1
|
Ismail A, Shadid HR, Huang Y, Hutten CG, Vasbinder A, Pizzo I, Catalan TC, Diaz KM, Kunkle P, Banerjee M, Rubenfire M, Brandt EJ, Williams G, Pop-Busui R, Hayek SS. Statin Therapy, Inflammation, and Outcomes in Patients Hospitalized for COVID-19: A Prospective Multicenter Cohort Study. Am J Med 2024; 137:1264-1271.e1. [PMID: 39179167 DOI: 10.1016/j.amjmed.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/16/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Statins are lipid-lowering agents with anti-inflammatory effects. Data surrounding the benefits of statins in patients with coronavirus disease 2019 (COVID-19) are conflicting. We sought to better understand the impact of statins in the context of COVID-19-related inflammation. METHODS We leveraged the International Study of Inflammation in COVID-19, a prospective multicenter cohort of patients hospitalized for COVID-19 between February 2020 and October 2022. Participants underwent systematic assessment of biomarkers of inflammation. We used logistic regression modeling and inverse probability-of-treatment weighting (IPTW) to examine the association between prior statin use and the composite outcome of in-hospital death, need for mechanical ventilation, and need for renal replacement therapy. RESULTS A total of 4464 patients were included in the study, of whom 1364 (27.5%) were taking a statin prior to admission. There were 1061 primary outcome events, including 540 deaths, 854 mechanical ventilation and 313 renal replacement therapy. Amongst biomarkers of inflammation, statin use was associated solely with lower levels of soluble urokinase plasminogen activator receptor (suPAR) after adjusting for known confounders. In multivariable logistic regression analysis, statin use was associated with lower odds of the composite outcome (adjusted odds ratio (aOR) 0.63, 95% CI [0.53-0.76]) compared to patients not on statins. Findings were consistent with IPTW (aOR 0.92, 95% CI [0.89- 0.95]). The proportion of the effect of statin on the primary outcome mediated by suPAR was estimated at 31.5%. CONCLUSION Prior-statin use is associated with improved outcomes and lower inflammation as measured by suPAR levels in patients hospitalized for COVID-19.
Collapse
Affiliation(s)
- Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical Branch, Galveston
| | - Husam R Shadid
- Department of Cardiology, University of Texas Health Houston, Houston
| | - Yiyuan Huang
- Department of Biostatistics, University of Michigan, Ann Arbor
| | | | - Alexi Vasbinder
- Department of Biobehavioral Nursing and Health Informatics, School of Nursing, University of Washington, Seattle
| | - Ian Pizzo
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | | | | | - Pennelope Kunkle
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical Branch, Galveston
| | | | - Melvyn Rubenfire
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor
| | - Eric J Brandt
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor
| | - Geoffery Williams
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor
| | - Rodica Pop-Busui
- Division of Endocrinology, Department of Medicine, University of Michigan, Ann Arbor
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical Branch, Galveston; Department of Internal Medicine, University of Michigan, Ann Arbor; On behalf of the International Study of Inflammation In COVID-19 (ISIC) Investigators.
| |
Collapse
|
2
|
Dabbaghi MM, Soleimani Roudi H, Safaei R, Baradaran Rahimi V, Fadaei MR, Askari VR. Unveiling the Mechanism of Protective Effects of Tanshinone as a New Fighter Against Cardiovascular Diseases: A Systematic Review. Cardiovasc Toxicol 2024; 24:1467-1509. [PMID: 39306819 DOI: 10.1007/s12012-024-09921-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 11/15/2024]
Abstract
Tanshinone, a natural compound found in the roots of Salvia miltiorrhiza, has been shown to possess various pharmacological properties, including anti-inflammatory, antioxidant, and cardiovascular protective effects. This article aims to review the literature on the cardiovascular protective effects of tanshinone and its underlying mechanisms. Tanshinone has been demonstrated to improve cardiac function, reduce oxidative stress, and inhibit inflammation in various animal models of cardiovascular diseases. Additionally, it has been shown to regulate multiple signaling pathways involved in the pathogenesis of cardiovascular diseases, such as the PI3K/AKT, MAPK, and NF-κB pathways. Clinical studies have also suggested that tanshinone may have therapeutic potential for treating cardiovascular diseases. In conclusion, tanshinone has emerged as a promising natural compound with significant cardiovascular protective effects, and further research is warranted to explore its potential clinical applications.
Collapse
Affiliation(s)
- Mohammad Mahdi Dabbaghi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Hesan Soleimani Roudi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Rozhan Safaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran.
| |
Collapse
|
3
|
Lisboa CD, Maciel de Souza JL, Gaspar CJ, Turck P, Ortiz VD, Teixeira Proença IC, Fernandes TRG, Fernandes E, Tasca S, Carraro CC, Belló-Klein A, Sander da Rosa Araujo A, Luz de Castro A. Melatonin effects on oxidative stress and on TLR4/NF-kβ inflammatory pathway in the right ventricle of rats with pulmonary arterial hypertension. Mol Cell Endocrinol 2024; 592:112330. [PMID: 39002930 DOI: 10.1016/j.mce.2024.112330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/19/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Pulmonary arterial hypertension (PAH) is characterised by an increase in mean pulmonary arterial pressure and a compromised the right ventricle (RV), together with progression to heart failure and premature death. Studies have evaluated the role of melatonin as a promising therapeutic strategy for PAH. The objective of this study was to evaluate melatonin's effects on oxidative stress and on the TLR4/NF-kβ inflammatory pathway in the RV of rats with PAH. Male Wistar rats were divided into the following groups: control, monocrotaline (MCT), and monocrotaline plus melatonin groups. These two last groups received one intraperitoneal injection of MCT (60 mg/kg) on the first day of experimental protocol. The monocrotaline plus melatonin group received 10 mg/kg/day of melatonin by gavage for 21 days. Echocardiographic analysis was performed, and the RV was collected for morphometric analysis oxidative stress and molecular evaluations. The main findings of the present study were that melatonin administration attenuated the reduction in RV function that was induced by monocrotaline, as assessed by TAPSE. In addition, melatonin prevented RV diastolic area reduction caused by PAH. Furthermore, animals treated with melatonin did not show an increase in ROS levels or in NF-kβ expression. In addition, the monocrotaline plus melatonin group showed a reduction in TLR4 expression when compared with control and monocrotaline groups. To our knowledge, this is the first study demonstrating a positive effect of melatonin on the TLR4/NF-kβ pathway in the RV of rats with PAH. In this sense, this study makes it possible to think of melatonin as a possible ally in mitigating RV alterations caused by PAH.
Collapse
Affiliation(s)
- Cristiane Dias Lisboa
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - José Luciano Maciel de Souza
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Custódio José Gaspar
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Patrick Turck
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Vanessa Duarte Ortiz
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Isabel Cristina Teixeira Proença
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Tânia Regina G Fernandes
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Elissa Fernandes
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Silvio Tasca
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Cristina Campos Carraro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Alexandre Luz de Castro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil.
| |
Collapse
|
4
|
Song R, Baker TL, Watters JJ, Kumar S. Obstructive Sleep Apnea-Associated Intermittent Hypoxia-Induced Immune Responses in Males, Pregnancies, and Offspring. Int J Mol Sci 2024; 25:1852. [PMID: 38339130 PMCID: PMC10856042 DOI: 10.3390/ijms25031852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Obstructive sleep apnea (OSA), a respiratory sleep disorder associated with cardiovascular diseases, is more prevalent in men. However, OSA occurrence in pregnant women rises to a level comparable to men during late gestation, creating persistent effects on both maternal and offspring health. The exact mechanisms behind OSA-induced cardiovascular diseases remain unclear, but inflammation and oxidative stress play a key role. Animal models using intermittent hypoxia (IH), a hallmark of OSA, reveal several pro-inflammatory signaling pathways at play in males, such as TLR4/MyD88/NF-κB/MAPK, miRNA/NLRP3, and COX signaling, along with shifts in immune cell populations and function. Limited evidence suggests similarities in pregnancies and offspring. In addition, suppressing these inflammatory molecules ameliorates IH-induced inflammation and tissue injury, providing new potential targets to treat OSA-associated cardiovascular diseases. This review will focus on the inflammatory mechanisms linking IH to cardiovascular dysfunction in males, pregnancies, and their offspring. The goal is to inspire further investigations into the understudied populations of pregnant females and their offspring, which ultimately uncover underlying mechanisms and therapeutic interventions for OSA-associated diseases.
Collapse
Affiliation(s)
- Ruolin Song
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Tracy L. Baker
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Jyoti J. Watters
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
| |
Collapse
|
5
|
|
6
|
Arnaud C, Billoir E, de Melo Junior AF, Pereira SA, O'Halloran KD, Monteiro EC. Chronic intermittent hypoxia-induced cardiovascular and renal dysfunction: from adaptation to maladaptation. J Physiol 2023; 601:5553-5577. [PMID: 37882783 DOI: 10.1113/jp284166] [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: 07/07/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
Chronic intermittent hypoxia (CIH) is the dominant pathological feature of human obstructive sleep apnoea (OSA), which is highly prevalent and associated with cardiovascular and renal diseases. CIH causes hypertension, centred on sympathetic nervous overactivity, which persists following removal of the CIH stimulus. Molecular mechanisms contributing to CIH-induced hypertension have been carefully delineated. However, there is a dearth of knowledge on the efficacy of interventions to ameliorate high blood pressure in established disease. CIH causes endothelial dysfunction, aberrant structural remodelling of vessels and accelerates atherosclerotic processes. Pro-inflammatory and pro-oxidant pathways converge on disrupted nitric oxide signalling driving vascular dysfunction. In addition, CIH has adverse effects on the myocardium, manifesting atrial fibrillation, and cardiac remodelling progressing to contractile dysfunction. Sympatho-vagal imbalance, oxidative stress, inflammation, dysregulated HIF-1α transcriptional responses and resultant pro-apoptotic ER stress, calcium dysregulation, and mitochondrial dysfunction conspire to drive myocardial injury and failure. CIH elaborates direct and indirect effects in the kidney that initially contribute to the development of hypertension and later to chronic kidney disease. CIH-induced morphological damage of the kidney is dependent on TLR4/NF-κB/NLRP3/caspase-1 inflammasome activation and associated pyroptosis. Emerging potential therapies related to the gut-kidney axis and blockade of aryl hydrocarbon receptors (AhR) are promising. Cardiorenal outcomes in response to intermittent hypoxia present along a continuum from adaptation to maladaptation and are dependent on the intensity and duration of exposure to intermittent hypoxia. This heterogeneity of OSA is relevant to therapeutic treatment options and we argue the need for better stratification of OSA phenotypes.
Collapse
Affiliation(s)
- Claire Arnaud
- Université Grenoble-Alpes INSERM U1300, Laboratoire HP2, Grenoble, France
| | - Emma Billoir
- Université Grenoble-Alpes INSERM U1300, Laboratoire HP2, Grenoble, France
| | | | - Sofia A Pereira
- iNOVA4Health, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Emilia C Monteiro
- iNOVA4Health, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
7
|
Zhang Y, Wu J, Dong E, Wang Z, Xiao H. Toll-like receptors in cardiac hypertrophy. Front Cardiovasc Med 2023; 10:1143583. [PMID: 37113698 PMCID: PMC10126280 DOI: 10.3389/fcvm.2023.1143583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) that can identify pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). TLRs play an important role in the innate immune response, leading to acute and chronic inflammation. Cardiac hypertrophy, an important cardiac remodeling phenotype during cardiovascular disease, contributes to the development of heart failure. In previous decades, many studies have reported that TLR-mediated inflammation was involved in the induction of myocardium hypertrophic remodeling, suggesting that targeting TLR signaling might be an effective strategy against pathological cardiac hypertrophy. Thus, it is necessary to study the mechanisms underlying TLR functions in cardiac hypertrophy. In this review, we summarized key findings of TLR signaling in cardiac hypertrophy.
