1
|
Cheung J, Cheung BMY, Yiu KH, Tse HF, Chan YH. Role of metabolic dysfunction-associated fatty liver disease in atrial fibrillation and heart failure: molecular and clinical aspects. Front Cardiovasc Med 2025; 12:1573841. [PMID: 40264510 PMCID: PMC12011764 DOI: 10.3389/fcvm.2025.1573841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MASLD) is a rising global health concern. In addition to direct hepatic complications, extra-hepatic complications, including cardiovascular diseases (CVD), type 2 diabetes (T2D), gastroesophageal reflux disease, chronic kidney disease and some malignancies, are increasingly recognized. CVD, including atrial fibrillation (AF) and heart failure (HF), is the leading cause of death in patients with MASLD. External factors, including excess energy intake, sedentary lifestyle and xenobiotic use, induce inflammation-related complications. MASLD, AF, and HF are associated with immune system activation, including the reprogramming of immune cells and the establishment of immune memory. Emerging evidence suggests that the heart and the liver cross-talk with each other through the diverse spectrum of autocrine, paracrine and endocrine mechanisms. Pro-inflammatory cytokines produced from the liver and the heart circulate systemically to orchestrate metabolic derangements that promote the systematic immune dysregulation in the heart-liver axis and the development of end-organ complications. Cardio-hepatic syndrome describes the clinical and biochemical evidence of hepatic dysfunction and cardiac pathology due to the interaction between the heart and the liver. Activation of inflammatory cascades, oxidative stress and immune system dysregulation underlie key mechanisms in bringing about such pathological changes. This review focuses on the current clinical and molecular evidence about the heart-liver cross-talk. It summarizes the epidemiological and pathophysiological associations of MASLD, AF and HF. In addition, we will discuss how repurposing currently available and emerging pharmacotherapies may help tackle the cardiovascular risks resulting from MASLD.
Collapse
Affiliation(s)
- Jamie Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Bernard Man-Yung Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kai-Hang Yiu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, Shenzhen Hong Kong University Hospital, Hong Kong SAR, China
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, Shenzhen Hong Kong University Hospital, Hong Kong SAR, China
| | - Yap-Hang Chan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Experimental Medicine and Immunotherapeutics, Cambridge University Hospitals NHS Foundation Trust/University of Cambridge, Cambridge, United Kingdom
- Department of Cardiology, Royal Papworth Hospital, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
2
|
Hill AP, Iqbal SB, Case BC, Shankar AA, Merdler I. Acute Coronary Syndrome and Rheumatic Disease. J Clin Med 2025; 14:1490. [PMID: 40094966 PMCID: PMC11900616 DOI: 10.3390/jcm14051490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Patients with rheumatic disease and immune disorders have been noted to show an earlier development of atherosclerosis and to present with acute coronary syndrome. These diseases disproportionately affect women, and patients frequently have a higher number of comorbidities and other risk factors. Inflammation has long been known to play a role in the development of coronary artery disease. Early studies have shown some benefit in treatment targeting inflammation. While this has not been realized for all populations, there remains potential in treatment with targeted and individualized therapies. Especially since these diseases are associated with a worse prognosis, management benefits from the multidisciplinary expertise of cardiologists, rheumatologists, and other providers. However, the prevention and treatment of underlying rheumatic disease remains essential. This review will seek to highlight prior studies and future directions in the treatment of acute coronary syndrome in patients with rheumatologic disease.
Collapse
Affiliation(s)
- Andrew P. Hill
- Section of Cardiology, MedStar Washington Hospital Center and Georgetown University, Washington, DC 20007, USA;
| | - Shaikh B. Iqbal
- Section of Internal Medicine, MedStar Union Memorial Hospital and Georgetown University, Washington, DC 20007, USA;
| | - Brian C. Case
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC 20010, USA;
| | | | | |
Collapse
|
3
|
Cairns M, Marais E, Joseph D, Essop MF. The Role of Chronic Stress in the Pathogenesis of Ischemic Heart Disease in Women. Compr Physiol 2025; 15:e70000. [PMID: 39903543 PMCID: PMC11793136 DOI: 10.1002/cph4.70000] [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: 10/16/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Psychological stress has emerged as a critical risk factor for cardiovascular disease, especially in women. While female participation in clinical research has improved, sex-specific data analysis and reporting often remain inadequate, limiting our ability to draw definitive conclusions for women. Conversely, preclinical studies consistently demonstrate adverse effects of stress on female health, yet the molecular mechanisms underlying this association remain elusive. Evidence suggests that female IHD pathogenesis is more complex than in males, involving multiple factors, including inflammation, contractile dysfunction, bioenergetic impairment, and remodeling. However, many of these mechanisms are primarily derived from male studies, and molecular investigations in female models are limited, hindering our understanding of the underlying biological pathways. This is particularly concerning given the increasing prevalence of ischemic heart disease in postmenopausal women. In order to fully elucidate the impact of stress on female cardiac health and develop targeted interventions, further preclinical research on female models is essential.
