to severe pulmonary disease. SARS-CoV-2 induces neurological symptoms, likely contributing to morbidity and mortality through unknown mechanisms. Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid with pleiotropic properties and functions in many tissues, including the nervous system. S1P regulates neurogenesis and inflammation and it is implicated in multiple sclerosis (MS). Notably, Fingolimod (FTY720), a modulator of S1P receptors, has been approved for the treatment of MS and is being tested for COVID-19. Here, we discuss the putative role of S1P on viral infection and in the modulation of inflammation and survival in the stem cell niche of the olfactory epithelium. This could help to design therapeutic strategies based on S1P-mediated signaling to limit or overcome the host-virus interaction, virus propagation and the pathogenesis and complications involving the nervous system.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Viale GB Morgagni 50, 50134 Firenze, Italy;
- Interuniversity Institute of Myology, University of Firenze, 50134 Firenze, Italy
| | - Mercedes Garcia-Gil
- Unit of Physiology, Department of Biology, University of Pisa, via S. Zeno 31, 56127 Pisa, Italy;
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Viale GB Morgagni 50, 50134 Firenze, Italy;
- Interuniversity Institute of Myology, University of Firenze, 50134 Firenze, Italy
| |
Collapse
|
34
|
Ouyang J, Shu Z, Chen S, Xiang H, Lu H. The role of sphingosine 1-phosphate and its receptors in cardiovascular diseases. J Cell Mol Med 2020; 24:10290-10301. [PMID: 32803879 PMCID: PMC7521328 DOI: 10.1111/jcmm.15744] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/12/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
There are many different types of cardiovascular diseases, which impose a huge economic burden due to their extremely high mortality rates, so it is necessary to explore the underlying mechanisms to achieve better supportive and curative care outcomes. Sphingosine 1‐phosphate (S1P) is a bioactive lipid mediator with paracrine and autocrine activities that acts through its cell surface S1P receptors (S1PRs) and intracellular signals. In the circulatory system, S1P is indispensable for both normal and disease conditions; however, there are very different views on its diverse roles, and its specific relevance to cardiovascular pathogenesis remains elusive. Here, we review the synthesis, release and functions of S1P, specifically detail the roles of S1P and S1PRs in some common cardiovascular diseases, and then address several controversial points, finally, we focus on the development of S1P‐based therapeutic approaches in cardiovascular diseases, such as the selective S1PR1 modulator amiselimod (MT‐1303) and the non‐selective S1PR1 and S1PR3 agonist fingolimod, which may provide valuable insights into potential therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Ouyang
- Center for Experimental Medical Research, the Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhihao Shu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medical Research, the Third Xiangya Hospital of Central South University, Changsha, China
| | - Hongwei Lu
- Center for Experimental Medical Research, the Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
35
|
Magaye RR, Savira F, Hua Y, Xiong X, Huang L, Reid C, Flynn B, Kaye D, Liew D, Wang BH. Exogenous dihydrosphingosine 1 phosphate mediates collagen synthesis in cardiac fibroblasts through JAK/STAT signalling and regulation of TIMP1. Cell Signal 2020; 72:109629. [PMID: 32278008 DOI: 10.1016/j.cellsig.2020.109629] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 02/03/2023]
Abstract
Cardiac fibrosis and myocyte hypertrophy are hallmarks of the cardiac remodelling process in cardiomyopathies such as heart failure (HF). Dyslipidemia or dysregulation of lipids contribute to HF. The dysregulation of high density lipoproteins (HDL) could lead to altered levels of other lipid metabolites that are bound to it such as sphingosine-1- phosphate (S1P). Recently, it has been shown that S1P and its analogue dihydrosphingosine-1-phosphate (dhS1P) are bound to HDL in plasma. The effects of dhS1P on cardiac cells have been obscure. In this study, we show that extracellular dhS1P is able to increase collagen synthesis in neonatal rat cardiac fibroblasts (NCFs) and cause hypertrophy of neonatal cardiac myocytes (NCMs). The janus kinase/signal transducer and activator (JAK/STAT) signalling pathway was involved in the increased collagen synthesis by dhS1P, through sustained increase of tissue inhibitor of matrix metalloproteinase 1 (TIMP1). Extracellular dhS1P increased phosphorylation levels of STAT1 and STAT3 proteins, also caused an early increase in gene expression of transforming growth factor-β (TGFβ), and sustained increase in TIMP1. Inhibition of JAKs led to inhibition of TIMP1 and TGFβ gene and protein expression. We also show that dhS1P is able to cause NCM hypertrophy through S1P-receptor-1 (S1PR1) signalling which is opposite to that of its analogue, S1P. Taken together, our results show that dhS1P increases collagen synthesis in cardiac fibroblasts causing fibrosis through dhS1P-JAK/STAT-TIMP1 signalling.