Collapse
Affiliation(s)
- Yanan Zhang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Department of Clinical Laboratory, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jimin Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- Department of Clinical Laboratory, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Correspondence: Zhanli Wang Han Xiao
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University Third Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Correspondence: Zhanli Wang Han Xiao
| |
Collapse
|
8
|
Zhou J, Lin H, Lv T, Hao J, Zhang H, Sun S, Yang J, Chi J, Guo H. Inappropriate Activation of TLR4/NF-κB is a Cause of Heart Failure. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2022. [DOI: 10.15212/cvia.2022.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Significance: Heart failure, a disease with extremely high incidence, is closely associated with inflammation and oxidative stress. The Toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) pathway plays an important role in the occurrence and development of heart failure.
Recent advances: Previous studies have shown that TLR4/NF-κB causes heart failure by inducing oxidative stress and inflammation; damaging the endothelia; promoting fibrosis; and inducing myocardial hypertrophy, apoptosis, pyroptosis, and autophagy.
Critical issues: Understanding the pathogenesis of heart failure is essential for the treatment of this disease. In this review, we outline the mechanisms underlying TLR4/NF-κB pathway-mediated heart failure and discuss drugs that alleviate heart failure by regulating the TLR4/NF-κB pathway.
Future directions: During TLR4/NF-κB overactivation, interventions targeting specific receptor antagonists may effectively alleviate heart failure, thus providing a basis for the development of new anti-heart failure drugs.
Collapse
Affiliation(s)
- Jiedong Zhou
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People’s Hospital Shaoxing Hospital, Shaoxing, China
| | - Tingting Lv
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Jinjin Hao
- Zhejiang University School of Medicine, Hangzhou, China
| | - Hanlin Zhang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Shimin Sun
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Juntao Yang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People’s Hospital Shaoxing Hospital, Shaoxing, China
| | - Hangyuan Guo
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
9
|
Protective effects of statins on COVID-19 risk, severity and fatal outcome: a nationwide Swedish cohort study. Sci Rep 2022; 12:12047. [PMID: 35835835 PMCID: PMC9282150 DOI: 10.1038/s41598-022-16357-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/08/2022] [Indexed: 12/11/2022] Open
Abstract
The impact of statins on COVID-19 remains unclear. This study aims to investigate whether statin exposure assessed both in the population and in well-defined cohorts of COVID-19 patients may affect the risk and severity of COVID-19 using nationwide Swedish population-based register data. A population ≥ 40 years was selected by age/sex-stratified random sampling from the Swedish population on 1 Jan 2020. COVID-19 outcomes were identified from the SmiNet database, the National Patient Register and/or Cause-of-Death Register and linked with the National Prescribed Drug Register and sociodemographic registers. Statin exposure was defined as any statin prescriptions in the year before index date. In Cox regressions, confounding was addressed using propensity score ATT (Average Treatment effect in the Treated) weighting. Of 572,695 individuals in the overall cohort, 22.3% had prior statin treatment. After ATT weighting, protective effects were observed among statin user for hospitalization and COVID-19 death in the overall cohort and onset cohort. In the hospitalized cohort, statin use was only associated with lower risk for death (HR = 0.86, 95% CI 0.79–0.95), but not ICU admission. Statin-treated individuals appear to have lower COVID-19 mortality than nonusers, whether assessed in the general population, from COVID-19 onset or from hospitalization.
Collapse
|
10
|
Ghati N, Bhatnagar S, Mahendran M, Thakur A, Prasad K, Kumar D, Dwivedi T, Mani K, Tiwari P, Gupta R, Mohan A, Saxena A, Guleria R, Deepti S. Statin and aspirin as adjuvant therapy in hospitalised patients with SARS-CoV-2 infection: a randomised clinical trial (RESIST trial). BMC Infect Dis 2022; 22:606. [PMID: 35810307 PMCID: PMC9270743 DOI: 10.1186/s12879-022-07570-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/27/2022] [Indexed: 01/21/2023] Open
Abstract
Background Statins and aspirin have been proposed for treatment of COVID-19 because of their anti-inflammatory and anti-thrombotic properties. Several observational studies have shown favourable results. There is a need for a randomised controlled trial. Methods In this single-center, open-label, randomised controlled trial, 900 RT-PCR positive COVID-19 patients requiring hospitalisation, were randomly assigned to receive either atorvastatin 40 mg (Group A, n = 224), aspirin 75 mg (Group B, n = 225), or both (Group C, n = 225) in addition to standard of care for 10 days or until discharge whichever was earlier or only standard of care (Group D, n = 226). The primary outcome variable was clinical deterioration to WHO Ordinal Scale for Clinical Improvement ≥ 6. The secondary outcome was change in serum C-reactive protein, interleukin-6, and troponin I. Results The primary outcome occurred in 25 (2.8%) patients: 7 (3.2%) in Group A, 3 (1.4%) in Group B, 8 (3.6%) in Group C, and 7 (3.2%) in Group D. There was no difference in primary outcome across the study groups (P = 0.463). Comparison of all patients who received atorvastatin or aspirin with the control group (Group D) also did not show any benefit [Atorvastatin: HR 1.0 (95% CI 0.41–2.46) P = 0.99; Aspirin: HR 0.7 (95% CI 0.27–1.81) P = 0.46]. The secondary outcomes revealed lower serum interleukin-6 levels among patients in Groups B and C. There was no excess of adverse events. Conclusions Among patients admitted with mild to moderate COVID-19 infection, additional treatment with aspirin, atorvastatin, or a combination of the two does not prevent clinical deterioration. Trial Registry Number CTRI/2020/07/026791 (http://ctri.nic.in; registered on 25/07/2020) Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07570-5.
Collapse
Affiliation(s)
- Nirmal Ghati
- Department of Cardiology, Jai Prakash Narayan Apex Trauma Center, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sushma Bhatnagar
- Department of Onco-Anaesthesia, Dr. B.R.A Institute-Rotary Cancer Hospital, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Manjit Mahendran
- Department of Cardiology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Abhishek Thakur
- Department of Cardiology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Kshitij Prasad
- Department of Cardiology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Devesh Kumar
- Department of Cardiology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Tanima Dwivedi
- Department of Laboratory Medicine, National Cancer Institute (Jhajjar, Haryana), All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Kalaivani Mani
- Department of Biostatistics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Pawan Tiwari
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Ritu Gupta
- Department of Laboratory Oncology, Dr. B.R.A Institute-Rotary Cancer Hospital, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anita Saxena
- Department of Cardiology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Randeep Guleria
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Siddharthan Deepti
- Department of Cardiology, All India Institute of Medical Sciences (AIIMS), New Delhi, India. .,Department of Cardiology, Cardiothoracic Sciences Centre, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
11
|
Mahmoudi A, Heydari S, Markina YV, Barreto GE, Sahebkar A. Role of statins in regulating molecular pathways following traumatic brain injury: A system pharmacology study. Biomed Pharmacother 2022; 153:113304. [PMID: 35724514 DOI: 10.1016/j.biopha.2022.113304] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/28/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious disorder with debilitating physical and psychological complications. Previous studies have indicated that genetic factors have a critical role in modulating the secondary phase of injury in TBI. Statins have interesting pleiotropic properties such as antiapoptotic, antioxidative, and anti-inflammatory effects, which make them a suitable class of drugs for repurposing in TBI. In this study, we aimed to explore how statins modulate proteins and pathways involved in TBI using system pharmacology. We first explored the target associations with statins in two databases to discover critical clustering groups, candidate hub and critical hub genes in the network of TBI, and the possible connections of statins with TBI-related genes. Our results showed 1763 genes associated with TBI. Subsequently, the analysis of centralities in the PPI network displayed 55 candidate hub genes and 15 hub genes. Besides, MCODE analysis based on threshold score:10 determined four modular clusters. Intersection analysis of genes related to TBI and statins demonstrated 204 shared proteins, which suggested that statins influence 31 candidate hub and 9 hub genes. Moreover, statins had the highest interaction with MCODE1. The biological processes of the 31 shared proteins are related to gene expression, inflammation, antioxidant activity, and cell proliferation. Biological enriched pathways showed Programmed Cell Death proteins, AGE-RAGE signaling pathway, C-type lectin receptor signalling pathway, and MAPK signaling pathway as top clusters. In conclusion, statins could target several critical post-TBI genes mainly involved in inflammation and apoptosis, supporting the previous research results as a potential therapeutic agent.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177899191, the Islamic Republic of Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran
| | - Sahar Heydari
- Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, the Islamic Republic of Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran
| | - Yuliya V Markina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBI "Petrovsky National Research Center of Surgery", 3 Tsyurupy Str., 117418, Moscow, the Russian Federation
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran.
| |
Collapse
|
12
|
González-Candia A, Candia AA, Paz A, Mobarec F, Urbina-Varela R, del Campo A, Herrera EA, Castillo RL. Cardioprotective Antioxidant and Anti-Inflammatory Mechanisms Induced by Intermittent Hypobaric Hypoxia. Antioxidants (Basel) 2022; 11:antiox11061043. [PMID: 35739940 PMCID: PMC9220055 DOI: 10.3390/antiox11061043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022] Open
Abstract
More than 80 million people live and work (in a chronic or intermittent form) above 2500 masl, and 35 million live in the Andean Mountains. Furthermore, in Chile, it is estimated that 100,000 people work in high-altitude shifts, where stays in the lowlands are interspersed with working visits in the highlands. Acute exposure to high altitude has been shown to induce oxidative stress in healthy human lowlanders due to increased free radical formation and decreased antioxidant capacity. However, intermittent hypoxia (IH) induces preconditioning in animal models, generating cardioprotection. Here, we aim to describe the responses of a cardiac function to four cycles of intermittent hypobaric hypoxia (IHH) in a rat model. The twelve adult Wistar rats were randomly divided into two equal groups, a four-cycle of IHH and a normobaric hypoxic control. Intermittent hypoxia was induced in a hypobaric chamber in four continuous cycles (1 cycle = 4 days of hypoxia + 4 days of normoxia), reaching a barometric pressure equivalent to 4600 m of altitude (428 Torr). At the end of the fourth cycle, cardiac structural and functional variables were also determined by echocardiography; furthermore, cardiac oxidative stress biomarkers (4-Hydroxynonenal, HNE; nitrotyrosine, NT), antioxidant enzymes, and NLRP3 inflammasome panel expression are also determined. Our results show a higher ejection and a shortening fraction of the left ventricle function by the end of the fourth cycle. Furthermore, cardiac tissue presented a decreased expression of antioxidant proteins. However, a decrease in IL-1β, TNF-αn, and oxidative stress markers is observed in IHH compared to normobaric hypoxic controls. Non-significant differences were found in protein levels of NLRP3 and caspase-1. IHH exposure determines structural and functional heart changes. These findings suggest that initial states of IHH are beneficial for cardiovascular function and protection.
Collapse
Affiliation(s)
| | - Alejandro A. Candia
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
- Department for the Woman and Newborn Health Promotion, Universidad de Chile, Santiago 7500922, Chile
| | - Adolfo Paz
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
| | - Fuad Mobarec
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
| | - Rodrigo Urbina-Varela
- Laboratorio de Fisiología y Bioenergética Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; (R.U.-V.); (A.d.C.)
| | - Andrea del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; (R.U.-V.); (A.d.C.)
| | - Emilio A. Herrera
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile; (A.A.C.); (A.P.); (F.M.)