Collapse
Affiliation(s)
- Megan Cairns
- Division of Medical PhysiologyCentre for Cardio‐Metabolic Research in Africa (CARMA)Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Erna Marais
- Division of Medical PhysiologyCentre for Cardio‐Metabolic Research in Africa (CARMA)Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Danzil Joseph
- Department of Physiological Sciences, Center for Cardio‐Metabolic Research in Africa (CARMA)Stellenbosch UniversityStellenboschSouth Africa
| | - M. Faadiel Essop
- Division of Medical PhysiologyCentre for Cardio‐Metabolic Research in Africa (CARMA)Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| |
Collapse
|
4
|
Duenas S, McGee Z, Mhatre I, Mayilvahanan K, Patel KK, Abdelhalim H, Jayprakash A, Wasif U, Nwankwo O, Degroat W, Yanamala N, Sengupta PP, Fine D, Ahmed Z. Computational approaches to investigate the relationship between periodontitis and cardiovascular diseases for precision medicine. Hum Genomics 2024; 18:116. [PMID: 39427205 PMCID: PMC11491019 DOI: 10.1186/s40246-024-00685-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
Periodontitis is a highly prevalent inflammatory illness that leads to the destruction of tooth supporting tissue structures and has been associated with an increased risk of cardiovascular disease (CVD). Precision medicine, an emerging branch of medical treatment, aims can further improve current traditional treatment by personalizing care based on one's environment, genetic makeup, and lifestyle. Genomic databases have paved the way for precision medicine by elucidating the pathophysiology of complex, heritable diseases. Therefore, the investigation of novel periodontitis-linked genes associated with CVD will enhance our understanding of their linkage and related biochemical pathways for targeted therapies. In this article, we highlight possible mechanisms of actions connecting PD and CVD. Furthermore, we delve deeper into certain heritable inflammatory-associated pathways linking the two. The goal is to gather, compare, and assess high-quality scientific literature alongside genomic datasets that seek to establish a link between periodontitis and CVD. The scope is focused on the most up to date and authentic literature published within the last 10 years, indexed and available from PubMed Central, that analyzes periodontitis-associated genes linked to CVD. Based on the comparative analysis criteria, fifty-one genes associated with both periodontitis and CVD were identified and reported. The prevalence of genes associated with both CVD and periodontitis warrants investigation to assess the validity of a potential linkage between the pathophysiology of both diseases.
Collapse
Affiliation(s)
- Sophia Duenas
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Zachary McGee
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Ishani Mhatre
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Karthikeyan Mayilvahanan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Kush Ketan Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Atharv Jayprakash
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Uzayr Wasif
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Oluchi Nwankwo
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - William Degroat
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Naveena Yanamala
- Division of Cardiovascular Diseases and Hypertension, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
| | - Partho P Sengupta
- Division of Cardiovascular Diseases and Hypertension, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
| | - Daniel Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, 110 Bergen Street, Newark, NJ, US
| | - Zeeshan Ahmed
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA.
- Division of Cardiovascular Diseases and Hypertension, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA.
- Department of Medicine, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
5
|
Baumgardt SL, Fang J, Fu X, Liu Y, Xia Z, Zhao M, Chen L, Mishra R, Gunasekaran M, Saha P, Forbess JM, Bosnjak ZJ, Camara AKS, Kersten JR, Thorp EB, Kaushal S, Ge ZD. Genetic deletion or pharmacologic inhibition of histone deacetylase 6 protects the heart against ischaemia/reperfusion injury by limiting tumour necrosis factor alpha-induced mitochondrial injury in experimental diabetes. Cardiovasc Res 2024; 120:1456-1471. [PMID: 39001869 PMCID: PMC11472425 DOI: 10.1093/cvr/cvae144] [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: 04/20/2023] [Revised: 09/01/2023] [Accepted: 04/06/2024] [Indexed: 07/15/2024] Open
Abstract
AIMS The histone deacetylase 6 (HDAC6) inhibitor, tubastatin A (TubA), reduces myocardial ischaemia/reperfusion injury (MIRI) in type 1 diabetic rats. It remains unclear whether HDAC6 regulates MIRI in type 2 diabetic animals. Diabetes augments the activity of HDAC6 and the generation of tumour necrosis factor alpha (TNF-α) and impairs mitochondrial complex I (mCI). Here, we examined how HDAC6 regulates TNF-α production, mCI activity, mitochondria, and cardiac function in type 1 and type 2 diabetic mice undergoing MIRI. METHODS AND RESULTS HDAC6 knockout, streptozotocin-induced type 1 diabetic, and obese type 2 diabetic db/db mice underwent MIRI in vivo or ex vivo in a Langendorff-perfused system. We found that MIRI and diabetes additively augmented myocardial HDAC6 activity and generation of TNF-α, along with cardiac mitochondrial fission, low bioactivity of mCI, and low production of adenosine triphosphate. Importantly, genetic disruption of HDAC6 or TubA decreased TNF-α levels, mitochondrial fission, and myocardial mitochondrial nicotinamide adenine dinucleotide levels in ischaemic/reperfused diabetic mice, concomitant with augmented mCI activity, decreased infarct size, and improved cardiac function. Moreover, HDAC6 knockout or TubA treatment decreased left ventricular dilation and improved cardiac systolic function 28 days after MIRI. H9c2 cardiomyocytes with and without HDAC6 knockdown were subjected to hypoxia/reoxygenation injury in the presence of high glucose. Hypoxia/reoxygenation augmented HDAC6 activity and TNF-α levels and decreased mCI activity. These negative effects were blocked by HDAC6 knockdown. CONCLUSION HDAC6 is an essential negative regulator of MIRI in diabetes. Genetic deletion or pharmacologic inhibition of HDAC6 protects the heart from MIRI by limiting TNF-α-induced mitochondrial injury in experimental diabetes.