Collapse
Affiliation(s)
- Ruth R Magaye
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia
| | - Feby Savira
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Xiong
- Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia; Shanghai Institute of Heart Failure, Research Centre for Translational Medicine, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai 200120, China
| | - Li Huang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia; School of Public Health School, Curtin University, Perth, Australia
| | - Bernard Flynn
- Australian Translational Medicinal Chemistry Facility, Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - David Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia
| | - Bing H Wang
- Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Monash Centre of Cardiovascular Research and Education in therapeutics, Melbourne, Australia.
| |
Collapse
|
36
|
Jozefczuk E, Guzik TJ, Siedlinski M. Significance of sphingosine-1-phosphate in cardiovascular physiology and pathology. Pharmacol Res 2020; 156:104793. [PMID: 32278039 DOI: 10.1016/j.phrs.2020.104793] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/07/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a signaling lipid, synthetized by sphingosine kinases (SPHK1 and SPHK2), that affects cardiovascular function in various ways. S1P signaling is complex, particularly since its molecular action is reliant on the differential expression of its receptors (S1PR1, S1PR2, S1PR3, S1PR4, S1PR5) within various tissues. Significance of this sphingolipid is manifested early in vertebrate development as certain defects in S1P signaling result in embryonic lethality due to defective vasculo- or cardiogenesis. Similar in the mature organism, S1P orchestrates both physiological and pathological processes occurring in the heart and vasculature of higher eukaryotes. S1P regulates cell fate, vascular tone, endothelial function and integrity as well as lymphocyte trafficking, thus disbalance in its production and signaling has been linked with development of such pathologies as arterial hypertension, atherosclerosis, endothelial dysfunction and aberrant angiogenesis. Number of signaling mechanisms are critical - from endothelial nitric oxide synthase through STAT3, MAPK and Akt pathways to HDL particles involved in redox and inflammatory balance. Moreover, S1P controls both acute cardiac responses (cardiac inotropy and chronotropy), as well as chronic processes (such as apoptosis and hypertrophy), hence numerous studies demonstrate significance of S1P in the pathogenesis of hypertrophic/fibrotic heart disease, myocardial infarction and heart failure. This review presents current knowledge concerning the role of S1P in the cardiovascular system, as well as potential therapeutic approaches to target S1P signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- E Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - T J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland; Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - M Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland; Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
37
|
Józefczuk E, Nosalski R, Saju B, Crespo E, Szczepaniak P, Guzik TJ, Siedlinski M. Cardiovascular Effects of Pharmacological Targeting of Sphingosine Kinase 1. Hypertension 2020; 75:383-392. [PMID: 31838904 PMCID: PMC7055939 DOI: 10.1161/hypertensionaha.119.13450] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/11/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023]
Abstract
High blood pressure is a risk factor for cardiovascular diseases. Ang II (angiotensin II), a key pro-hypertensive hormone, mediates target organ consequences such as endothelial dysfunction and cardiac hypertrophy. S1P (sphingosine-1-phosphate), produced by Sphk1 (sphingosine kinase 1), plays a pivotal role in the pathogenesis of hypertension and downstream organ damage, as it controls vascular tone and regulates cardiac remodeling. Accordingly, we aimed to examine if pharmacological inhibition of Sphk1 using selective inhibitor PF543 can represent a useful vasoprotective and cardioprotective anti-hypertensive strategy in vivo. PF543 was administered intraperitoneally throughout a 14-day Ang II-infusion in C57BL6/J male mice. Pharmacological inhibition of Sphk1 improved endothelial function of arteries of hypertensive mice that could be mediated via decrease in eNOS (endothelial nitric oxide synthase) phosphorylation at T495. This effect was independent of blood pressure. Importantly, PF543 also reduced cardiac hypertrophy (heart to body weight ratio, 5.6±0.2 versus 6.4±0.1 versus 5.9±0.2 mg/g; P<0.05 for Sham, Ang II+placebo, and Ang II+PF543-treated mice, respectively). Mass spectrometry revealed that PF543 elevated cardiac sphingosine, that is, Sphk1 substrate, content in vivo. Mechanistically, RNA-Seq indicated a decreased expression of cardiac genes involved in actin/integrin organization, S1pr1 signaling, and tissue remodeling. Indeed, downregulation of Rock1 (Rho-associated coiled-coil containing protein kinase 1), Stat3 (signal transducer and activator of transcription 3), PKC (protein kinase C), and ERK1/2 (extracellular signal-regulated kinases 1/2) level/phosphorylation by PF543 was observed. In summary, pharmacological inhibition of Sphk1 partially protects against Ang II-induced cardiac hypertrophy and endothelial dysfunction. Therefore, it may represent a promising target for harnessing residual cardiovascular risk in hypertension.