- International Center for Andean Studies (INCAS), University of Chile, Putre 1070000, Chile
- Correspondence: (E.A.H.); or (R.L.C.); Tel.: +56-982-337-566 (R.L.C.)
| | - Rodrigo L. Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Unidad de Paciente Crítico, Hospital del Salvador, Santiago 7500922, Chile
- Correspondence: (E.A.H.); or (R.L.C.); Tel.: +56-982-337-566 (R.L.C.)
| |
Collapse
|
13
|
Rommasi F, Nasiri MJ, Mirsaeidi M. Immunomodulatory agents for COVID-19 treatment: possible mechanism of action and immunopathology features. Mol Cell Biochem 2022; 477:711-726. [PMID: 35013850 PMCID: PMC8747854 DOI: 10.1007/s11010-021-04325-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023]
Abstract
The novel coronavirus pandemic has emerged as one of the significant medical-health challenges of the current century. The World Health Organization has named this new virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since the first detection of SARS-CoV-2 in November 2019 in Wuhan, China, physicians, researchers, and others have made it their top priority to find drugs and cures that can effectively treat patients and reduce mortality rates. The symptoms of Coronavirus Disease 2019 (COVID-19) include fever, dry cough, body aches, and anosmia. Various therapeutic compounds have been investigated and applied to mitigate the symptoms in COVID-19 patients and cure the disease. Degenerative virus analyses of the infection incidence and COVID-19 have demonstrated that SARS-CoV-2 penetrates the pulmonary alveoli's endothelial cells through Angiotensin-Converting Enzyme 2 (ACE2) receptors on the membrane, stimulates various signaling pathways and causes excessive secretion of cytokines. The continuous triggering of the innate and acquired immune system, as well as the overproduction of pro-inflammatory factors, cause a severe condition in the COVID-19 patients, which is called "cytokine storm". It can lead to acute respiratory distress syndrome (ARDS) in critical patients. Severe and critical COVID-19 cases demand oxygen therapy and mechanical ventilator support. Various drugs, including immunomodulatory and immunosuppressive agents (e.g., monoclonal antibodies (mAbs) and interleukin antagonists) have been utilized in clinical trials. However, the studies and clinical trials have documented diverging findings, which seem to be due to the differences in these drugs' possible mechanisms of action. These drugs' mechanism of action generally includes suppressing or modulating the immune system, preventing the development of cytokine storm via various signaling pathways, and enhancing the blood vessels' diameter in the lungs. In this review article, multiple medications from different drug families are discussed, and their possible mechanisms of action are also described.
Collapse
Affiliation(s)
- Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mirsaeidi
- Department of Pulmonary and Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
14
|
Sagris M, Theofilis P, Antonopoulos AS, Oikonomou E, Tsioufis K, Tousoulis D. Genetic Predisposition and Inflammatory Inhibitors in COVID-19: Where Do We Stand? Biomedicines 2022; 10:biomedicines10020242. [PMID: 35203452 PMCID: PMC8868779 DOI: 10.3390/biomedicines10020242] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) and the resulting coronavirus disease-19 (COVID-19) have led to a global pandemic associated with high fatality rates. COVID-19 primarily manifests in the respiratory system as an acute respiratory distress syndrome following viral entry through the angiotensin-converting enzyme-2 (ACE2) that is present in pulmonary epithelial cells. Central in COVID-19 is the burst of cytokines, known as a “cytokine storm”, and the subsequent widespread endothelial activation, leading to cardiovascular complications such as myocarditis, arrhythmias, and adverse vascular events, among others. Genetic alterations may play an additive, detrimental role in the clinical course of patients with COVID-19, since gene alterations concerning ACE2, major histocompatibility complex class I, and toll-like receptors may predispose patients to a worse clinical outcome. Since the role of inflammation is quintessential in COVID-19, pharmacologic inhibition of various signaling pathways such as the interleukin-1 and -6, tumor necrosis factor-alpha, interferon gamma, Janus kinase-signal transducer and activator of transcription, and granulocyte–macrophage colony-stimulating factor may ameliorate the prognosis following timely administration. Finally, frequently used, non-specific anti-inflammatory agents such as corticosteroids, statins, colchicine, and macrolides represent additional therapeutic considerations.
Collapse
Affiliation(s)
- Marios Sagris
- 1st Cardiology Clinic, School of Medicine, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (K.T.); (D.T.)
- Correspondence: ; Tel.: +30-21-3208-8099; Fax: +30-21-3208-8676
| | - Panagiotis Theofilis
- 1st Cardiology Clinic, School of Medicine, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Alexios S. Antonopoulos
- 1st Cardiology Clinic, School of Medicine, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- 1st Cardiology Clinic, School of Medicine, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (K.T.); (D.T.)
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, University of Athens Medical School, 157 72 Athens, Greece
| | - Kostas Tsioufis
- 1st Cardiology Clinic, School of Medicine, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- 1st Cardiology Clinic, School of Medicine, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece; (P.T.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| |
Collapse
|
15
|
COVID-19, the Pandemic of the Century and Its Impact on Cardiovascular Diseases. CARDIOLOGY DISCOVERY 2021; 1:233-258. [PMID: 34888547 PMCID: PMC8638821 DOI: 10.1097/cd9.0000000000000038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/19/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection likely ranks among the deadliest diseases in human history. As with other coronaviruses, SARS-CoV-2 infection damages not only the lungs but also the heart and many other organs that express angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV-2. COVID-19 has upended lives worldwide. Dietary behaviors have been altered such that they favor metabolic and cardiovascular complications, while patients have avoided hospital visits because of limited resources and the fear of infection, thereby increasing out-hospital mortality due to delayed diagnosis and treatment. Clinical observations show that sex, age, and race all influence the risk for SARS-CoV-2 infection, as do hypertension, obesity, and pre-existing cardiovascular conditions. Many hospitalized COVID-19 patients suffer cardiac injury, acute coronary syndromes, or cardiac arrhythmia. SARS-CoV-2 infection may lead to cardiomyocyte apoptosis and necrosis, endothelial cell damage and dysfunction, oxidative stress and reactive oxygen species production, vasoconstriction, fibrotic and thrombotic protein expression, vascular permeability and microvascular dysfunction, heart inflammatory cell accumulation and activation, and a cytokine storm. Current data indicate that COVID-19 patients with cardiovascular diseases should not discontinue many existing cardiovascular therapies such as ACE inhibitors, angiotensin receptor blockers, steroids, aspirin, statins, and PCSK9 inhibitors. This review aims to furnish a framework relating to COVID-19 and cardiovascular pathophysiology.
Collapse
|
16
|
Bayer AL, Alcaide P. MyD88: At the heart of inflammatory signaling and cardiovascular disease. J Mol Cell Cardiol 2021; 161:75-85. [PMID: 34371036 PMCID: PMC8629847 DOI: 10.1016/j.yjmcc.2021.08.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide and is associated with systemic inflammation. In depth study of the cell-specific signaling mechanisms mediating the inflammatory response is vital to improving anti-inflammatory therapies that reduce mortality and morbidity. Cellular damage in the cardiovascular system results in the release of damage associated molecular patterns (DAMPs), also known as "alarmins," which activate myeloid cells through the adaptor protein myeloid differentiation primary response 88 (MyD88). MyD88 is broadly expressed in most cell types of the immune and cardiovascular systems, and its role often differs in a cardiovascular disease context and cell specific manner. Herein we review what is known about MyD88 in the setting of a variety of cardiovascular diseases, discussing cell specific functions and the relative contributions of MyD88-dependent vs. independent alarmin triggered inflammatory signaling. The widespread involvement of these pathways in cardiovascular disease, and their largely unexplored complexity, sets the stage for future in depth mechanistic studies that may place MyD88 in both immune and non-immune cell types as an attractive target for therapeutic intervention in cardiovascular disease.
Collapse
Affiliation(s)
- Abraham L Bayer
- Department of Immunology, Tufts University School of Medicine. 136 Harrison Ave, Boston, MA 02111, United States of America.
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine. 136 Harrison Ave, Boston, MA 02111, United States of America.
| |
Collapse
|
17
|
Alves PHR, Ferron AJT, Costa MR, Hasimoto FK, Gregolin CS, Garcia JL, Campos DHSD, Cicogna AC, Mattei LD, Moreto F, Bazan SGZ, Francisqueti-Ferron FV, Corrêa CR. Relationship between Innate Immune Response Toll-Like Receptor 4 (TLR-4) and the Pathophysiological Process of Obesity Cardiomyopathy. Arq Bras Cardiol 2021; 117:91-99. [PMID: 34320076 PMCID: PMC8294729 DOI: 10.36660/abc.20190788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 06/16/2020] [Indexed: 12/23/2022] Open
Abstract
Fundamento A obesidade é uma condição inflamatória crônica de baixo grau relacionada a distúrbios cardíacos. No entanto, o mecanismo responsável pela inflamação cardíaca relacionada à obesidade não é claro. O receptor do tipo toll 4 (TLR-4) pertence a um receptor da família das transmembranas, responsável pela resposta imune, cuja ativação estimula a produção de citocinas pró-inflamatórias. Objetivo Testar se a ativação do receptor TLR-4 participa do processo de cardiomiopatia da obesidade, devido à produção de citocinas por meio da ativação do NF-ĸB. Métodos Ratos Wistar machos foram randomizados em dois grupos: o grupo controle (C, n = 8 animais) que recebeu dieta padrão/água e o grupo obeso (OB, n = 8 animais) que foi alimentado com dieta rica em açúcar e gordura e água mais 25% de sacarose por 30 semanas. Análise nutricional: peso corporal, índice de adiposidade, alimentos, água e ingestão calórica. Análise de distúrbios relacionados à obesidade: glicose plasmática, ácido úrico e triglicerídeos, HOMA-IR, pressão arterial sistólica, TNF-α no tecido adiposo. A análise cardíaca incluiu: expressão das proteínas TLR-4 e NF-ĸB, níveis de TNF-α e IL-6. Comparação pelo teste t de
Student
não pareado ou teste de Mann-Whitney com um valor de p <0,05 como estatisticamente significativo. Resultados O grupo OB apresentou obesidade, glicose elevada, triglicerídeos, ácido úrico, HOMA, pressão arterial sistólica e TNF-α no tecido adiposo. O grupo OB apresentou remodelação cardíaca e disfunção diastólica. A expressão de TLR-4 e NF-ĸB e os níveis de citocinas foram maiores em OB. Conclusão Nossos achados concluem que, em uma condição obesogênica, a inflamação derivada da ativação do TLR-4 cardíaco pode ser um mecanismo capaz de levar à remodelação e disfunção cardíaca.
Collapse
Affiliation(s)
- Pedro Henrique Rizzi Alves
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | - Artur Junio Togneri Ferron
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | - Mariane Róvero Costa
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | - Fabiana Kurokawa Hasimoto
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | - Cristina Schmitt Gregolin
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil.,Universidade Federal de Mato Grosso , Sinop , MT - Brasil
| | - Jéssica Leite Garcia
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | | | - Antônio Carlos Cicogna
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | - Letícia de Mattei
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | - Fernando Moreto
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | - Silméia Garcia Zanati Bazan
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| | | | - Camila Renata Corrêa
- Universidade Estadual Paulista Júlio de Mesquita Filho Câmpus de Botucatu Faculdade de Medicina , Botucatu , SP - Brasil
| |
Collapse
|
18
|
Zhang XB, Cheng HJ, Yuan YT, Chen Y, Chen YY, Chiu KY, Zeng HQ. Atorvastatin attenuates intermittent hypoxia-induced myocardial oxidative stress in a mouse obstructive sleep apnea model. Aging (Albany NY) 2021; 13:18870-18878. [PMID: 34289453 PMCID: PMC8351704 DOI: 10.18632/aging.203339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Chronic intermittent hypoxia (CIH), a hallmark of obstructive sleep apnea (OSA), is associated with various cardiovascular diseases. In the present study, we assessed the effect of the lipid reducing agent atorvastatin on CIH-induced myocardial oxidative stress and apoptosis in a mouse OSA model. Forty-eight C57BL/6J mice were evenly divided among normoxia + vehicle, normoxia + atorvastatin, CIH + vehicle, and CIH + atorvastatin groups. CIH consisted of a hypoxia-reoxygenation cycle in which oxygen concentrations fluctuated from 21% to 6% and back over two minutes for 8 hours each day (30 events/hour). CIH exposure continued for 12 weeks. Atorvastatin (5 mg/kg) was administered from week 6 through the end of the experiment. CIH increased malondialdehyde levels and decreased superoxide dismutase activity, total antioxidant capacity, and nuclear factor erythroid 2-related factor 2 levels in cardiac tissue, indicating a reduction in antioxidant activity. Atorvastatin significantly reversed those effects (p < 0.05). CIH also increased B-cell lymphoma 2-associated protein X and cleaved caspased-3 levels as well as the myocardial apoptotic rate, as indicated by terminal deoxynucleotidyl transferase dUTP nick-end labeling. Atorvastatin had no effect on those changes (p > 0.05). Thus, atorvastatin administration exerts antioxidant but not anti-apoptotic effects after CIH and may therefore have therapeutic potential in OSA patients with cardiovascular comorbidities.