Collapse
MESH Headings
- Animals
- Myocardial Reperfusion Injury/enzymology
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/genetics
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/drug effects
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/drug therapy
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Histone Deacetylase 6/metabolism
- Histone Deacetylase 6/antagonists & inhibitors
- Histone Deacetylase 6/genetics
- Histone Deacetylase Inhibitors/pharmacology
- Mice, Knockout
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Mice, Inbred C57BL
- Hydroxamic Acids/pharmacology
- Mitochondrial Dynamics/drug effects
- Male
- Electron Transport Complex I/metabolism
- Electron Transport Complex I/genetics
- Isolated Heart Preparation
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/pathology
- Signal Transduction
- Mice
- Myocardial Infarction/enzymology
- Myocardial Infarction/pathology
- Myocardial Infarction/metabolism
- Myocardial Infarction/prevention & control
- Myocardial Infarction/genetics
- Myocardial Infarction/physiopathology
- Ventricular Function, Left/drug effects
- Indoles
Collapse
Affiliation(s)
- Shelley L Baumgardt
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
| | - Xuebin Fu
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Yanan Liu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, The People’s Republic of China
| | - Ming Zhao
- The Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, IL 60611, USA
| | - Ling Chen
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Rachana Mishra
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Muthukumar Gunasekaran
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Progyaparamita Saha
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Joseph M Forbess
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Zeljko J Bosnjak
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
| | - Judy R Kersten
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
| | - Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, IL 60611, USA
| | - Sunjay Kaushal
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Zhi-Dong Ge
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53206, USA
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, 303 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, IL 60611, USA
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, IL 60611, USA
| |
Collapse
|
6
|
Napiórkowska-Baran K, Doligalska A, Drozd M, Czarnowska M, Łaszczych D, Dolina M, Szymczak B, Schmidt O, Bartuzi Z. Management of a Patient with Cardiovascular Disease Should Include Assessment of Primary and Secondary Immunodeficiencies: Part 2-Secondary Immunodeficiencies. Healthcare (Basel) 2024; 12:1977. [PMID: 39408157 PMCID: PMC11477378 DOI: 10.3390/healthcare12191977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Cardiovascular diseases are among the most common chronic diseases, generating high social and economic costs. Secondary immunodeficiencies occur more often than primary ones and may result from the co-occurrence of specific diseases, treatment, nutrient deficiencies and non-nutritive bio-active compounds that result from the industrial nutrient practices. OBJECTIVES The aim of this article is to present selected secondary immunodeficiencies and their impact on the cardiovascular system. RESULTS The treatment of a patient with cardiovascular disease should include an assess-ment for immunodeficiencies, because the immune and cardiovascular systems are closely linked. CONCLUSIONS Immune system dysfunctions can significantly affect the course of cardiovascular diseases and their treatment. For this reason, comprehensive care for a patient with cardiovascular disease requires taking into account potential immunodeficiencies, which can have a significant impact on the patient's health.
Collapse
Affiliation(s)
- Katarzyna Napiórkowska-Baran
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| | - Agata Doligalska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Magdalena Drozd
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marta Czarnowska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Dariusz Łaszczych
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marcin Dolina
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Bartłomiej Szymczak
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Oskar Schmidt
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Zbigniew Bartuzi
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| |
Collapse
|
7
|
Salah A, Bouzid F, Dhouib W, Benmarzoug R, Triki N, Rebai A, Kharrat N. Integrative Bioinformatics Approaches to Uncover Hub Genes and Pathways Involved in Cardiovascular Diseases. Cell Biochem Biophys 2024; 82:2107-2127. [PMID: 38809349 DOI: 10.1007/s12013-024-01319-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Cardiovascular diseases (CVD) represent a significant global health challenge resulting from a complex interplay of genetic, environmental, and lifestyle factors. However, the molecular pathways and genetic factors involved in the onset and progression of CVDs remain incompletely understood. Here, we performed an integrative bioinformatic analysis to highlight specific genes and signaling pathways implicated in the pathogenesis of 80 CVDs. Differentially expressed genes (DEGs) were identified through the integrated analysis of microarray and GWAS datasets. Then, hub genes were identified after gene ontology functional annotation analysis and protein-protein internet (PPI) analysis. In addition, pathways were identified through KEGG and gene ontology enrichment analyses. A total of 821 hub genes related to 80 CVDs were identified, including 135 common and frequent CVD-associated genes. TNF, IL6, VEGFA, and TGFB.1 genes were the central core genes expressed in 50% or more of CVDs, confirming that the inflammation is a key pathological feature of CVDs. Analysis of hub genes by KEGG enrichment revealed predominant enrichment in 201 KEGG pathways, of which the AGE-RAGE signaling pathway in diabetic complications was identified as the common key KEGG implicated in 62 CVDs. In addition, the outcomes showed an overrepresentation in pathways categorized under human diseases, particularly in the subcategories of infectious diseases and cancers, which may be common risk factors for CVDs. In conclusion, this powerful approach for in silico fine-mapping of genes and pathways allowed the identification of determinant hubs genes and pathways implicated in the pathogenesis of CVDs which could be employed in developing more targeted and effective interventions for preventing, diagnosing, and treating CVDs. The function of these hub genes in CVDs needs further exploration to elucidate their biological characteristics.
Collapse
Affiliation(s)
- Awatef Salah
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia.
| | - Fériel Bouzid
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Wala Dhouib
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Riadh Benmarzoug
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Nesrine Triki
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ahmed Rebai
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Najla Kharrat
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
8
|
Shah M, Arumugam S. Exploring putative drug properties associated with TNF-alpha inhibition and identification of potential targets in cardiovascular disease using machine learning-assisted QSAR modeling and virtual reverse pharmacology approach. Mol Divers 2024; 28:2263-2287. [PMID: 38954070 DOI: 10.1007/s11030-024-10921-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Cardiovascular disease is a chronic inflammatory disease with high mortality rates. TNF-alpha is pro-inflammatory and associated with the disease, but current medications have adverse effects. Therefore, efficient inhibitors are urgently needed as alternatives. This study represents a structural-activity relationship investigation of TNF-alpha, curated from the ChEMBL database. Exploratory data analysis was performed to visualize the physicochemical properties of different bioactivity groups. The extracted molecules were subjected to PubChem and SubStructure fingerprints, and a QSAR-based Random Forest (QSAR-RF) model was generated using the WEKA tool. The QSAR random Forest model was built based on the SubStructure fingerprint with a correlation coefficient of 0.992 and 0.716 as the respective tenfold cross-validation scores. The variance important plot (VIP) method was used to extract the important features for TNF-alpha inhibition. The Substructure-based QSAR-RF (SS-QSAR-RF) model was validated using molecules from PubChem and ZINC databases. The generated model also predicts the pIC50 value of the molecules selected from the docking study followed by molecular dynamic simulation with the time step of 100 ns. Through virtual reverse pharmacology, we determined the main drug targets from the top four hit compounds obtained via molecular docking study. Our analysis included an integrated bioinformatics approach to pinpoint crucial targets like EGRF, HSP900A1, STAT3, PSEN1, AKT1, and MDM2. Further, GO and KEGG pathways analysis identified relevant cardiovascular disease-related pathways for the hub gene involved. However, this study provides valuable insights, it is important to note that it lacks experimental application. Future research may benefit from conducting in-vitro and in-vivo studies.