Collapse
Affiliation(s)
- Ewelina Józefczuk
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| | - Ryszard Nosalski
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Blessy Saju
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Eva Crespo
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Piotr Szczepaniak
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| | - Tomasz Jan Guzik
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasglow, United Kingdom (R.N., B.S., E.C., T.J.G.)
| | - Mateusz Siedlinski
- From the Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland (E.J., R.N., P.S., T.J.G., M.S.)
| |
Collapse
|
38
|
Diarte-Añazco EMG, Méndez-Lara KA, Pérez A, Alonso N, Blanco-Vaca F, Julve J. Novel Insights into the Role of HDL-Associated Sphingosine-1-Phosphate in Cardiometabolic Diseases. Int J Mol Sci 2019; 20:ijms20246273. [PMID: 31842389 PMCID: PMC6940915 DOI: 10.3390/ijms20246273] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023] Open
Abstract
Sphingolipids are key signaling molecules involved in the regulation of cell physiology. These species are found in tissues and in circulation. Although they only constitute a small fraction in lipid composition of circulating lipoproteins, their concentration in plasma and distribution among plasma lipoproteins appears distorted under adverse cardiometabolic conditions such as diabetes mellitus. Sphingosine-1-phosphate (S1P), one of their main representatives, is involved in regulating cardiomyocyte homeostasis in different models of experimental cardiomyopathy. Cardiomyopathy is a common complication of diabetes mellitus and represents a main risk factor for heart failure. Notably, plasma concentration of S1P, particularly high-density lipoprotein (HDL)-bound S1P, may be decreased in patients with diabetes mellitus, and hence, inversely related to cardiac alterations. Despite this, little attention has been given to the circulating levels of either total S1P or HDL-bound S1P as potential biomarkers of diabetic cardiomyopathy. Thus, this review will focus on the potential role of HDL-bound S1P as a circulating biomarker in the diagnosis of main cardiometabolic complications frequently associated with systemic metabolic syndromes with impaired insulin signaling. Given the bioactive nature of these molecules, we also evaluated its potential of HDL-bound S1P-raising strategies for the treatment of cardiometabolic disease.
Collapse
Affiliation(s)
- Elena M. G. Diarte-Añazco
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
| | - Karen Alejandra Méndez-Lara
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| | - Antonio Pérez
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei d’Endocrinologia, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Núria Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei d’Endocrinologia, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Francisco Blanco-Vaca
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| | - Josep Julve
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| |
Collapse
|
39
|
Ishimaru K, Yoshioka K, Kano K, Kurano M, Saigusa D, Aoki J, Yatomi Y, Takuwa N, Okamoto Y, Proia RL, Takuwa Y. Sphingosine kinase-2 prevents macrophage cholesterol accumulation and atherosclerosis by stimulating autophagic lipid degradation. Sci Rep 2019; 9:18329. [PMID: 31797978 PMCID: PMC6892873 DOI: 10.1038/s41598-019-54877-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is the major cause of ischemic coronary heart diseases and characterized by the infiltration of cholesterol-accumulating macrophages in the vascular wall. Although sphingolipids are implicated in atherosclerosis as both membrane components and lipid mediators, the precise role of sphingolipids in atherosclerosis remains elusive. Here, we found that genetic deficiency of sphingosine kinase-2 (SphK2) but not SphK1 aggravates the formation of atherosclerotic lesions in mice with ApoE deficiency. Bone marrow chimaera experiments show the involvement of SphK2 expressed in bone marrow-derived cells. In macrophages, deficiency of SphK2, a major SphK isoform in this cell type, results in increases in cellular sphingosine and ceramides. SphK2-deficient macrophages have increases in lipid droplet-containing autophagosomes and autolysosomes and defective lysosomal degradation of lipid droplets via autophagy with an impaired luminal acidic environment and proteolytic activity in the lysosomes. Transgenic overexpression of SphK1 in SphK2-deficient mice rescued aggravation of atherosclerosis and abnormalities of autophagosomes and lysosomes in macrophages with reductions of sphingosine, suggesting at least partial overlapping actions of two SphKs. Taken together, these results indicate that SphK2 is required for autophagosome- and lysosome-mediated catabolism of intracellular lipid droplets to impede the development of atherosclerosis; therefore, SphK2 may be a novel target for treating atherosclerosis.
Collapse
Affiliation(s)
- Kazuhiro Ishimaru
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan.