Collapse
Affiliation(s)
- Xiao-Bin Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province, People's Republic of China
| | - Hui-Juan Cheng
- Department of Medical Affairs, Zhongshan Hospital, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province, People's Republic of China
| | - Ya-Ting Yuan
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province, People's Republic of China
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province, People's Republic of China
| | - Yi-Yuan Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province, People's Republic of China
| | - Kam Yu Chiu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province, People's Republic of China
| | - Hui-Qing Zeng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province, People's Republic of China
| |
Collapse
|
19
|
Lu J, Ma X, Gao WC, Zhang X, Fu Y, Liu Q, Tian L, Qin XD, Yang W, Zheng HY, Zheng CB. Gastrodin Exerts Cardioprotective Action via Inhibition of Insulin-Like Growth Factor Type 2/Insulin-Like Growth Factor Type 2 Receptor Expression in Cardiac Hypertrophy. ACS OMEGA 2021; 6:16763-16774. [PMID: 34250336 PMCID: PMC8264851 DOI: 10.1021/acsomega.1c00797] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/10/2021] [Indexed: 05/05/2023]
Abstract
Pathological cardiac hypertrophy is commonly associated with an upregulation of fetal genes, fibrosis, cardiac dysfunction, and heart failure. Previous studies have demonstrated that gastrodin (GAS) exerts cardioprotective action in the treatment of cardiac hypertrophy. However, the mechanism by which GAS protects against cardiac hypertrophy is yet to be elucidated. A mouse model of myocardial hypertrophy was established using an angiotensin II (Ang II) induction. GAS (5 or 50 mg/kg/d) was orally administered every day starting 7 days prior to the Ang II infusion combined with sham-operated controls. Heart samples from each group were collected for RNA sequencing. Using bioinformatics analysis, the key differentially expressed genes (DEGs) that are involved in reversing cardiac function were identified. Through bioinformatics analysis, the key DEGs that are involved in GAS's inhibition of Ang II-induced abnormal gene expression within the heart were identified. This was further validated using quantitative real-time PCR and Western blotting in neonatal rat cardiomyocytes (NRCMs). Oral administration of GAS significantly suppressed the Ang II-induced increase in heart size and heart weight to body weight. Furthermore, pretreatment of the NRCMs with GAS led to a dose-dependent inhibition of Ang II-induced increases in Nppb mRNA expression. We identified 620 upregulated and 87 downregulated Ang II-induced DEGs II, among which the expression patterns of 58 and 146 genes were inverted by low-dose and high-dose GAS, respectively. These inverted DEGs were found to be mainly enriched in the biological processes of regulation of Ras protein signal transduction, heart contraction, covalent chromatin modification, glucose metabolism, and positive regulation of cell cycle. Among them, the insulin-like growth factor type 2 (Igf2) gene, which was found to be highly reversed and downregulated by GAS, served as a core gene linking energy metabolism, immune regulation, and systemic development. Subsequent functional verification demonstrated that IGF2, and its receptor IGF2R, is one of the targets of GAS that helps protect against cardiac hypertrophy. Taken together, we have identified, for the first time, IGF2/IGF2R as a potential target influenced by GAS in the prevention of cardiac hypertrophy.
Collapse
Affiliation(s)
- Jun Lu
- Department
of Pharmacology, Guilin Medical University, Guilin 541199, China
| | - Xin Ma
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Wen-Cong Gao
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Xin Zhang
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Yuanling Fu
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Qian Liu
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Lixiang Tian
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Xiao-Dan Qin
- Department
of Pharmacology, Guilin Medical University, Guilin 541199, China
| | - Weimin Yang
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Hong-Yi Zheng
- Key
Laboratory of Animal Models and Human Diseases Mechanisms of Chinese
Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Chang-Bo Zheng
- School
of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology
for Natural Products, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
20
|
Kollias A, Kyriakoulis KG, Kyriakoulis IG, Nitsotolis T, Poulakou G, Stergiou GS, Syrigos K. Statin use and mortality in COVID-19 patients: Updated systematic review and meta-analysis. Atherosclerosis 2021; 330:114-121. [PMID: 34243953 PMCID: PMC8233054 DOI: 10.1016/j.atherosclerosis.2021.06.911] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/02/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Statin therapy is administered to patients with high cardiovascular risk. These patients are also at risk for severe course of coronavirus disease 2019 (COVID-19). Statins exhibit not only cardioprotective but also immunomodulatory and anti-inflammatory effects. This study performed a systematic review of published evidence regarding statin treatment and COVID-19 related mortality. METHODS A systematic PubMed/Embase search was performed from February 10, 2020 until March 05, 2021 for studies in COVID-19 patients that reported adjusted hazard or odds ratio for death in statin users versus non-users. RESULTS 22 studies fulfilled the inclusion criteria and were included in the systematic review. Meta-analysis of 10 studies (n = 41,807, weighted age 56 ± 8 years, men 51%, hypertension 34%, diabetes 21%, statin users 14%) that reported adjusted hazard ratios for mortality in statin users versus non-users showed pooled estimate at 0.65 (95% confidence intervals [CI] 0.53, 0.81). Meta-analysis of 6 studies that reported continuation of statin therapy during hospitalization (58-100% of patients) revealed a pooled hazard ratio of 0.54 (95% CI 0.47, 0.62). Meta-analysis of 12 studies (n = 72,881, weighted age 65 ± 2 years, men 54%, hypertension 66%, diabetes 43%, statin users 30%) that reported adjusted odds ratios for mortality showed pooled estimate at 0.65 (95% CI 0.55, 0.78). Multivariable meta-regression analysis did not reveal any significant association of hazard or odds ratios with anthropometric characteristics or comorbidities. CONCLUSIONS This meta-analysis of retrospective observational studies showed that statin therapy was associated with an about 35% decrease in the adjusted risk of mortality in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Anastasios Kollias
- Third Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Sotiria Hospital, Athens, Greece.
| | - Konstantinos G Kyriakoulis
- Third Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Sotiria Hospital, Athens, Greece
| | - Ioannis G Kyriakoulis
- Third Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Sotiria Hospital, Athens, Greece
| | - Thomas Nitsotolis
- Third Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Sotiria Hospital, Athens, Greece
| | - Garyphallia Poulakou
- Third Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Sotiria Hospital, Athens, Greece
| | - George S Stergiou
- Third Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Sotiria Hospital, Athens, Greece
| | - Konstantinos Syrigos
- Third Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Sotiria Hospital, Athens, Greece
| |
Collapse
|
21
|
Studying the Effect of Taking Statins before Infection in the Severity Reduction of COVID-19 with Machine Learning. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9995073. [PMID: 34250095 PMCID: PMC8218917 DOI: 10.1155/2021/9995073] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/25/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
Statins can help COVID-19 patients' treatment because of their involvement in angiotensin-converting enzyme-2. The main objective of this study is to evaluate the impact of statins on COVID-19 severity for people who have been taking statins before COVID-19 infection. The examined research patients include people that had taken three types of statins consisting of Atorvastatin, Simvastatin, and Rosuvastatin. The case study includes 561 patients admitted to the Razi Hospital in Ghaemshahr, Iran, during February and March 2020. The illness severity was encoded based on the respiratory rate, oxygen saturation, systolic pressure, and diastolic pressure in five categories: mild, medium, severe, critical, and death. Since 69.23% of participants were in mild severity condition, the results showed the positive effect of Simvastatin on COVID-19 severity for people that take Simvastatin before being infected by the COVID-19 virus. Also, systolic pressure for this case study is 137.31, which is higher than that of the total patients. Another result of this study is that Simvastatin takers have an average of 95.77 mmHg O2Sat; however, the O2Sat is 92.42, which is medium severity for evaluating the entire case study. In the rest of this paper, we used machine learning approaches to diagnose COVID-19 patients' severity based on clinical features. Results indicated that the decision tree method could predict patients' illness severity with 87.9% accuracy. Other methods, including the K-nearest neighbors (KNN) algorithm, support vector machine (SVM), Naïve Bayes classifier, and discriminant analysis, showed accuracy levels of 80%, 68.8%, 61.1%, and 85.1%, respectively.
Collapse
|
22
|
Chen F, Chen ZQ, Zhong GL, Zhu JJ. Nicorandil inhibits TLR4/MyD88/NF-κB/NLRP3 signaling pathway to reduce pyroptosis in rats with myocardial infarction. Exp Biol Med (Maywood) 2021; 246:1938-1947. [PMID: 34034564 DOI: 10.1177/15353702211013444] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pyroptosis is an inflammatory cell death that regulates cardiomyocyte loss after myocardial infarction. Reports indicate that nicorandil has a strong anti-inflammatory effect and protects the myocardium from myocardial infarction. However, its relationship with pyroptosis is largely unreported. Here, we investigated to influence and mechanism of action of nicorandil on cardiomyocyte pyroptosis. Forty Sprague Dawley rats were randomly assigned to sham, MI, MI + nicorandil, and MI + nicorandil + TAK242 groups (10 per group). Myocardial infarction modeling was performed through ligation of the anterior descending branch of the left coronary artery. The function of cardiac was evaluated through echocardiography, detection of myocardial adenine nucleotides, cTnI, LDH, TTC, and HE staining. Moreover, we used qRT-PCR, immunohistochemistry, and Western blotting to examine the expression of pyroptosis-related molecules and the inflammasome pathway of TLR4/MyD88/NF-κB/NLRP3. Myocardial infarction caused the activation of GSDMD, aggravated myocardial injury, and triggered cardiac dysfunction. Myocardial infarction induced pyroptotic cell death, manifested as upregulation in mRNA and protein levels associated with pyroptosis, including caspase-1 cleavage and increased expression of IL-1β and IL-18. These changes were mitigated by nicorandil. The achieved data implicate that myocardial infarction induces pyroptosis via the TLR4/MyD88/NF-κB/NLRP3 pathway, which can be inhibited by nicorandil pretreatment. Therefore, nicorandil exerts cardioprotective effects by activating KATP channels, and at least in part through inhibition of the TLR4/MyD88/NF-κB/NLRP3 pathway to reduce myocardial infarction-induced pyroptosis. As such, it is a potential therapy for ischemic heart disease.
Collapse
Affiliation(s)
- Feng Chen
- Department of Emergency, Guangxi Medical University First Affiliated Hospital, Nanning 530021, China
| | - Zhi-Qing Chen
- Department of Cardiology, Guangxi Medical University First Affiliated Hospital, Nanning 530021, China
| | - Gui-Ling Zhong
- Department of Cardiology, Guangxi Medical University First Affiliated Hospital, Nanning 530021, China
| | - Ji-Jin Zhu
- Department of Emergency, Guangxi Medical University First Affiliated Hospital, Nanning 530021, China.,Department of Cardiology, Guangxi Medical University First Affiliated Hospital, Nanning 530021, China
| |
Collapse
|
23
|
Greco S, D'Amuri A, Giorgini E, Luciani F, Lopreiato M, Fortunato V, Scopa A, Vestita G, Capatti E, Passaro A. Role of Statins in Coronavirus-Related Disease (COVID-19): A Retrospective Cohort Study in Northern Italy. High Blood Press Cardiovasc Prev 2021; 28:355-364. [PMID: 33905094 PMCID: PMC8077187 DOI: 10.1007/s40292-021-00452-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023] Open
Abstract
Introduction The outbreak by SARS-CoV-2 has rapidly spread worldwide. The need for specific treatments to adequately stop the inflammatory response and its sequelae is day by day more urgent and many therapeutic strategies were performed since COVID-19 burst in the last months. Statins were thought to be effective against this novel coronavirus for their anti-inflammatory properties, even if the real effects on COVID patients are still partially unexplored. Methods We retrospectively evaluated 501 adult patients, consecutively admitted to the two COVID-hospitals of Ferrara’s territory, and divided them into two groups: ST = patients on statin therapy on admission and NST=patients not on statin therapy on admission. We searched for differences between groups in terms of anamnestic, clinical and laboratory data and then in terms of COVID-19 outcomes. Results We found significant differences between groups in terms of age, comorbidities, procalcitonin and CPK serum levels: ST patients were older, more comorbid, with lower procalcitonin and higher CPK serum levels. Male sex was, together with the Charlson Comorbidity Index, an independent predictor of needing intensification of care, while age only was a good predictor of in-hospital and 100-day mortality. Differences were also found in the survival functions between the two groups. Conclusions After a period of observation of 100 days, ST patients, despite their older age and their greater load of comorbidities, have similar survival functions to NST patients. If adjusted for age and CCI the survival functions of ST group are considerably more favourable than those of the second group. Supplementary Information The online version contains supplementary material available at 10.1007/s40292-021-00452-y.