Collapse
Affiliation(s)
- Manisha Shah
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Sivakumar Arumugam
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
9
|
Padial-Molina M, Gonzalez-Perez G, Martin-Morales N, Sanchez-Fernandez E, O'Valle F, Galindo-Moreno P. Periostin in the relation between periodontal disease and atherosclerotic coronary artery disease: A pilot randomized clinical study. J Periodontal Res 2024; 59:446-457. [PMID: 38140743 DOI: 10.1111/jre.13229] [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: 09/12/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVE The aim of this study was to analyze the effects of periodontal treatment on markers of atherosclerotic coronary artery disease and circulating levels of periostin. BACKGROUND Periostin is necessary for periodontal stability, but it is highly present in atherosclerotic plaques. Treatment of periodontal disease, with low levels of local periostin, is thought to reduce systemic levels of periostin. Thus, this may contribute to cardiovascular health. METHODS A pilot randomized controlled clinical trial was designed to include patients with severe periodontal disease and history of atherosclerotic coronary artery disease. Samples of gingival crevicular fluid (GCF) and serum were collected before and after periodontal treatment by periodontal surgery or non-surgical therapy. The levels of several markers of inflammation and cardiovascular damage were evaluated including CRP, IFN-γ, IL-1ß, IL-10, MIP-1α, periostin, and TNF-α in GCF and CRP, Fibrinogen, IFN-γ, IL-1ß, IL-6, IL-10, L-Selectin, MIP-1α, Periostin, TNF-α, and vWF in serum. RESULTS A total of 22 patients with an average of 56 years old were recruited for participating in this study. Twenty of them were male. Most of them (82%) had suffered an acute myocardial event and underwent surgery for placing 1, 2, or 3 stents in the coronary arteries more than 6 months ago but less than 1 year. The treatment of periodontal disease resulted in an overall improvement of all periodontal parameters. Regarding the evaluation of GCF and serum, a significant increase of periostin in the GCF was observed after periodontal surgery. In contrast, although other markers in GCF and serum improved, no significant correlations were found. CONCLUSION Treatment of periodontal disease through periodontal surgery induces a local and transient increase in the levels of periostin in the gingival crevicular fluid. The effects on systemic markers of inflammation and cardiovascular function have not been confirmed.
Collapse
Affiliation(s)
- Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Gloria Gonzalez-Perez
- PhD Program in Clinical Medicine and Public Health, University of Granada, Granada, Spain
| | - Natividad Martin-Morales
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- PhD Program in Biomedicine, University of Granada, Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, Granada, Spain
| | - Elena Sanchez-Fernandez
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Francisco O'Valle
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, Granada, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| |
Collapse
|
10
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
11
|
Aldokhayyil M, Gomez DH, Cook MD, Kavazis AN, Roberts MD, Geetha T, Brown MD. Influence of Race and High Laminar Shear Stress on TNFR1 Signaling in Endothelial Cells. Int J Mol Sci 2023; 24:14723. [PMID: 37834170 PMCID: PMC10572906 DOI: 10.3390/ijms241914723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Tumor necrosis factor (TNF) binding to endothelial TNF receptor-I (TNFR-I) facilitates monocyte recruitment and chronic inflammation, leading to the development of atherosclerosis. In vitro data show a heightened inflammatory response and atherogenic potential in endothelial cells (ECs) from African American (AA) donors. High laminar shear stress (HSS) can mitigate some aspects of racial differences in endothelial function at the cellular level. We examined possible racial differences in TNF-induced monocyte adhesion and TNFR1 signaling complex expression/activity, along with the effects of HSS. Tohoku Hospital Pediatrics-1 (THP-1) monocytes were used in a co-culture system with human umbilical vein ECs (HUVECs) from Caucasian American (CA) and AA donors to examine racial differences in monocyte adhesion. An in vitro exercise mimetic model was applied to investigate the potential modulatory effect of HSS. THP-1 adherence to ECs and TNF-induced nuclear factor kappa B (NF-κB) DNA binding were elevated in AA ECs compared to CA ECs, but not significantly. We report no significant racial differences in the expression of the TNFR-I signaling complex. Application of HSS significantly increased the expression and shedding of TNFR-I and the expression of TRAF3, and decreased the expression of TRAF5 in both groups. Our data does not support TNF-induced NF-κB activation as a potential mediator of racial disparity in this model. Other pathways and associated factors activated by the TNFR1 signaling complex are recommended targets for future research.
Collapse
Affiliation(s)
- Maitha Aldokhayyil
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
- Department of Physical Therapy, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Dulce H. Gomez
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
| | - Marc D. Cook
- Department of Kinesiology, North Carolina Agriculture and Technology State University, Greensboro, NC 27411, USA
| | | | | | - Thangiah Geetha
- Department of Nutritional Sciences, College of Human Sciences, Auburn University, Auburn, AL 36849, USA
| | | |
Collapse
|
12
|
Bansal N, Pasricha C, Kumari P, Jangra S, Kuar R, Singh R. A comprehensive overview of juvenile idiopathic arthritis: From pathophysiology to management. Autoimmun Rev 2023; 22:103337. [PMID: 37068698 DOI: 10.1016/j.autrev.2023.103337] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
Rheumatoid Arthritis (RA) is a progressive autoimmune disease. It is among the most widespread chronic illnesses in children, with an annual incidence of 1.6 to 23 new instances per 100,000 adolescents. About 1 child in every 1000 develops Juvenile Idiopathic Arthritis (JIA) type of chronic arthritis. The cause of JIA is not well known but what known is that it involves inflammation of the synovium and destruction of tissues in joints which can cause early-onset of oligo articular JIA. It is challenging to diagnose the condition in some children who initially complain of pain and joint swelling as there is no blood test discovered that can confirm the diagnoses of JIA. As JIA patients are immunosuppressed due to the use of drugs, making them vulnerable to catch infections like COVID 19 which can lead to cardiovascular diseases having high rate of morbidity and mortality. The comorbidity like Diabetes has higher incidence in these patients resulting in synergistic effect on inflammation. Currently, the connection of genetics in JIA provides evidence that HLA Class I and II alleles have a role in the pathophysiology of various subtypes of JIA which includes inflammation in the axial skeletal. The primary objective of therapy in juvenile idiopathic arthritis is the suppression of clinical symptoms. The pharmacological approach includes use of medications like DMARDs, NSAIDs etc. and non-pharmacological approach includes physiotherapy, which helps in restoring normal joint function and herbs as adjuvants which has the benefit of no side effects.