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan
| | - Kuniyuki Kano
- Department of Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, 980-8578, Japan
| | - Makoto Kurano
- Department of Laboratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, 113-0033, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, 980-8573, Japan
| | - Junken Aoki
- Department of Biochemistry, Graduate School of Pharmaceutical Science, Tohoku University, Sendai, 980-8578, Japan
| | - Yutaka Yatomi
- Department of Laboratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, 113-0033, Japan
| | - Noriko Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan.,Department of Health and Medical Sciences, Ishikawa Prefectural Nursing University, Kahoku, 929-1210, Japan
| | - Yasuo Okamoto
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, 20892, USA
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, 920-8640, Japan.
| |
Collapse
|
40
|
Abstract
GPCRs (G-protein [guanine nucleotide-binding protein]-coupled receptors) play a central physiological role in the regulation of cardiac function in both health and disease and thus represent one of the largest class of surface receptors targeted by drugs. Several antagonists of GPCRs, such as βARs (β-adrenergic receptors) and Ang II (angiotensin II) receptors, are now considered standard of therapy for a wide range of cardiovascular disease, such as hypertension, coronary artery disease, and heart failure. Although the mechanism of action for GPCRs was thought to be largely worked out in the 80s and 90s, recent discoveries have brought to the fore new and previously unappreciated mechanisms for GPCR activation and subsequent downstream signaling. In this review, we focus on GPCRs most relevant to the cardiovascular system and discuss traditional components of GPCR signaling and highlight evolving concepts in the field, such as ligand bias, β-arrestin-mediated signaling, and conformational heterogeneity.
Collapse
Affiliation(s)
- Jialu Wang
- From the Department of Medicine (J.W., C.G., H.A.R.)
| | | | - Howard A Rockman
- From the Department of Medicine (J.W., C.G., H.A.R.).,Department of Cell Biology (H.A.R.).,Department of Molecular Genetics and Microbiology (H.A.R.), Duke University Medical Center, Durham, NC
| |
Collapse
|
41
|
Cardiac Fibroblast p38 MAPK: A Critical Regulator of Myocardial Remodeling. J Cardiovasc Dev Dis 2019; 6:jcdd6030027. [PMID: 31394846 PMCID: PMC6787752 DOI: 10.3390/jcdd6030027] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 12/15/2022] Open
Abstract
The cardiac fibroblast is a remarkably versatile cell type that coordinates inflammatory, fibrotic and hypertrophic responses in the heart through a complex array of intracellular and intercellular signaling mechanisms. One important signaling node that has been identified involves p38 MAPK; a family of kinases activated in response to stress and inflammatory stimuli that modulates multiple aspects of cardiac fibroblast function, including inflammatory responses, myofibroblast differentiation, extracellular matrix turnover and the paracrine induction of cardiomyocyte hypertrophy. This review explores the emerging importance of the p38 MAPK pathway in cardiac fibroblasts, describes the molecular mechanisms by which it regulates the expression of key genes, and highlights its potential as a therapeutic target for reducing adverse myocardial remodeling.
Collapse
|
42
|
Sun L, Yu M, Zhou T, Zhang S, He G, Wang G, Gang X. Current advances in the study of diabetic cardiomyopathy: From clinicopathological features to molecular therapeutics (Review). Mol Med Rep 2019; 20:2051-2062. [PMID: 31322242 DOI: 10.3892/mmr.2019.10473] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/29/2019] [Indexed: 11/06/2022] Open
Abstract
The incidence of diabetes mellitus has become a major public health concern due to lifestyle alterations. Moreover, the complications associated with diabetes mellitus deeply influence the quality of life of patients. Diabetic cardiomyopathy (DC) is a type of diabetes mellitus complication characterized by functional and structural damage in the myocardium but not accompanied by coronary arterial disease. Currently, diagnosing and preventing DC is still a challenge for physicians due to its atypical symptoms. For this reason, it is necessary to summarize the current knowledge on DC, especially in regards to the underlying molecular mechanisms toward the goal of developing useful diagnostic approaches and effective drugs based on these mechanisms. There exist several review articles which have focused on these points, but there still remains a lot to learn from published studies. In this review, the features, diagnosis and molecular mechanisms of DC are reviewed. Furthermore, potential therapeutic and prophylactic drugs are discussed.