Collapse
Affiliation(s)
- Salvatore Greco
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Andrea D'Amuri
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Enrico Giorgini
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Francesco Luciani
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Mariarosaria Lopreiato
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Valeria Fortunato
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Alessandro Scopa
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Grazia Vestita
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy
| | - Eleonora Capatti
- Medical Department, University Hospital of Ferrara Arcispedale Sant'Anna, Via Aldo Moro, 8 Cona, 44124, Ferrara, Italy
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari, 46, 44121, Ferrara, Italy.
- Medical Department, University Hospital of Ferrara Arcispedale Sant'Anna, Via Aldo Moro, 8 Cona, 44124, Ferrara, Italy.
| |
Collapse
|
24
|
Jia X, Virani SS. Statins in COVID-19 infection: A rehash of old themes or truly a new hope? J Clin Lipidol 2021; 15:399-401. [PMID: 33994121 PMCID: PMC8059260 DOI: 10.1016/j.jacl.2021.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/04/2021] [Accepted: 04/11/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Xiaoming Jia
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Salim S Virani
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, United States; Health Policy, Quality & Informatics Program, Michael E. DeBakey Veterans Affairs Medical Center Health Services Research and Development Center for Innovations, Houston, TX 77030, United States; Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, United States; Section of Health Services Research, Department of Medicine, Baylor College of Medicine, Houston TX, United States.
| |
Collapse
|
25
|
Marić I, Oskotsky T, Kosti I, Le B, Wong RJ, Shaw GM, Sirota M, Stevenson DK. Decreased Mortality Rate Among COVID-19 Patients Prescribed Statins: Data From Electronic Health Records in the US. Front Med (Lausanne) 2021; 8:639804. [PMID: 33614688 PMCID: PMC7887302 DOI: 10.3389/fmed.2021.639804] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The severe respiratory illness due to SARS-CoV-2, the virus responsible for coronavirus disease 2019 (COVID-19), is triggered by an intense pro-inflammatory host response. Statins, prescribed primarily for lipid reduction, are known to have anti-inflammatory and immunomodulatory properties and have been associated with a reduced mortality rate among COVID-19 patients taking statins as reported in two recent retrospective studies. However, a meta-analysis that included nine studies showed that statin use did not improve in-hospital outcomes of those with COVID-19. In addition, concerns regarding the use of statins and an increase in COVID-19 infections have been raised, as statins may increase the expression of angiotensin-converting enzyme 2 (ACE2), the primary receptor for the SARS-CoV-2 virus. Our goal was to investigate the effect of statins in COVID-19 patients in a large, diverse patient population across the United States containing nearly 120,000 patients diagnosed with COVID-19. We used propensity score matching of demographics, comorbidities, and medication indication to compare statin-treated patients (N = 2,297) with matched controls (N = 4,594). We observed a small, but statistically significant, decrease in mortality among patients prescribed statins (16.1%) when compared with matched COVID-19-positive controls (18.0 to 20.6%). These results support previous evidence that statins do not increase COVID-19-related mortality and may, in fact, have a mitigating effect on severity of the disease reflected in a slight reduction in mortality. Mixed findings on effects of statins in COVID-19 patients reported in the literature should prompt prospective randomized controlled trials in order to define better who might be advantaged with respect to clinical outcomes.
Collapse
Affiliation(s)
- Ivana Marić
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Tomiko Oskotsky
- Department of Pediatrics, UCSF, San Francisco, CA, United States.,Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, United States
| | - Idit Kosti
- Department of Pediatrics, UCSF, San Francisco, CA, United States.,Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, United States
| | - Brian Le
- Department of Pediatrics, UCSF, San Francisco, CA, United States.,Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, United States
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Marina Sirota
- Department of Pediatrics, UCSF, San Francisco, CA, United States.,Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, United States
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
26
|
Esfehani RJ, Vojdanparast M, Soleimanpour S, Ferns GA, Avan A. The Potential Impact of Statins in the Treatment of Patients with COVID-19 Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1352:149-158. [DOI: 10.1007/978-3-030-85109-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
27
|
Levett JY, Raparelli V, Mardigyan V, Eisenberg MJ. Cardiovascular Pathophysiology, Epidemiology, and Treatment Considerations of Coronavirus Disease 2019 (COVID-19): A Review. CJC Open 2021; 3:28-40. [PMID: 33458630 PMCID: PMC7801216 DOI: 10.1016/j.cjco.2020.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/02/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is rapidly evolving, with important cardiovascular considerations. The presence of underlying cardiovascular risk factors and established cardiovascular disease (CVD) may affect the severity and clinical management of patients with COVID-19. We conducted a review of the literature to summarize the cardiovascular pathophysiology, risk factors, clinical presentations, and treatment considerations of COVID-19 patients with underlying CVD. Angiotensin-converting enzyme 2 (ACE2) has been identified as a functional receptor for the SARS-CoV-2 virus, and it is associated with the cardiovascular system. Hypertension, diabetes, and CVD are the most common comorbidities in COVID-19 patients, and these factors have been associated with the progression and severity of COVID-19. However, elderly populations, who develop more-severe COVID-19 complications, are naturally exposed to these comorbidities, underscoring the possible confounding of age. Observational data support international cardiovascular societies' recommendations to not discontinue ACE inhibitor/angiotensin-receptor blocker therapy in patients with guideline indications for fear of the increased risk of SARS-CoV-2 infection, severe disease, or death. In addition to the cardiotoxicity of experimental antivirals and potential interactions of experimental therapies with cardiovascular drugs, several strategies for cardiovascular protection have been recommended in COVID-19 patients with underlying CVD. Troponin elevation is associated with increased risk of in-hospital mortality and adverse outcomes in patients with COVID-19. Cardiovascular care teams should have a high index of suspicion for fulminant myocarditis-like presentations being SARS-CoV-2 positive, and remain vigilant for cardiovascular complications in COVID-19 patients.
Collapse
Affiliation(s)
- Jeremy Y. Levett
- Center of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital/McGill University, Montreal, Quebec, Canada
- Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Valeria Raparelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Vartan Mardigyan
- Division of Cardiology, Jewish General Hospital/McGill University, Montreal, Quebec, Canada
| | - Mark J. Eisenberg
- Center of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital/McGill University, Montreal, Quebec, Canada
- Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Division of Cardiology, Jewish General Hospital/McGill University, Montreal, Quebec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Mitacchione G, Schiavone M, Curnis A, Arca M, Antinori S, Gasperetti A, Mascioli G, Severino P, Sabato F, Caracciolo MM, Arabia G, D'Erasmo L, Viecca M, Mancone M, Galli M, Forleo GB. Impact of prior statin use on clinical outcomes in COVID-19 patients: data from tertiary referral hospitals during COVID-19 pandemic in Italy. J Clin Lipidol 2021; 15:68-78. [PMID: 33390341 PMCID: PMC7833194 DOI: 10.1016/j.jacl.2020.12.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/04/2020] [Accepted: 12/15/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Epidemiological evidence suggests that anti-inflammatory and immunomodulatory properties of statins may reduce the risk of infections and infection-related complications. OBJECTIVE We aimed to assess the impact of prior statin use on coronavirus disease (COVID-19) severity and mortality. METHODS In this observational multicenter study, consecutive patients hospitalized for COVID-19 were enrolled. In-hospital mortality and severity of COVID-19 assessed with National Early Warning Score (NEWS) were deemed primary and secondary outcomes, respectively. Propensity score (PS) matching was used to obtain balanced cohorts. RESULTS Among 842 patients enrolled, 179 (21%) were treated with statins before admission. Statin patients showed more comorbidities and more severe COVID-19 (NEWS 4 [IQR 2-6] vs 3 [IQR 2-5], p < 0.001). Despite having similar rates of intensive care unit admission, noninvasive ventilation, and mechanical ventilation, statin users appeared to show higher mortality rates. After balancing pre-existing relevant clinical conditions that could affect COVID-19 prognosis with PS matching, statin therapy confirmed its association with a more severe disease (NEWS ≥5 61% vs. 48%, p = 0.025) but not with in-hospital mortality (26% vs. 28%, p = 0.185). At univariate logistic regression analysis, statin use was confirmed not to be associated with mortality (OR 0.901; 95% CI: 0.537 to 1.51; p = 0.692) and to be associated with a more severe disease (NEWS≥5 OR 1.7; 95% CI 1.067-2.71; p = 0.026). CONCLUSIONS Our results did not confirm the supposed favorable effects of statin therapy on COVID-19 outcomes. Conversely, they suggest that statin use should be considered as a proxy of underlying comorbidities, which indeed expose to increased risks of more severe COVID-19.
Collapse
Affiliation(s)
| | - Marco Schiavone
- Department of Cardiology, Luigi Sacco - University Hospital, Milan, Italy
| | - Antonio Curnis
- Department of Cardiology, Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - Marcello Arca
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Spinello Antinori
- Department of Infectious Diseases, ASST-Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Alessio Gasperetti
- Department of Cardiology, Luigi Sacco - University Hospital, Milan, Italy
| | - Giosuè Mascioli
- Department of Arrhythmology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Paolo Severino
- Department of Clinical Internal, Anesthesiological and Cardiovascular Science, Sapienza University of Rome, Rome, Italy
| | - Federica Sabato
- Department of Cardiology, Luigi Sacco - University Hospital, Milan, Italy
| | - Maria M Caracciolo
- Department of Cardiology, Luigi Sacco - University Hospital, Milan, Italy
| | - Gianmarco Arabia
- Department of Cardiology, Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - Laura D'Erasmo
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Maurizio Viecca
- Department of Cardiology, Luigi Sacco - University Hospital, Milan, Italy
| | - Massimo Mancone
- Department of Clinical Internal, Anesthesiological and Cardiovascular Science, Sapienza University of Rome, Rome, Italy
| | - Massimo Galli
- Department of Infectious Diseases, ASST-Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Giovanni B Forleo
- Department of Cardiology, Luigi Sacco - University Hospital, Milan, Italy
| |
Collapse
|
29
|
Barkas F, Milionis H, Anastasiou G, Liberopoulos E. Statins and PCSK9 inhibitors: What is their role in coronavirus disease 2019? Med Hypotheses 2021; 146:110452. [PMID: 33333472 PMCID: PMC7724447 DOI: 10.1016/j.mehy.2020.110452] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/22/2020] [Accepted: 12/05/2020] [Indexed: 02/06/2023]
Abstract
Statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors interfere with several pathophysiological pathways of coronavirus disease 2019 (COVID-19). Statins may have a direct antiviral effect on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by inhibiting its main protease. Statin-induced up-regulation of angiotensin-converting enzyme 2 (ACE2) may also be beneficial, whereas cholesterol reduction might significantly suppress SARS-CoV-2 by either blocking its host-cell entry through the disruption of lipid rafts or by inhibiting its replication. Available human studies have shown beneficial effects of statins and PCSK9 inhibitors on pneumonia and sepsis. These drugs may act as immunomodulators in COVID-19 and protect against major complications, such as acute respiratory distress syndrome and cytokine release syndrome. Considering their antioxidative, anti-arrhythmic, antithrombotic properties and their beneficial effect on endothelial dysfunction, along with the increased risk of mortality of patients at high cardiovascular risk infected by SARS-CoV-2, statins and PCSK9 inhibitors might prove effective against the cardiovascular and thromboembolic complications of COVID-19. On the whole, randomized clinical trials are needed to establish routine use of statins and PCSK9 inhibitors in the treatment of SARS-CoV-2 infection. In the meantime, it is recommended that lipid-lowering therapy should not be discontinued in COVID-19 patients unless otherwise indicated.