Collapse
Affiliation(s)
- Nancy Bansal
- Chitkara College of Pharmacy, Chitakara University, Punjab, India
| | - Chirag Pasricha
- Chitkara College of Pharmacy, Chitakara University, Punjab, India
| | - Pratima Kumari
- Chitkara College of Pharmacy, Chitakara University, Punjab, India
| | - Sarita Jangra
- Chitkara College of Pharmacy, Chitakara University, Punjab, India
| | - Rupinder Kuar
- Chitkara College of Pharmacy, Chitakara University, Punjab, India
| | - Ravinder Singh
- Chitkara College of Pharmacy, Chitakara University, Punjab, India.
| |
Collapse
|
13
|
Matsiras D, Bezati S, Ventoulis I, Verras C, Parissis J, Polyzogopoulou E. Gut Failure: A Review of the Pathophysiology and Therapeutic Potentials in the Gut-Heart Axis. J Clin Med 2023; 12:2567. [PMID: 37048650 PMCID: PMC10095379 DOI: 10.3390/jcm12072567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite considerable advances in the field, heart failure (HF) still poses a significant disease burden among affected individuals since it continues to cause high morbidity and mortality rates. Inflammation is considered to play a key role in disease progression, but the exact underlying pathophysiological mechanisms involved have not yet been fully elucidated. The gut, as a potential source of inflammation, could feasibly explain the state of low-grade inflammation seen in patients with chronic HF. Several derangements in the composition of the microbiota population, coupled with an imbalance between favorable and harmful metabolites and followed by gut barrier disruption and eventually bacterial translocation, could contribute to cardiac dysfunction and aggravate HF. On the other hand, HF-associated congestion and hypoperfusion alters intestinal function, thereby creating a vicious cycle. Based on this evidence, novel pharmaceutical agents have been developed and their potential therapeutic use has been tested in both animal and human subjects. The ultimate goal in these efforts is to reverse the aforementioned intestinal derangements and block the inflammation cascade. This review summarizes the gut-related causative pathways implicated in HF pathophysiology, as well as the associated therapeutic interventions described in the literature.
Collapse
Affiliation(s)
- Dionysis Matsiras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christos Verras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
14
|
Bezati S, Velliou M, Ventoulis I, Simitsis P, Parissis J, Polyzogopoulou E. Infection as an under-recognized precipitant of acute heart failure: prognostic and therapeutic implications. Heart Fail Rev 2023:10.1007/s10741-023-10303-8. [PMID: 36897491 PMCID: PMC9999079 DOI: 10.1007/s10741-023-10303-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 03/11/2023]
Abstract
As the prevalence of heart failure (HF) continues to rise, prompt diagnosis and management of various medical conditions, which may lead to HF exacerbation and result in poor patient outcomes, are of paramount importance. Infection has been identified as a common, though under-recognized, precipitating factor of acute heart failure (AHF), which can cause rapid development or deterioration of HF signs and symptoms. Available evidence indicates that infection-related hospitalizations of patients with AHF are associated with higher mortality, protracted length of stay, and increased readmission rates. Understanding the intricate interaction of both clinical entities may provide further therapeutic strategies to prevent the occurrence of cardiac complications and improve prognosis of patients with AHF triggered by infection. The purpose of this review is to investigate the incidence of infection as a causative factor in AHF, explore its prognostic implications, elucidate the underlying pathophysiological mechanisms, and highlight the basic principles of the initial diagnostic and therapeutic interventions in the emergency department.
Collapse
Affiliation(s)
- Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, Chaidari, 12462, Athens, Greece.
| | - Maria Velliou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, Chaidari, 12462, Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, Keptse Area, Ptolemaida, 50200, Greece
| | - Panagiotis Simitsis
- National and Kapodistrian University of Athens, 2nd Department of Cardiology, Heart Failure Unit, Attikon University Hospital, Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, Chaidari, 12462, Athens, Greece.,Emergency Medicine Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, Chaidari, 12462, Athens, Greece.,Emergency Medicine Department, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Baumgardt SL, Fang J, Fu X, Liu Y, Xia Z, Zhao M, Chen L, Mishra R, Gunasekaran M, Saha P, Forbess JM, Bosnjak ZJ, Camara AKS, Kersten JR, Thorp E, Kaushal S, Ge ZD. Augmentation of Histone Deacetylase 6 Activity Impairs Mitochondrial Respiratory Complex I in Ischemic/Reperfused Diabetic Hearts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.21.529462. [PMID: 36865233 PMCID: PMC9980088 DOI: 10.1101/2023.02.21.529462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
BACKGROUND Diabetes augments activity of histone deacetylase 6 (HDAC6) and generation of tumor necrosis factor α (TNFα) and impairs the physiological function of mitochondrial complex I (mCI) which oxidizes reduced nicotinamide adenine dinucleotide (NADH) to nicotinamide adenine dinucleotide to sustain the tricarboxylic acid cycle and β-oxidation. Here we examined how HDAC6 regulates TNFα production, mCI activity, mitochondrial morphology and NADH levels, and cardiac function in ischemic/reperfused diabetic hearts. METHODS HDAC6 knockout, streptozotocin-induced type 1 diabetic, and obese type 2 diabetic db/db mice underwent myocardial ischemia/reperfusion injury in vivo or ex vivo in a Langendorff-perfused system. H9c2 cardiomyocytes with and without HDAC6 knockdown were subjected to hypoxia/reoxygenation injury in the presence of high glucose. We compared the activities of HDAC6 and mCI, TNFα and mitochondrial NADH levels, mitochondrial morphology, myocardial infarct size, and cardiac function between groups. RESULTS Myocardial ischemia/reperfusion injury and diabetes synergistically augmented myocardial HDCA6 activity, myocardial TNFα levels, and mitochondrial fission and inhibited mCI activity. Interestingly, neutralization of TNFα with an anti-TNFα monoclonal antibody augmented myocardial mCI activity. Importantly, genetic disruption or inhibition of HDAC6 with tubastatin A decreased TNFα levels, mitochondrial fission, and myocardial mitochondrial NADH levels in ischemic/reperfused diabetic mice, concomitant with augmented mCI activity, decreased infarct size, and ameliorated cardiac dysfunction. In H9c2 cardiomyocytes cultured in high glucose, hypoxia/reoxygenation augmented HDAC6 activity and TNFα levels and decreased mCI activity. These negative effects were blocked by HDAC6 knockdown. CONCLUSIONS Augmenting HDAC6 activity inhibits mCI activity by increasing TNFα levels in ischemic/reperfused diabetic hearts. The HDAC6 inhibitor, tubastatin A, has high therapeutic potential for acute myocardial infarction in diabetes.