Collapse
Affiliation(s)
- Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ming Yu
- Department of Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tong Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Siwen Zhang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guangyu He
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
43
|
Deng S, Zhou X, Ge Z, Song Y, Wang H, Liu X, Zhang D. Exosomes from adipose-derived mesenchymal stem cells ameliorate cardiac damage after myocardial infarction by activating S1P/SK1/S1PR1 signaling and promoting macrophage M2 polarization. Int J Biochem Cell Biol 2019; 114:105564. [PMID: 31276786 DOI: 10.1016/j.biocel.2019.105564] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 01/11/2023]
Abstract
Exosomes derived from mesenchymal stem cells (MSCs) are known to participate in myocardial repair after myocardial infarction (MI), but the mechanism remains unclear. Here, we isolated exosomes from adipose-derived MSCs (ADSCs) and examined their effect on MI-induced cardiac damage. To examine the underlying mechanism, H9c2 cells, cardiac fibroblasts, and HAPI cells were used to study the effect of ADSC-exosomes on hypoxia-induced H9c2 apoptosis, TGF-β1-induced fibrosis of cardiac fibroblasts, and hypoxia-induced macrophage M1 polarization using qRT-PCR, western blot, ELISA, immunohistochemistry, immunofluorescence and flow cytometry. ADSC-exosome treatment mitigated MI-induced cardiac damage by suppressing cardiac dysfunction, cardiac apoptosis, cardiac fibrosis, and inflammatory responses in vitro and in vivo. In addition, ADSC-exosome treatment promoted macrophage M2 polarization. Further experiments found that S1P/SK1/S1PR1 signaling was involved in the ADSC-exosome-mediated myocardial repair. Silencing of S1PR1 reversed the inhibitory effect of ADSC-exosomes on MI-induced cardiac apoptosis and fibrosis in vitro. ADSC-exosome-induced macrophage M2 polarization was also reversed after downregulation of S1PR1 under hypoxia conditions, which promoted NFκB and TGF-β1 expression, and suppressed the MI-induced cardiac fibrosis and inflammatory response. In sum, these results indicate that ADSC-derived exosomes ameliorate cardiac damage after MI by activating S1P/SK1/S1PR1 signaling and promoting macrophage M2 polarization.
Collapse
Affiliation(s)
- Shengqiong Deng
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - Xianjin Zhou
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200135, China.
| | - Zhiru Ge
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - Yuting Song
- Ningxia Medical University, Ningxia, 750000, China; Sino-French Cooperative Central Lab, Shanghai Gongli Hospital, Secondary Military Medical University, Shanghai, 200135, China.
| | - Hairong Wang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - Xinghui Liu
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - Denghai Zhang
- Sino-French Cooperative Central Lab, Shanghai Gongli Hospital, Secondary Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
44
|
Eid RA, Alkhateeb MA, El-Kott AF, Eleawa SM, Zaki MSA, Alaboodi SA, Salem Al-Shudiefat AAR, Aldera H, Alnamar NM, Alassiri M, Khalil MA. A high-fat diet rich in corn oil induces cardiac fibrosis in rats by activating JAK2/STAT3 and subsequent activation of ANG II/TGF-1β/Smad3 pathway: The role of ROS and IL-6 trans-signaling. J Food Biochem 2019; 43:e12952. [PMID: 31368573 DOI: 10.1111/jfbc.12952] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/20/2022]
Abstract
This study compared the effect of low-fat diet (LFD) and high-fat diet rich in corn oil (HFD-CO) on left ventricular (LV) fibrosis in rats and examined their effect of angiotensin II (ANG II), JAK/STAT, and TGF-1β/smad3 pathways. As compared to LFD which didn't affect any of the measured parameters, HFD-CO-induced type 2 diabetes phenotype and increased LV collagen synthesis. Mechanistically, it increased LV levels of ROS, ANG II, ACE, IL-6, s-IL-6Rα, TGF-β1, Smad-3, and activities of JAK1/2 and STAT1/3. AG490, a JAK2 inhibitor, partially ameliorated these effect while Losartan, an AT1 inhibitor completely abolished collagen synthesis. However, with both treatments, levels of ANG II, IL-6, and s-IL-6Rα, and activity of JAK1/STAT3 remained high, all of which were normalized by co-administration of NAC or IL-6 neutralizing antibody. In conclusion: HFD-CO enhances LV collage synthesis by activation of JAK1/STAT3/ANG II/TGF-1β/smad3 pathway. PRACTICAL APPLICATIONS: We report that chronic consumption of a high-fat diet rich in corn oil (HFD-CO) induces diabetes mellitus phenotype 2 associated with left ventricular (LV) cardiac fibrosis in rats. The findings of this study show that HFD-CO, and through the increasing generation of ROS and IL-6 levels and shedding, could activate LV JAK1/2-STAT1/3 and renin-angiotensin system (RAS) signaling pathways, thus creating a positive feedback between the two which ultimately leads to activation of TGF-1β/Smad3 fibrotic pathway. Herein, we also report a beneficial effect of the antioxidant, NAC, or IL-6 neutralizing antibody in preventing such adverse effects of such HFD-CO. However, this presents a warning message to the current sudden increase in idiopathic cardiac disorders, especially with the big shift in our diets toward n-6 PUFA.