Collapse
Affiliation(s)
- Fotios Barkas
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Greece
| | - Haralampos Milionis
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Greece
| | - Georgia Anastasiou
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Greece
| | - Evangelos Liberopoulos
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Greece.
| |
Collapse
|
30
|
Daniels LB, Sitapati AM, Zhang J, Zou J, Bui QM, Ren J, Longhurst CA, Criqui MH, Messer K. Relation of Statin Use Prior to Admission to Severity and Recovery Among COVID-19 Inpatients. Am J Cardiol 2020; 136:149-155. [PMID: 32946859 PMCID: PMC7492151 DOI: 10.1016/j.amjcard.2020.09.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 12/26/2022]
Abstract
The impact of statins, angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers (ARBs) on coronavirus disease 2019 (COVID-19) severity and recovery is important given their high prevalence of use among individuals at risk for severe COVID-19. We studied the association between use of statin/angiotensin-converting enzyme inhibitors/ARB in the month before hospital admission, with risk of severe outcome, and with time to severe outcome or disease recovery, among patients hospitalized for COVID-19. We performed a retrospective single-center study of all patients hospitalized at University of California San Diego Health between February 10, 2020 and June 17, 2020 (n = 170 hospitalized for COVID-19, n = 5,281 COVID-negative controls). Logistic regression and competing risks analyses were used to investigate progression to severe disease (death or intensive care unit admission), and time to discharge without severe disease. Severe disease occurred in 53% of COVID-positive inpatients. Median time from hospitalization to severe disease was 2 days; median time to recovery was 7 days. Statin use prior to admission was associated with reduced risk of severe COVID-19 (adjusted OR 0.29, 95%CI 0.11 to 0.71, p < 0.01) and faster time to recovery among those without severe disease (adjusted HR for recovery 2.69, 95%CI 1.36 to 5.33, p < 0.01). The association between statin use and severe disease was smaller in the COVID-negative cohort (p for interaction = 0.07). There was potential evidence of faster time to recovery with ARB use (adjusted HR 1.92, 95%CI 0.81 to 4.56). In conclusion, statin use during the 30 days prior to admission for COVID-19 was associated with a lower risk of developing severe COVID-19, and a faster time to recovery among patients without severe disease.
Collapse
|
31
|
Crimi E, Benincasa G, Figueroa-Marrero N, Galdiero M, Napoli C. Epigenetic susceptibility to severe respiratory viral infections and its therapeutic implications: a narrative review. Br J Anaesth 2020; 125:1002-1017. [PMID: 32828489 PMCID: PMC7438995 DOI: 10.1016/j.bja.2020.06.060] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
Abstract
The emergence of highly pathogenic strains of influenza virus and coronavirus (CoV) has been responsible for large epidemic and pandemic outbreaks characterised by severe pulmonary illness associated with high morbidity and mortality. One major challenge for critical care is to stratify and minimise the risk of multi-organ failure during the stay in the intensive care unit (ICU). Epigenetic-sensitive mechanisms, including deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) methylation, histone modifications, and non-coding RNAs may lead to perturbations of the host immune-related transcriptional programmes by regulating chromatin structure and gene expression patterns. Viruses causing severe pulmonary illness can use epigenetic-regulated mechanisms during host-pathogen interaction to interfere with innate and adaptive immunity, adequacy of inflammatory response, and overall outcome of viral infections. For example, Middle East respiratory syndrome-CoV and H5N1 can affect host antigen presentation through DNA methylation and histone modifications. The same mechanisms would presumably occur in patients with coronavirus disease 2019, in which tocilizumab may epigenetically reduce microvascular damage. Targeting epigenetic pathways by immune modulators (e.g. tocilizumab) or repurposed drugs (e.g. statins) may provide novel therapeutic opportunities to control viral-host interaction during critical illness. In this review, we provide an update on epigenetic-sensitive mechanisms and repurposed drugs interfering with epigenetic pathways which may be clinically suitable for risk stratification and beneficial for treatment of patients affected by severe viral respiratory infections.
Collapse
Affiliation(s)
- Ettore Crimi
- College of Medicine, University of Central Florida, Orlando, FL, USA; Department of Anesthesiology and Critical Care Medicine, Ocala Health, Ocala, FL, USA.
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, Naples, Italy
| | - Neisaliz Figueroa-Marrero
- College of Medicine, University of Central Florida, Orlando, FL, USA; Department of Anesthesiology and Critical Care Medicine, Ocala Health, Ocala, FL, USA
| | - Massimiliano Galdiero
- Department of Experimental Medicine, Section of Microbiology and Virology, University Hospital, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, Naples, Italy; IRCCS SDN, Naples, Italy
| |
Collapse
|
32
|
Xiao Z, Kong B, Yang H, Dai C, Fang J, Qin T, Huang H. Key Player in Cardiac Hypertrophy, Emphasizing the Role of Toll-Like Receptor 4. Front Cardiovasc Med 2020; 7:579036. [PMID: 33324685 PMCID: PMC7725871 DOI: 10.3389/fcvm.2020.579036] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Toll-like receptor 4 (TLR4), a key pattern recognition receptor, initiates the innate immune response and leads to chronic and acute inflammation. In the past decades, accumulating evidence has implicated TLR4-mediated inflammatory response in regulation of myocardium hypertrophic remodeling, indicating that regulation of the TLR4 signaling pathway may be an effective strategy for managing cardiac hypertrophy's pathophysiology. Given TLR4's significance, it is imperative to review the molecular mechanisms and roles underlying TLR4 signaling in cardiac hypertrophy. Here, we comprehensively review the current knowledge of TLR4-mediated inflammatory response and its interaction ligands and co-receptors, as well as activation of various intracellular signaling. We also describe the associated roles in promoting immune cell infiltration and inflammatory mediator secretion, that ultimately cause cardiac hypertrophy. Finally, we provide examples of some of the most promising drugs and new technologies that have the potential to attenuate TLR4-mediated inflammatory response and prevent or reverse the ominous cardiac hypertrophy outcomes.
Collapse
Affiliation(s)
- Zheng Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hongjie Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chang Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jin Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Tianyou Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
33
|
Kow CS, Hasan SS. Meta-analysis of Effect of Statins in Patients with COVID-19. Am J Cardiol 2020; 134:153-155. [PMID: 32891399 PMCID: PMC7419280 DOI: 10.1016/j.amjcard.2020.08.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/04/2020] [Indexed: 12/29/2022]
|
34
|
Minz MM, Bansal M, Kasliwal RR. Statins and SARS-CoV-2 disease: Current concepts and possible benefits. Diabetes Metab Syndr 2020; 14:2063-2067. [PMID: 33120281 PMCID: PMC7582042 DOI: 10.1016/j.dsx.2020.10.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS Inflammation-mediated tissue injury is the major mechanism involved in the pathogenesis of coronavirus disease 2019 (COVID-2019), caused by Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2). Statins have well-established anti-inflammatory, anti-thrombotic and immuno-modulatory effects. They may also influence viral entry into human cells. METHODS A literature search was done using PubMed and Google search engines to prepare a narrative review on this topic. RESULTS Statins interact with several different signaling pathways to exert their anti-inflammatory and vasculoprotective effects. They also variably affect cholesterol content of cell membranes and interfere with certain coronavirus enzymes involved in receptor-binding. Both these actions may influence SARS-CoV-2 entry into human cells. Statins also upregulate expression of angiotensin-converting enzyme 2 receptors on cell surfaces which may promote viral entry into the cells but at the same time, may minimize tissue injury through production of angiotensin [1-7]. The net impact of these different effects on COVID-19 pathogenesis is not clear. However, the retrospective clinical studies have shown that statin use is potentially associated with lower risk of developing severe illness and mortality and a faster time to recovery in patients with COVID-19. CONCLUSIONS Early observations suggest beneficial effect of statin use on the clinical outcomes in COVID-19. Prospective randomized studies as well as well-designed laboratory studies are required to confirm these observations and to elucidate the mechanisms of such benefits, if proven.
Collapse
Affiliation(s)
- Madhu Mary Minz
- Department of Cardiology, Medanta- the Medicity, Gurgaon, India
| | - Manish Bansal
- Department of Cardiology, Medanta- the Medicity, Gurgaon, India.
| | - Ravi R Kasliwal
- Department of Cardiology, Medanta- the Medicity, Gurgaon, India
| |
Collapse
|
35
|
Rizk JG, Kalantar-Zadeh K, Mehra MR, Lavie CJ, Rizk Y, Forthal DN. Pharmaco-Immunomodulatory Therapy in COVID-19. Drugs 2020; 80:1267-1292. [PMID: 32696108 PMCID: PMC7372203 DOI: 10.1007/s40265-020-01367-z] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 associated coronavirus disease 2019 (COVID-19) illness is a syndrome of viral replication in concert with a host inflammatory response. The cytokine storm and viral evasion of cellular immune responses may play an equally important role in the pathogenesis, clinical manifestation, and outcomes of COVID-19. Systemic proinflammatory cytokines and biomarkers are elevated as the disease progresses towards its advanced stages, and correlate with worse chances of survival. Immune modulators have the potential to inhibit cytokines and treat the cytokine storm. A literature search using PubMed, Google Scholar, and ClinicalTrials.gov was conducted through 8 July 2020 using the search terms ‘coronavirus’, ‘immunology’, ‘cytokine storm’, ‘immunomodulators’, ‘pharmacology’, ‘severe acute respiratory syndrome 2’, ‘SARS-CoV-2’, and ‘COVID-19’. Specific immune modulators include anti-cytokines such as interleukin (IL)-1 and IL-6 receptor antagonists (e.g. anakinra, tocilizumab, sarilumab, siltuximab), Janus kinase (JAK) inhibitors (e.g. baricitinib, ruxolitinib), anti-tumor necrosis factor-α (e.g. adalimumab, infliximab), granulocyte–macrophage colony-stimulating factors (e.g. gimsilumab, lenzilumab, namilumab), and convalescent plasma, with promising to negative trials and other data. Non-specific immune modulators include human immunoglobulin, corticosteroids such as dexamethasone, interferons, statins, angiotensin pathway modulators, macrolides (e.g. azithromycin, clarithromycin), hydroxychloroquine and chloroquine, colchicine, and prostaglandin D2 modulators such as ramatroban. Dexamethasone 6 mg once daily (either by mouth or by intravenous injection) for 10 days may result in a reduction in mortality in COVID-19 patients by one-third for patients on ventilators, and by one-fifth for those receiving oxygen. Research efforts should focus not only on the most relevant immunomodulatory strategies but also on the optimal timing of such interventions to maximize therapeutic outcomes. In this review, we discuss the potential role and safety of these agents in the management of severe COVID-19, and their impact on survival and clinical symptoms.
Collapse
Affiliation(s)
- John G Rizk
- Edson College, Arizona State University, Phoenix, AZ, USA.
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology, Hypertension and Kidney Transplantation, University of California, Irvine, School of Medicine, Irvine, CA, USA.,Department of Epidemiology, University of California, Los Angeles, UCLA Fielding School of Public Health, Los Angeles, CA, USA.,Tibor Rubin VA Long Beach Healthcare System, Long Beach, CA, USA
| | - Mandeep R Mehra
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, LA, USA
| | - Youssef Rizk
- Department of Family Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Donald N Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA.,Department of Molecular Biology and Biochemistry, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|
36
|
Subir R, Jagat J M, Kalyan K G. Pros and cons for use of statins in people with coronavirus disease-19 (COVID-19). Diabetes Metab Syndr 2020; 14:1225-1229. [PMID: 32683320 PMCID: PMC7352102 DOI: 10.1016/j.dsx.2020.07.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/04/2020] [Accepted: 07/08/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND AIMS Morbidity and mortality from coronavirus disease 2019 (COVID-19) is higher among people with diabetes mellitus (DM), hypertension, and cardiovascular disease (CVD). Statins are used in the majority of people with DM and CVD. This mini-review discusses the current understanding of benefit-risk ratio of use of statins in COVID-19. METHODS We searched PubMed database using specific keywords related to our aims till June 12, 2020. Full text of relevant articles published in English language were retrieved and reviewed. RESULTS Statins, with their immunomodulatory, anti-inflammatory, anti-thrombotic, and anti-oxidant properties, have the potential to reduce severity of lung injury in, and mortality from, severe acute respiratory syndrome-coronavirus 2 (SARS-CoV2) infections. Statin-induced upregulation of angiotensin-converting enzyme-2 (ACE2) has the potential to reduce lung injury from excess angiotensin II. By disrupting lipid rafts, statins have the potential to reduce viral entry into cells. However, benefit-risk ratio of its complex interaction with MYD88 gene expression on outcomes in COVID-19, and the putative role of low serum LDL cholesterol in increasing severity of SARS-CoV2 infection need further clarification. CONCLUSIONS People with COVID-19, who are already on statins for an underlying co-morbid condition, should continue on it unless there are specific contraindications. De-novo use of statins in people with COVID-19 with no underlying co-morbidity might be beneficial but awaits substantiation in clinical trials; till that time, de novo use of statins in COVID 19 should be limited to a clinical trial setting.