Collapse
Affiliation(s)
- Shelley L. Baumgardt
- Departments of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53206
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53206
| | - Xuebin Fu
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
| | - Yanan Liu
- Departments of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53206
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, The People’s Republic of China
| | - Ming Zhao
- The Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, Illinois 60611
| | - Ling Chen
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
| | - Rachana Mishra
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
| | - Muthukumar Gunasekaran
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
| | - Progyaparamita Saha
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
| | - Joseph M. Forbess
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
| | - Zeljko J. Bosnjak
- Departments of Medicine and Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53206
| | - Amadou KS Camara
- Departments of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53206
| | - Judy R. Kersten
- Departments of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53206
| | - Edward Thorp
- Departments of Pathology and Pediatrics, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, Illinois 60611
| | - Sunjay Kaushal
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
| | - Zhi-Dong Ge
- Departments of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53206
- Cardiovascular-Thoracic Surgery and the Heart Center, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Departments of Pediatrics and Surgery, Feinberg School of Medicine, Northwestern University, 225 E. Chicago Avenue, Chicago, Illinois 60611
- Departments of Pathology and Pediatrics, Feinberg School of Medicine, Northwestern University, 300 E. Superior Avenue, Chicago, Illinois 60611
| |
Collapse
|
16
|
Yang Y, Ma Y, Yu S, Lin Z, Yan C, Wang Y, Yuan Q, Meng Z, Yan G, Wu Z, Tang H, Peng Z, Huang J, Zhuang G. TIPE2 knockout reduces myocardial cell damage by inhibiting IFN-γ-mediated ferroptosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166566. [PMID: 36216021 DOI: 10.1016/j.bbadis.2022.166566] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/18/2022] [Accepted: 10/04/2022] [Indexed: 11/19/2022]
Abstract
Acute rejection of the transplanted heart is mediated by oxidative programmed cell death through the synergistic effects of the innate and adaptive immune systems. However, the role of ferroptosis, a newly discovered form of oxidative cell death, has not been widely evaluated. Tumor necrosis factor-α-induced protein-8 like 2 (TNFAIP8L2), also known as TIPE2, is required for maintaining immune homeostasis. To characterize the role of TIPE2 in mediating heart allografts, BALB/c hearts were transplanted into C57BL/6 wild-type (WT) and TIPE2-/- recipient mice. In TIPE2-/- recipient mice, allograft injury in BALB/c allograft hearts was significantly reduced through the inhibition of allograft ferroptosis. On day 3 and day 6 post-transplantation, the numbers of CD3+, CD4+, and CD8+ cells among splenocytes and draining lymph node cells were significantly decreased, and the activation of CD4+ and CD8+ cells in grafts was decreased in TIPE2-/- recipient mice compared with WT mice. Moreover, CD4+ and CD8+ T cells in TIPE2-/- recipient mice were characterized by deficient capacities for interferon-γ (IFN-γ) production through the TBK1 signaling axis and increased glutathione peroxidase 4 (GPX4). In cell experiments, treatment with IFN-γ enhanced ferroptosis-specific lipid peroxidation in myocardial cells and correlated inversely with GPX4 expression. Mechanistically, IFN-γ administration decreased the expression of GPX4 by inhibiting MEK/ERK phosphorylation. In summary, our findings demonstrated that TIPE2 deficiency inhibits T-cell production of IFN-γ to reduce ferroptosis in allografts by restraining lipid peroxidation.
Collapse
Affiliation(s)
- Yan Yang
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen, China
| | - Yunhan Ma
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen, China
| | - Shengnan Yu
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen, China
| | - Zeyang Lin
- Department of Pathology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changxiu Yan
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen, China
| | - Yinan Wang
- Department of Basic Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qian Yuan
- Department of Basic Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhe Meng
- Department of Basic Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China; Department of Obstetrics and Gynecology, Dongfang Affiliated Hospital of Xiamen University, Fuzhou, China
| | - Guoliang Yan
- Department of Basic Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhengxin Wu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Huamei Tang
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen, China; Department of General Surgery, Xiang An Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhihai Peng
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen, China; Department of General Surgery, Xiang An Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Jiyi Huang
- The Fifth Hospital of Xiamen, Xiamen, Fujian, China; The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Guohong Zhuang
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
17
|
Doukbi E, Soghomonian A, Sengenès C, Ahmed S, Ancel P, Dutour A, Gaborit B. Browning Epicardial Adipose Tissue: Friend or Foe? Cells 2022; 11:991. [PMID: 35326442 PMCID: PMC8947372 DOI: 10.3390/cells11060991] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
The epicardial adipose tissue (EAT) is the visceral fat depot of the heart which is highly plastic and in direct contact with myocardium and coronary arteries. Because of its singular proximity with the myocardium, the adipokines and pro-inflammatory molecules secreted by this tissue may directly affect the metabolism of the heart and coronary arteries. Its accumulation, measured by recent new non-invasive imaging modalities, has been prospectively associated with the onset and progression of coronary artery disease (CAD) and atrial fibrillation in humans. Recent studies have shown that EAT exhibits beige fat-like features, and express uncoupling protein 1 (UCP-1) at both mRNA and protein levels. However, this thermogenic potential could be lost with age, obesity and CAD. Here we provide an overview of the physiological and pathophysiological relevance of EAT and further discuss whether its thermogenic properties may serve as a target for obesity therapeutic management with a specific focus on the role of immune cells in this beiging phenomenon.