Collapse
Affiliation(s)
- Refaat A Eid
- Department of Clinica Pathology and Anatomy, College of Medicine, King Khalid University (KKU), Abha, Kingdom of Saudi Arabia
| | - Mahmoud A Alkhateeb
- Basic Medical Sciences (Physiology Section), College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Kingdom of Saudi Arabia
| | - Attalla Farag El-Kott
- Department of Biology, College of Science, King Khalid University (KKU), Abha, Kingdom of Saudi Arabia.,Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Samy M Eleawa
- Department of Applied Medical Sciences, College of Health Sciences, PAAET, Safat, Kuwait
| | - Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University (KKU), Abha, Kingdom of Saudi Arabia.,Department of Histology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sultan Abdullah Alaboodi
- Central laboratories, Huraymala General Hospital, Ministry of Health, Riyadh, Kingdom of Saudi Arabia
| | | | - Hussain Aldera
- Basic Medical Sciences (Physiology Section), College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Kingdom of Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Kingdom of Saudi Arabia
| | | | - Mohammed Alassiri
- Basic Medical Sciences (Physiology Section), College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Kingdom of Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Mohammad A Khalil
- Department of Basic Medical Sciences, College of Medicine, King Fahid Medical City (KFMC), Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
45
|
Alqarni I, Bassiouni YA, Badr AM, Ali RA. Telmisartan and/or chlorogenic acid attenuates fructose-induced non-alcoholic fatty liver disease in rats: Implications of cross-talk between angiotensin, the sphingosine kinase/sphingoine-1-phosphate pathway, and TLR4 receptors. Biochem Pharmacol 2019; 164:252-262. [PMID: 31004566 DOI: 10.1016/j.bcp.2019.04.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022]
Abstract
Renin-angiotensin-aldosterone system (RAS) has been implicated in non-alcoholic fatty liver disease (NAFLD); the most common cause of chronic liver diseases. There is accumulating evidence that altered TLR4 and Sphingosine kinase 1(SphK1)/sphingosine1phosphate (S1P) signaling pathways are key players in the pathogenesis of NAFLD. Cross talk of the sphingosine signaling pathway, toll-4 (TLR4) receptors, and angiotensin II was reported in various tissues. Therefore, the aim of this study was to define the contribution of these two pathways to the hepatoprotective effects of telmisartan and/or chlorogenic acid (CGA) in NAFLD. CGA is a strong antioxidant that was previously reported to inhibit angiotensin converting enzyme. Male Wistar rats were treated with either high-fructose, with or without telmisartan, CGA, telmisartan + CGA for 8 weeks. Untreated NAFL rats showed characteristics of NAFLD, as evidenced by significant increase in the body weight, insulin resistance, and serum hepatotoxicity markers (Alanine and Aspartate transaminases) and lipids as compared to the negative control group, in addition to characteristic histopathological alterations. Treatment with either telmisartan and/or CGA improved aforementioned parameters, in addition to upregulation of antioxidant enzymes (Superoxide dismutase and Glutathione peroxidase). Effect of inhibiting RAS on both sphingosine pathway and TLR4 was evident by the suppressing effect of telmisartan and/or CGA on high fructose-induced upregulation of hepatic SPK1 and S1P, in addition to concomitant up-regulation of Sphingosine-1-Phosphate receptor (S1PR)3 protein level and increased expression of S1PR1 and TLR4. As TLR4 and SPK/S1P signaling pathways play important roles in the progression of liver inflammation, the effect on sphingosine pathway and TLR4 was associated with decreased concentrations of inflammatory markers, enzyme kB kinase (IKK), nuclear factor-kB and tumor necrosis factor-α as compared to untreated NAFL group. In conclusion, the present data strongly suggests the cross-talk between angiotensin, the Sphingosine SPK/S1P Axis and TLR4 Receptors, and their role in the pathogenesis of fructose-induced NAFLD, and the protection afforded by drugs inhibiting RAS.