Collapse
Affiliation(s)
- Ray Subir
- Department of Endocrinology and Diabetes, Apollo Gleneagles Hospitals, Kolkata, India
| | - Mukherjee Jagat J
- Department of Endocrinology and Diabetes, Apollo Gleneagles Hospitals, Kolkata, India
| | | |
Collapse
|
37
|
Zhang XJ, Qin JJ, Cheng X, Shen L, Zhao YC, Yuan Y, Lei F, Chen MM, Yang H, Bai L, Song X, Lin L, Xia M, Zhou F, Zhou J, She ZG, Zhu L, Ma X, Xu Q, Ye P, Chen G, Liu L, Mao W, Yan Y, Xiao B, Lu Z, Peng G, Liu M, Yang J, Yang L, Zhang C, Lu H, Xia X, Wang D, Liao X, Wei X, Zhang BH, Zhang X, Yang J, Zhao GN, Zhang P, Liu PP, Loomba R, Ji YX, Xia J, Wang Y, Cai J, Guo J, Li H. In-Hospital Use of Statins Is Associated with a Reduced Risk of Mortality among Individuals with COVID-19. Cell Metab 2020; 32:176-187.e4. [PMID: 32592657 PMCID: PMC7311917 DOI: 10.1016/j.cmet.2020.06.015] [Citation(s) in RCA: 356] [Impact Index Per Article: 71.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Statins are lipid-lowering therapeutics with favorable anti-inflammatory profiles and have been proposed as an adjunct therapy for COVID-19. However, statins may increase the risk of SARS-CoV-2 viral entry by inducing ACE2 expression. Here, we performed a retrospective study on 13,981 patients with COVID-19 in Hubei Province, China, among which 1,219 received statins. Based on a mixed-effect Cox model after propensity score-matching, we found that the risk for 28-day all-cause mortality was 5.2% and 9.4% in the matched statin and non-statin groups, respectively, with an adjusted hazard ratio of 0.58. The statin use-associated lower risk of mortality was also observed in the Cox time-varying model and marginal structural model analysis. These results give support for the completion of ongoing prospective studies and randomized controlled trials involving statin treatment for COVID-19, which are needed to further validate the utility of this class of drugs to combat the mortality of this pandemic.
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Lijun Shen
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Yan-Ci Zhao
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fang Lei
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Ming-Ming Chen
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Huilin Yang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Liangjie Bai
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xiaohui Song
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Lijin Lin
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Meng Xia
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Feng Zhou
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianghua Zhou
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Lihua Zhu
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Qingbo Xu
- Centre for Clinic Pharmacology, The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Ping Ye
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430072, China
| | - Guohua Chen
- Department of Neurology, Wuhan First Hospital/Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan 430072, China
| | - Liming Liu
- Department of General Surgery, Ezhou Central Hospital, Ezhou 436000, China
| | - Weiming Mao
- Department of General Surgery, Huanggang Central Hospital, Huanggang 438000, China
| | - Youqin Yan
- Wuhan Seventh Hospital, Wuhan 430072, China
| | - Bing Xiao
- Department of Stomatology, Xiantao First People's Hospital, Xiantao 433000, China
| | - Zhigang Lu
- Department of Neurology, The First People's Hospital of Jingmen Affiliated to Hubei Minzu University, Jingmen 448000, China
| | - Gang Peng
- Department of Hepatobiliary and Pancreatic Surgery, Suizhou Central Hospital Affiliated to Hubei Medical College, Suizhou 441300, China
| | - Mingyu Liu
- The Ninth Hospital of Wuhan City, Wuhan 430072, China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital and Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China
| | - Luyu Yang
- Department of Intensive Care Unit, Wuhan Third Hospital & Tongren Hospital of Wuhan University, Wuhan, China
| | - Changjiang Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi 445000, China
| | - Haofeng Lu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Changjiang University, Jingzhou, China
| | - Xigang Xia
- Department of Hepatobiliary Surgery, Jingzhou Central Hospital, Jingzhou, China
| | - Daihong Wang
- Department of Hepatobiliary and Pancreatic Surgery, Xianning Central Hospital, Xianning, Hubei Province, China
| | - Xiaofeng Liao
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing-Hong Zhang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Juan Yang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Guang-Nian Zhao
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peter P Liu
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Epidemiology, University of California, San Diego, San Diego, CA, USA
| | - Yan-Xiao Ji
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yibin Wang
- Departments of Anesthesiology, Physiology, and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410000, China.
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine & Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China & Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
38
|
Abstract
The COVID-19 pandemic has driven unprecedented efforts to identify existing treatments that can be quickly and effectively repurposed to reduce morbidity and mortality. In this issue of Cell Metabolism, Zhang et al. (2020) report an association between statin use and improved outcomes in a large observational study of hospitalized COVID-19 patients. Given the widespread availability, low cost, and safety of statins, this promising result should be further investigated in randomized controlled trials.
Collapse
Affiliation(s)
- David C Fajgenbaum
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Kow CS, Thiruchelvam K, Hasan SS. Pharmacotherapeutic considerations for the management of cardiovascular diseases among hospitalized COVID-19 patients. Expert Rev Cardiovasc Ther 2020; 18:475-485. [PMID: 32700573 DOI: 10.1080/14779072.2020.1797492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Cardiovascular diseases (CVDs) are among the most frequently identified comorbidities in hospitalized patients with COVID-19. Patients with CV comorbidities are typically prescribed with long-term medications. We reviewed the management of co-medications prescribed for CVDs among hospitalized COVID-19 patients. AREAS COVERED There is no specific contraindication or caution related to COVID-19 on the use of antihypertensives unless patients develop severe hypotension from septic shock where all antihypertensives should be discontinued or severe hyperkalemia in which continuation of renin-angiotensin system inhibitors is not desired. The continuation of antiplatelet or statin is not desired when severe thrombocytopenia or severe transminitis develop, respectively. Patients with atrial fibrillation receiving oral anticoagulants, particularly those who are critically ill, should be considered for substitution to parenteral anticoagulants. EXPERT OPINION An individualized approach to medication management among hospitalized COVID-19 patients with concurrent CVDs would seem prudent with attention paid to changes in clinical conditions and medications intended for COVID-19. The decision to modify prescribed long-term CV medications should be entailed by close follow-up to check if a revision on the decision is needed, with resumption of any long-term CV medication before discharge if it is discontinued during hospitalization for COVID-19, to ensure continuity of care.
Collapse
Affiliation(s)
- Chia Siang Kow
- School of Postgraduate Studies, International Medical University , Kuala Lumpur, Malaysia
| | | | - Syed Shahzad Hasan
- School of Biomedical Sciences & Pharmacy, University of Newcastle , Callaghan, Australia.,Department of Pharmacy, University of Huddersfield , Huddersfield, UK
| |
Collapse
|
40
|
Castiglione V, Chiriacò M, Emdin M, Taddei S, Vergaro G. Statin therapy in COVID-19 infection. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2020; 6:258-259. [PMID: 32347925 PMCID: PMC7197622 DOI: 10.1093/ehjcvp/pvaa042] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 01/09/2023]
Affiliation(s)
| | - Martina Chiriacò
- Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.,Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michele Emdin
- Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.,Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giuseppe Vergaro
- Institute of Life Science, Scuola Superiore Sant'Anna, Pisa, Italy.,Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| |
Collapse
|
41
|
Abuo-Rahma GEDA, Mohamed MFA, Ibrahim TS, Shoman ME, Samir E, Abd El-Baky RM. Potential repurposed SARS-CoV-2 (COVID-19) infection drugs. RSC Adv 2020; 10:26895-26916. [PMID: 35515773 PMCID: PMC9055522 DOI: 10.1039/d0ra05821a] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 01/09/2023] Open
Abstract
The global outbreak of COVID-19 viral infection is associated with the absence of specific drug(s) for fighting this viral infection. About 10 million people are already infected, about 500 000 deaths all over the world to date. Great efforts have been made to find solutions for this viral infection, either vaccines, monoclonal antibodies, or small molecule drugs; this can stop the spread of infection to avoid the expected human, economic and social catastrophe associated with this infection. In the literature and during clinical trials in hospitals, several FDA approved drugs for different diseases have the potential to treat or reduce the severity of COVID-19. Repurposing of these drugs as potential agents to treat COVID-19 reduces the time and cost to find effective COVID-19 agents. This review article summarizes the present situation of transmission, pathogenesis and statistics of COVID-19 in the world. Moreover, it includes chemistry, mechanism of action at the molecular level of the possible drug molecules which are liable for redirection as potential COVID-19 therapeutic agents. This includes polymerase inhibitors, protease inhibitors, malaria drugs, lipid lowering statins, rheumatoid arthritis drugs and some miscellaneous agents. We offer research data and knowledge about the chemistry and biology of potential COVID-19 drugs for the research community in this field.
Collapse
Affiliation(s)
- Gamal El-Din A Abuo-Rahma
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University 61519 Minia Egypt +20 1003069431
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University 82524 Sohag Egypt
| | - Tarek S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University Jeddah 21589 Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University Zagazig 44519 Egypt
| | - Mai E Shoman
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University 61519 Minia Egypt +20 1003069431
| | - Ebtihal Samir
- Physical Chemistry, Department of Analytical Chemistry, Faculty of Pharmacy, Deraya University Minia 11566 Egypt
| | - Rehab M Abd El-Baky
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University 61519 Minia Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University Minia 11566 Egypt
| |
Collapse
|
42
|
Suehiro H, Fukuzawa K, Yoshida N, Kiuchi K, Takami M, Akita T, Tabata T, Takemoto M, Sakai J, Nakamura T, Yatomi A, Takahara H, Sonoda Y, Nakasone K, Yamamoto K, Suzuki A, Yamashita T, Hirata KI. Circulating intermediate monocytes and toll-like receptor 4 correlate with low-voltage zones in atrial fibrillation. Heart Vessels 2020; 35:1717-1726. [DOI: 10.1007/s00380-020-01647-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/05/2020] [Indexed: 01/06/2023]
|
43
|
Lin ZP, Lin HL, Yu XP, Zheng YJ, Cheng SY. TLR4 mediates inflammation and hepatic fibrosis induced by chronic intermittent hypoxia in rats. Mol Med Rep 2020; 22:651-660. [PMID: 32626927 PMCID: PMC7339543 DOI: 10.3892/mmr.2020.11134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/05/2020] [Indexed: 12/24/2022] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a common and complex disorder that is associated with liver injury. Moreover, previous studies have revealed that chronic intermittent hypoxia (CIH) is associated with the development of non-alcoholic fatty liver disease and hepatic fibrosis. However, the underlying molecular mechanisms remain largely unknown. The present study aimed to investigate whether chronic intermittent hypoxia induced hepatic fibrosis, in addition to determining its underlying mechanisms, in CIH model rats using immunohistochemistry, western blotting and reverse transcription-quantitative PCR. The present results suggested that CIH caused hepatic fibrosis and increased the expression levels of interleukin (IL)-1β, IL-8, monocyte chemotactic-1, tumor necrosis factor-α, intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in the liver; these conditions could be reversed by Toll-like receptor 4 (TLR4) short hairpin RNA lentivirus treatment. Moreover, immunohistochemistry and western blotting results indicated that TLR4 and NF-κB expression levels were significantly increased in the CIH and CIH-TLR4 empty vector lentivirus group. However, protein expression levels of TLR4, NF-κB, inhibitor of NF-κB and phosphorylated-mitogen-activated protein kinase (MAPK)-1 in the hypoxia/reoxygenation group were significantly higher compared with the control group (P<0.05), and these results were reversed by the MAPK inhibitor U0126 in vitro. Collectively, the present preliminary results suggested that inflammation and the TLR4/NF-κB/MAPK signaling pathway may be involved in CIH-induced liver fibrosis.