Collapse
Affiliation(s)
- Elisa Doukbi
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Astrid Soghomonian
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| | - Coralie Sengenès
- Stromalab, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, University of Toulouse, F-31100 Toulouse, France;
- Institut National de la Santé et de la Recherche Médicale, University Paul Sabatier, F-31100 Toulouse, France
| | - Shaista Ahmed
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Patricia Ancel
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Anne Dutour
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| | - Bénédicte Gaborit
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| |
Collapse
|
18
|
|
19
|
Mirna M, Holnthoner M, Topf A, Jirak P, Fejzic D, Paar V, Kellermair J, Blessberger H, Reiter C, Kammler J, Motloch LJ, Jung C, Kretzschmar D, Franz M, Alushi B, Lauten A, Hoppe UC, Steinwender C, Lichtenauer M. Tumor necrosis factor alpha-an underestimated risk predictor in patients undergoing transcatheter aortic valve replacement (TAVR)? J Clin Lab Anal 2021; 35:e23977. [PMID: 34562276 PMCID: PMC8605157 DOI: 10.1002/jcla.23977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/06/2021] [Accepted: 08/14/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Systemic inflammation has been identified as a major cardiovascular risk factor in patients undergoing transcatheter aortic valve replacement (TAVR), yet currently, it is not adequately portrayed in scores for pre-interventional risk assessment. The aim of this study was to investigate the predictive ability of TNF-α in TAVR. METHODS A total of 431 patients undergoing transfemoral TAVR were enrolled in this study. Blood samples were drawn prior to intervention, 24 h post-intervention, 4, 5, and 7 days post-intervention, and 1, 3, and 6 months post-TAVR. RESULTS In a univariate Cox proportional hazard analysis, plasma concentrations of TNF-α after 24 h and after 5 days were associated with mortality after 12 months (after 24 h: HR 1.002 (1.000-1.004), p = 0.028; after 5d: HR 1.003 (1.001-1.005), p = 0.013). This association remained significant even after correction for confounders in a multivariate Cox regression analysis. Additionally, cut-offs were calculated. Patients above the cut-off for TNF-α after 5d had a significantly worse 12-month mortality than patients below the cut-off (18.8% vs. 2.8%, p = 0.046). CONCLUSION Plasma levels of TNF-α after 24 h and 5 days were independently associated with 12-month mortality in patients undergoing TAVR. Thus, TNF-α could represent a novel biomarker for enhanced risk stratification in these patients.
Collapse
Affiliation(s)
- Moritz Mirna
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Mario Holnthoner
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Albert Topf
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Peter Jirak
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Dzeneta Fejzic
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Vera Paar
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Jörg Kellermair
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Hermann Blessberger
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Christian Reiter
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Jürgen Kammler
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Paracelsus Medical University of Salzburg, Linz, Austria
| | - Lukas J Motloch
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Dusseldorf, Germany
| | - Daniel Kretzschmar
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Marcus Franz
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Brunilda Alushi
- Department of General and Interventional Cardiology and Rhythmology, Helios Clinic, Erfurt, Germany
| | - Alexander Lauten
- Department of General and Interventional Cardiology and Rhythmology, Helios Clinic, Erfurt, Germany
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Clemens Steinwender
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Paracelsus Medical University of Salzburg, Linz, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
20
|
Phloretin Alleviates Arsenic Trioxide-Induced Apoptosis of H9c2 Cardiomyoblasts via Downregulation in Ca 2+/Calcineurin/NFATc Pathway and Inflammatory Cytokine Release. Cardiovasc Toxicol 2021; 21:642-654. [PMID: 34037972 DOI: 10.1007/s12012-021-09655-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/27/2021] [Indexed: 01/25/2023]
Abstract
Arsenic trioxide (ATO) is among the first-line chemotherapeutic drugs for treating acute promyelocytic leukemia patients, but its clinical use is hampered due to cardiotoxicity. The present investigation unveils the mechanism underlying ATO-induced oxidative stress that promotes calcineurin (a ubiquitous Ca2+/calmodulin-dependent serine/threonine phosphatase expressed only during sustained Ca2+ elevation) expression, inflammatory cytokine release and apoptosis in H9c2 cardiomyoblasts, and its possible modulation with phloretin (PHL, an antioxidant polyphenol present in apple peel). ATO caused Ca2+ overload resulting in elevated expression of calcineurin and its downstream transcriptional effector NFATc causing the release of cytokines such as IL-2, IL-6, MCP-1, IFN-γ, and TNF-α in H9c2 cardiomyoblast. There was a visible increase in the nuclear fraction of NF-κB and ROS-mediated apoptotic cell death. The expression levels of cardiac-specific genes (troponin, desmin, and caveolin-3) and genes of the apoptotic signaling pathway (BCL-2, BAX, IGF1, AKT, ERK1, -2, RAF1, and JNK) in response to ATO and PHL were studied. The putative binding mode and the potential ligand-target interactions of PHL with calcineurin using docking software (Autodock and iGEMDOCKv2) showed the high binding affinity of PHL to calcineurin. PHL co-treatment significantly reduced Ca2+ influx and normalized the expression of calcineurin, NFATc, NF-κB, and other cytokines. PHL co-treatment resulted in activation of BCL-2, IGF1, AKT, RAF1, ERK1, and ERK2 and inhibition of BAX and JNK. Overall, these results revealed that PHL has a protective effect against ATO-induced apoptosis and we propose calcineurin as a druggable target for the interaction of PHL in ATO cardiotoxicity in H9c2 cells.