Collapse
Affiliation(s)
- Iman Alqarni
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11459, Saudi Arabia
| | - Yieldez A Bassiouni
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11459, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Egypt
| | - Amira M Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11459, Saudi Arabia; Department of Pharmacology and Toxicology, College of Pharmacy, Ain Shams University, Heliopolis, Cairo, Egypt.
| | - Rehab A Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11459, Saudi Arabia
| |
Collapse
|
46
|
Brodowski L, Zindler T, von Hardenberg S, Schröder-Heurich B, von Kaisenberg CS, Frieling H, Hubel CA, Dörk T, von Versen-Höynck F. Preeclampsia-Associated Alteration of DNA Methylation in Fetal Endothelial Progenitor Cells. Front Cell Dev Biol 2019; 7:32. [PMID: 30949477 PMCID: PMC6436196 DOI: 10.3389/fcell.2019.00032] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 02/25/2019] [Indexed: 01/06/2023] Open
Abstract
Objective The pregnancy complication preeclampsia represents an independent risk factor for cardiovascular disease. Our previous research shows a diminished function of fetal endothelial colony-forming cells (ECFC), a proliferative subgroup of endothelial progenitor cells (EPC) in preeclampsia. The aim of this study was to further investigate whether DNA methylation of fetal EPC is affected in preeclampsia. Methods The genomic methylation pattern of fetal ECFC from uncomplicated and preeclamptic pregnancies was compared for 865918 CpG sites, and genes were classified into gene networks. Low and advanced cell culture passages were compared to explore whether expansion of fetal ECFC in cell culture leads to changes in global methylation status and if methylation characteristics in preeclampsia are maintained with increasing passage. Results A differential methylation pattern of fetal ECFC from preeclampsia compared to uncomplicated pregnancy was detected for a total of 1266 CpG sites in passage 3, and for 2362 sites in passage 5. Key features of primary networks implicated by methylation differences included cell metabolism, cell cycle and transcription and, more specifically, genes involved in cell-cell interaction and Wnt signaling. We identified an overlap between differentially regulated pathways in preeclampsia and cardiovascular system development and function. Cell culture passages 3 and 5 showed similar gene network profiles, and 1260 out of 1266 preeclampsia-associated methylation changes detected in passage 3 were confirmed in passage 5. Conclusion Methylation modification caused by preeclampsia is stable and detectable even in higher cell culture passages. An epigenetically modified endothelial precursor may influence both normal morphogenesis and postnatal vascular repair capacity. Further studies on epigenetic modifications in complicated pregnancies are needed to facilitate development of EPC based therapies for cardiovascular alterations.
Collapse
Affiliation(s)
- Lars Brodowski
- Department of Obstetrics and Gynecology, Hannover Medical School, Hanover, Germany
| | - Tristan Zindler
- Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hanover, Germany
| | | | | | | | - Helge Frieling
- Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hanover, Germany
| | - Carl A Hubel
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Thilo Dörk
- Department of Obstetrics and Gynecology, Hannover Medical School, Hanover, Germany
| | | |
Collapse
|
47
|
Kitano T, Usui S, Takashima SI, Inoue O, Goten C, Nomura A, Yoshioka K, Okajima M, Kaneko S, Takuwa Y, Takamura M. Sphigosine-1-phosphate receptor 1 promotes neointimal hyperplasia in a mouse model of carotid artery injury. Biochem Biophys Res Commun 2019; 511:179-184. [DOI: 10.1016/j.bbrc.2019.02.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/08/2019] [Indexed: 11/28/2022]
|
48
|
Wang E, He X, Zeng M. The Role of S1P and the Related Signaling Pathway in the Development of Tissue Fibrosis. Front Pharmacol 2019; 9:1504. [PMID: 30687087 PMCID: PMC6338044 DOI: 10.3389/fphar.2018.01504] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue fibrosis, including pulmonary fibrosis, hepatic fibrosis, and cardiac fibrosis, is an important stage in the development of many diseases. It can lead to structural damage and dysfunction and even severe carcinogenesis or death. There is currently no effective method for the treatment of fibrosis. At present, the molecular mechanism of tissue fibrosis has not yet been fully elucidated, but many studies have demonstrated that it is involved in conveying the complex messages between fibroblasts and various cytokines. Sphingosine 1-phosphate (S1P) is a naturally bioactive sphingolipid. S1P and the related signaling pathways are important intracellular metabolic pathways involved in many life activities, including cell proliferation, differentiation, apoptosis, and cellular signal transduction. Increasing evidence suggests that S1P and its signaling pathways play an important role in the development of tissue fibrosis; however, the mechanisms of these effects have not yet been fully elucidated, and even the role of S1P and its signaling pathways are still controversial. This article focuses on the role of S1P and the related signaling pathways in the development of fibrosis of lung, liver, heart, and other tissues, with emphasis on the application of inhibitors of some of molecules in the pathway in clinical treatment of fibrosis diseases.