Collapse
Affiliation(s)
- Zhi-Peng Lin
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Hui-Li Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Xue-Ping Yu
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Yi-Juan Zheng
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Si-Yu Cheng
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| |
Collapse
|
44
|
Koushki K, Shahbaz SK, Mashayekhi K, Sadeghi M, Zayeri ZD, Taba MY, Banach M, Al-Rasadi K, Johnston TP, Sahebkar A. Anti-inflammatory Action of Statins in Cardiovascular Disease: the Role of Inflammasome and Toll-Like Receptor Pathways. Clin Rev Allergy Immunol 2020; 60:175-199. [PMID: 32378144 PMCID: PMC7985098 DOI: 10.1007/s12016-020-08791-9] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis is one type of cardiovascular disease (CVD) in which activation of the NLRP3 inflammasome and toll-like receptor (TLR) pathways is implicated. One of the most effective treatments for atherosclerosis is the use of statin medications. Recent studies have indicated that statins, in addition to their lipid-lowering effects, exert inhibitory and/or stimulatory effects on the NLRP3 inflammasome and TLRs. Some of the statins lead to activation of the inflammasome and subsequently cause secretion of IL-1β and IL-18. Thus, these actions may further aggravate the disease. On the other hand, some statins cause inhibition of the inflammasome or TLRs and along with lipid-lowering, help to improve the disease by reducing inflammation. In this article, we discuss these contradictory studies and the mechanisms of action of statins on the NLRP3 inflammasome and TLR pathways. The dose-dependent effects of statins on the NLRP3 complex are related to their chemistry, pharmacokinetic properties, and danger signals. Lipophilic statins have more pleiotropic effects on the NLRP3 complex in comparison to hydrophilic statins. Statins can suppress TLR4/MyD88/NF-ĸB signaling and cause an immune response shift to an anti-inflammatory response. Furthermore, statins inhibit the NF-ĸB pathway by decreasing the expression of TLRs 2 and 4. Statins are cost-effective drugs, which should have a continued future in the treatment of atherosclerosis due to both their immune-modulating and lipid-lowering effects.
Collapse
Affiliation(s)
- Khadijeh Koushki
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sanaz Keshavarz Shahbaz
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Mashayekhi
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeinab Deris Zayeri
- Golestan Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Khalid Al-Rasadi
- Medical Research Centre, Sultan Qaboos University, Muscat, Oman
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
45
|
Dashti‐Khavidaki S, Khalili H. Considerations for Statin Therapy in Patients with COVID-19. Pharmacotherapy 2020; 40:484-486. [PMID: 32267560 PMCID: PMC7262253 DOI: 10.1002/phar.2397] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Simin Dashti‐Khavidaki
- Department of Clinical PharmacyFaculty of PharmacyTehran University of Medical SciencesTehranIran
| | - Hossein Khalili
- Department of Clinical PharmacyFaculty of PharmacyTehran University of Medical SciencesTehranIran
| |
Collapse
|
46
|
Role of the Nrdp1 in Brain Injury Induced by Chronic Intermittent Hypoxia in Rats via Regulating the Protein Levels of ErbB3. Neurotox Res 2020; 38:124-132. [PMID: 32200526 DOI: 10.1007/s12640-020-00195-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/21/2020] [Accepted: 03/13/2020] [Indexed: 01/27/2023]
Abstract
Obstructive sleep apnea syndrome (OSAS) is known as a repeated obstruction of the upper airway during sleep, leading to generalized hypoxia episodes and associated with cardiovascular and cerebrovascular diseases. We mainly explored the role of neuregulin receptor degradation protein-1 (Nrdp1, also known as FLRF) in brain injury induced by chronic intermittent hypoxia (CIH) in rats. Wistar rats were randomly divided into 4 groups (n = 12 per group), including the sham + adeno-associated virus-NC (AAV-NC) group, the sham + AAV-siNrdp1 group, the IH-4w (intermittent hypoxia for 4 weeks) + AAV-NC group, and the IH-4w + AAV-siNrdp1 group. Morphologic changes in brain tissue were observed by hematoxylin and eosin (HE) staining. Apoptosis in the hippocampus was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining. Spatial learning and memory were assessed by the Morris water maze test. The expression of Nrdp1 mRNA and protein in the hippocampus was detected by qualitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. The concentration of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in serum was detected via enzyme-linked immunosorbent assay (ELISA) kits. Nrdp1 expression was increased after intermittent hypoxia exposure over time. Western blotting and H&E results showed that pathological changes of hippocampus neurons in chronic intermittent hypoxia rat were diminished by shNrdp1. Western blotting and TUNEL staining showed that apoptotic cells in the hippocampus of CIH rats were decreased by shNrdp1. The Morris water maze results proved that shNrdp1 improved spatial learning performance of chronic intermittent hypoxia rats. ELISA kits results showed that CIH-induced inflammatory response was decreased by shNrdp1. Western blotting and qRT-PCR results showed protein expression of ErbB3 in the hippocampus of CIH rats. Nrdp1 could regulate ErbB3 protein levels in brain-injured rats with CIH, which demonstrates that Nrdp1 is a potential therapeutic target in the cognition deficits associated with OSAS.
Collapse
|
47
|
Deng Y, Liu K, Pan Y, Ren J, Shang J, Chen L, Liu H. TLR2 antagonism attenuates the hippocampal neuronal damage in a murine model of sleep apnea via inhibiting neuroinflammation and oxidative stress. Sleep Breath 2020; 24:1613-1621. [PMID: 32170671 DOI: 10.1007/s11325-020-02030-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 01/28/2020] [Accepted: 02/05/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) in humans chronically promotes the neuronal damage in the hippocampus. Toll-like receptor 2 (TLR2) is pivotal for the development of numerous hippocampal diseases. Chronic intermittent hypoxia (CIH) is a prominent feature of OSA. Here in our study, the effects of TLR2 antagonism on the neural damage elicited by CIH were examined. METHODS Ortho-vanillin (O-vanillin) is an inhibitor of TLR2. Adult male mice were subjected to 8 h of intermittent hypoxia per day with or without O-vanillin for 28 days. Neuronal damage, the number of microglia, the interaction of TLR2 with its adapter protein myeloid differentiation factor 88 (MYD88), the expressions of inflammatory cytokines, and the oxidative stress were observed. RESULTS O-vanillin inhibited the increased interaction of TLR2 and MyD88, the activation of NFκB, the aggregation of microglia, the overexpression of proinflammatory agents, the elevation of oxidative stress, and hippocampal neuron cell apoptosis induced by CIH. CONCLUSIONS Our experiments indicate that TLR2 antagonism may alleviate the hippocampal neuronal damage caused by CIH via inhibiting neuroinflammation and oxidative stress.
Collapse
Affiliation(s)
- Yan Deng
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Kui Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Yueying Pan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Jie Ren
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Jin Shang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Lei Chen
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China.
| |
Collapse
|
48
|
Peña-Ortega F. Clinical and experimental aspects of breathing modulation by inflammation. Auton Neurosci 2018; 216:72-86. [PMID: 30503161 DOI: 10.1016/j.autneu.2018.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 12/19/2022]
Abstract
Neuroinflammation is produced by local or systemic alterations and mediated mainly by glia, affecting the activity of various neural circuits including those involved in breathing rhythm generation and control. Several pathological conditions, such as sudden infant death syndrome, obstructive sleep apnea and asthma exert an inflammatory influence on breathing-related circuits. Consequently breathing (both resting and ventilatory responses to physiological challenges), is affected; e.g., responses to hypoxia and hypercapnia are compromised. Moreover, inflammation can induce long-lasting changes in breathing and affect adaptive plasticity; e.g., hypoxic acclimatization or long-term facilitation. Mediators of the influences of inflammation on breathing are most likely proinflammatory molecules such as cytokines and prostaglandins. The focus of this review is to summarize the available information concerning the modulation of the breathing function by inflammation and the cellular and molecular aspects of this process. I will consider: 1) some clinical and experimental conditions in which inflammation influences breathing; 2) the variety of experimental approaches used to understand this inflammatory modulation; 3) the likely cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO 76230, México.
| |
Collapse
|
49
|
Bahrami A, Parsamanesh N, Atkin SL, Banach M, Sahebkar A. Effect of statins on toll-like receptors: a new insight to pleiotropic effects. Pharmacol Res 2018; 135:230-238. [PMID: 30120976 DOI: 10.1016/j.phrs.2018.08.014] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/27/2022]
Abstract
The toll-like receptors (TLRs) are a class of transmembrane-spanning receptors that are sentinels of both innate and adaptive immunity. Statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) are the most commonly prescribed therapeutic agents for treating hypercholesterolemia globally. However, statin therapy appears to have pleiotropic effects including attenuation of chronic low-grade inflammation and modulation of TLR activity. Statins through abolition of TLR4 expression and regulation of the TLR4/Myd88/NF-κB signaling pathway may slow the progression of atherosclerosis and other inflammatory diseases. In this review, we have focused on the impact and mechanism of action of statins on cardiovascular and non-cardiovascular diseases.
Collapse
Affiliation(s)
- Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Negin Parsamanesh
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
50
|
Zhang Y, Su X, Zou F, Xu T, Pan P, Hu C. Toll-like receptor-4 deficiency alleviates chronic intermittent hypoxia-induced renal injury, inflammation, and fibrosis. Sleep Breath 2018; 23:503-513. [PMID: 30099700 DOI: 10.1007/s11325-018-1704-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Obstructive sleep apnea (OSA)-associated chronic kidney disease is mainly caused by chronic intermittent hypoxia (CIH) triggered renal damage. This study aims to investigate the role of toll-like receptor-4 (TLR4) in underlying mechanism involved chronic intermittent hypoxia (CIH)-induced renal damage. METHODS C57BL/6J mice with normal TLR4 (TLR4 WT) or deficient TLR4 (TLR4 KO) were divided into four groups and exposed to normal air (NA) and CIH: TLR4 WT + NA, TLR4 KO + NA, TLR4 WT + CIH, and TLR4 KO + CIH. CIH lasted for 8 h/day and 7 days/week for 6 weeks. Renal injury and inflammation were evaluated by histology and ELISA. Renal tubular apoptosis, macrophages, and fibroblasts recruitment were determined by TUNEL assay, immunofluorescence, and western blot. RESULTS In response to CIH, TLR4 deficiency alleviated renal histological injury, renal dysfunction, and fibrosis. TLR4 deficiency ameliorated renal dysfunction (serum BUN and creatinine) and tubular endothelial apoptosis determined by immunofluorescence staining of CD31 and TUNEL, and western blot of apoptotic protein (caspase-3, c-caspase-3, and Bax/Bcl-2 ratio). Furthermore, we also found TLR4 deficiency abrogated CIH-induced macrophages (CD68) and fibroblasts (α-SMA) recruitment, further reducing expression of extra-cellular matrix protein (collagen I and collagen IV) and inflammatory cytokines release (IL-6, TNF-α, and MCP-1). Finally, we used immunohistochemistry to demonstrate that TLR4 deficiency attenuated increased expression of MyD88 and NF-kB p65 after CIH treatment. CONCLUSIONS Our data suggest that TLR4 plays a vital role in CIH-induced renal injury, inflammation and fibrosis, and inhibition of TLR4 probably provides a therapeutic potential for CIH-induced kidney damage.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Respiratory Medicine, Xiangya Hospital, Key Cite of National Clinical Research Center for Respiratory Disease, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xiaoli Su
- Department of Respiratory Medicine, Xiangya Hospital, Key Cite of National Clinical Research Center for Respiratory Disease, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Fangfang Zou
- Department of Respiratory Medicine, Xiangya Hospital, Key Cite of National Clinical Research Center for Respiratory Disease, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Tengjuan Xu
- Department of Respiratory Medicine, Xiangya Hospital, Key Cite of National Clinical Research Center for Respiratory Disease, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Pinhua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Key Cite of National Clinical Research Center for Respiratory Disease, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Key Cite of National Clinical Research Center for Respiratory Disease, Central South University, Changsha, 410008, Hunan, People's Republic of China
| |
Collapse
|