Collapse
Affiliation(s)
- Erjin Wang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xingxuan He
- Department of Human Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ming Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
49
|
Kurdi M, Zgheib C, Booz GW. Recent Developments on the Crosstalk Between STAT3 and Inflammation in Heart Function and Disease. Front Immunol 2018; 9:3029. [PMID: 30619368 PMCID: PMC6305745 DOI: 10.3389/fimmu.2018.03029] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/07/2018] [Indexed: 12/27/2022] Open
Abstract
The transcription factor STAT3 has a protective function in the heart. Until recently, the role of STAT3 in hypertension-induced cardiac hypertrophy was unsettled. Earlier studies revealed that global reduction of STAT3 activity reduced cardiac hypertrophy with hypertension, but caused a disruption of myofilaments and increased contractile dysfunction. However, newer studies with cardiomyocyte-specific deletion of STAT3 indicate that STAT3 does not cause cardiac hypertrophy with increased blood pressure. Rather, cardiac STAT3 is important for maintaining metabolic homeostasis, and loss of STAT3 in cardiomyocytes makes the heart more susceptible to chronic pathological insult, for example by disrupting glucose metabolism and protective signaling networks via the upregulation of certain microRNAs. This scenario has implications for understanding peripartum cardiomyopathy as well. In viral myocarditis, STAT3 opposes the initiation of the dilated phenotype by maintaining membrane integrity via the expression of dystrophin. STAT3 signaling was also found to attenuate myocarditis by polarizing macrophages to a less inflammatory phenotype. On the other hand, STAT3 contributes to immune-mediated myocarditis due to IL-6-induced complement component C3 production in the liver, as well as the differentiation of Th17 cells, which play a role in initiation and development of myocarditis. Besides canonical signaling pathways, unphosphorylated STAT3 (U-STAT3) and redox-activated STAT3 have been shown to couple to transcription in the heart. In addition, tissue signaling cytokines such as IL-22 and IL-17 have been proposed to have actions on the heart that involve STAT3, but are not fully defined. Understanding the novel and often protective aspects of STAT3 in the myocardium could lead to new therapeutic approaches to treat heart disease.
Collapse
Affiliation(s)
- Mazen Kurdi
- Faculty of Sciences, Department of Chemistry and Biochemistry, and The Laboratory of Experimental and Clinical Pharmacology, Lebanese University, Beirut, Lebanon
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, School of Medicine, University of Colorado Denver, Anschutz Medical Campus and Colorado Children's Hospital, Aurora, CO, United States
| | - George W. Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
50
|
Yuza K, Nakajima M, Nagahashi M, Tsuchida J, Hirose Y, Miura K, Tajima Y, Abe M, Sakimura K, Takabe K, Wakai T. Different Roles of Sphingosine Kinase 1 and 2 in Pancreatic Cancer Progression. J Surg Res 2018; 232:186-194. [PMID: 30463717 DOI: 10.1016/j.jss.2018.06.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/11/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pancreatic cancer is a disease with poor prognosis, and development of new treatments is necessary. Sphingosine-1-phosphate (S1P), a bioactive lipid mediator produced by sphingosine kinases (SphK1 and SphK2), plays a critical role in progression of many types of cancer. However, little is known about the role of sphingosine kinases in pancreatic cancer. This study investigated the roles of sphingosine kinases in pancreatic cancer progression. MATERIALS AND METHODS S1P levels in pancreatic cancer and noncancerous pancreatic tissue were measured in 10 patients. We generated PAN02 murine pancreatic cancer cell lines with a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system genes 9 (Cas9)-mediated deletion of SphK1 or SphK2 and assessed cell growth and migration. In an animal model, we assessed the survival of mice injected with PAN02 cells intraperitoneally. RESULTS S1P levels in the pancreatic cancer tissue were significantly higher than those in noncancerous tissue. SphK1 knockout (KO) cells showed greater proliferation and migration than wild type (WT) cells, and SphK2 KO cells showed less proliferation and migration than WT cells. Animal experiments showed that the survival of mice injected with SphK1 KO cells was significantly shorter than those injected with WT cells, and the survival of mice injected with SphK2 KO cells was longer than those injected with WT cells. Surprisingly, cytotoxic assay using gemcitabine showed that SphK1 KO cells survived less than WT cells, and SphK2 KO cells survived more than WT cells. CONCLUSIONS S1P produced by SphK1 and SphK2 may have different functions in pancreatic cancer cells. Targeting both SphK1 and SphK2 may be a potential strategy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Kizuki Yuza
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Masato Nakajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan.
| | - Junko Tsuchida
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Yuki Hirose
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Kohei Miura
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Yosuke Tajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata City, Niigata, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata City, Niigata, Japan
| | - Kazuaki Takabe
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan; Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York; Department of Surgery, University at Buffalo Jacobs School of Medicine and Biosciences, the State University of New York, Buffalo, New York
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| |
Collapse
|