Collapse
|
21
|
Poore TS, Taylor-Cousar JL, Zemanick ET. Cardiovascular complications in cystic fibrosis: A review of the literature. J Cyst Fibros 2021; 21:18-25. [PMID: 34140249 DOI: 10.1016/j.jcf.2021.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/30/2021] [Accepted: 04/26/2021] [Indexed: 12/26/2022]
Abstract
Cystic fibrosis is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, leading to dysfunction of the CFTR protein. CFTR dysfunction leads to disease in the respiratory and gastrointestinal systems. Disorders of the cardiovascular system in individuals with CF are usually attributed to secondary effects from progressive lung disease. However, CFTR has been localized to vascular endothelium and smooth muscle, suggesting that CFTR dysfunction may directly impact cardiovascular function. As treatments for CF improve and life-expectancy increases, the risk of vascular disease may increase in prevalence related to primary and secondary CFTR dysfunction, chronic systemic inflammation, nutritional health and hyperglycemia in individuals with CF related diabetes. Here we review the available literature on CF and the cardiovascular system, examining the secondary effects and evidence for direct CFTR dysfunction in the heart, aorta, pulmonary vessels, and vasculature, as well as future directions and treatment options.
Collapse
Affiliation(s)
- T Spencer Poore
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Edith T Zemanick
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
22
|
El Moheb M, Jia Z, Qin H, El Hechi MW, Nordestgaard AT, Lee JM, Han K, Kaafarani HMA. The Obesity Paradox in Elderly Patients Undergoing Emergency Surgery: A Nationwide Analysis. J Surg Res 2021; 265:195-203. [PMID: 33951584 DOI: 10.1016/j.jss.2021.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/15/2021] [Accepted: 02/27/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Obesity has long been considered a risk factor for postoperative adverse events in surgery. We sought to study the impact of body mass index (BMI) on the clinical outcomes of the high-risk emergency general surgery (EGS) elderly patients. METHODS All EGS ≥65 years old patients in the 2007-2016 ACS-NSQIP database, identified using the variables 'emergency' and 'surgspec,' were included. Patients were classified into five groups: normal weight: BMI <25 kg/m2, overweight: BMI ≥25 kg/m2 and <30 kg/m2, Class I: BMI ≥30 kg/m2 and <35 kg/m2, Class II: BMI ≥35 kg/m2 and <40 kg/m2, and Class III: BMI ≥40 kg/m2. Patients with BMI<18.5 kg/m2 were excluded. Multivariable logistic regression models were built to assess the relationship between obesity and 30-day postoperative mortality, overall morbidity, and individual postoperative complications after adjusting for demographics (e.g., age, gender), comorbidities (e.g., diabetes mellitus, heart failure), laboratory tests (e.g., white blood cell count, albumin), and operative complexity (e.g., ASA classification). RESULTS A total of 78,704 patients were included, of which 26,011 were overweight (33.1%), 13,897 (17.6%) had Class I obesity, 5904 (7.5%) had Class II obesity, and 4490 (5.7%) had Class III obesity. On multivariable analyses, compared to the nonobese, patients who are overweight or with Class I-III obesity paradoxically had a lower risk of mortality, bleeding requiring transfusion, pneumonia, stroke and myocardial infarction (MI). Additionally, the incidence of MI and stroke decreased in a stepwise fashion as BMI progressed from overweight to severely obese (MI: OR: 0.84 [0.73-0.95], OR: 0.73 [0.62-0.86], OR: 0.66 [0.52-0.83], OR: 0.51 [0.38-0.68]; stroke: OR: 0.80 [0.65-0.99], OR: 0.79 [0.62-1.02], OR: 0.71 [0.50-1.00], OR: 0.43 [0.28-0.68]). CONCLUSION In our study of elderly EGS patients, overweight and obese patients had a lower risk of mortality, bleeding requiring transfusion, pneumonia, reintubation, stroke, and MI. Further studies are needed to confirm and investigate the obesity paradox in this patient population.
Collapse
Affiliation(s)
- Mohamad El Moheb
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Boston, Massachusetts
| | - Zhenyi Jia
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Boston, Massachusetts; Department of General Surgery, Shanghai Sixth People's hospital affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Huanlong Qin
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Majed W El Hechi
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Boston, Massachusetts
| | - Ask T Nordestgaard
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Boston, Massachusetts; Department of Anesthesia, Centre of Head and Orthopaedics 4231, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jae Moo Lee
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Boston, Massachusetts
| | - Kelsey Han
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Boston, Massachusetts
| | - Haytham M A Kaafarani
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
23
|
Kotru S, Klimuntowski M, Ridha H, Uddin Z, Askhar AA, Singh G, Howlader MMR. Electrochemical sensing: A prognostic tool in the fight against COVID-19. Trends Analyt Chem 2021; 136:116198. [PMID: 33518850 PMCID: PMC7825925 DOI: 10.1016/j.trac.2021.116198] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The COVID-19 pandemic has devastated the world, despite all efforts in infection control and treatment/vaccine development. Hospitals are currently overcrowded, with health statuses of patients often being hard to gauge. Therefore, methods for determining infection severity need to be developed so that high-risk patients can be prioritized, resources can be efficiently distributed, and fatalities can be prevented. Electrochemical prognostic biosensing of various biomarkers may hold promise in solving these problems as they are low-cost and provide timely results. Therefore, we have reviewed the literature and extracted the most promising biomarkers along with their most favourable electrochemical sensors. The biomarkers discussed in this paper are C-reactive protein (CRP), interleukins (ILs), tumour necrosis factor alpha (TNFα), interferons (IFNs), glutamate, breath pH, lymphocytes, platelets, neutrophils and D-dimer. Metabolic syndrome is also discussed as comorbidity for COVID-19 patients, as it increases infection severity and raises chances of becoming infected. Cannabinoids, especially cannabidiol (CBD), are discussed as a potential adjunct therapy for COVID-19 as their medicinal properties may be desirable in minimizing the neurodegenerative or severe inflammatory damage caused by severe COVID-19 infection. Currently, hospitals are struggling to provide adequate care; thus, point-of-care electrochemical sensor development needs to be prioritized to provide an approximate prognosis for hospital patients. During and following the immediate aftermath of the pandemic, electrochemical sensors can also be integrated into wearable and portable devices to help patients monitor recovery while returning to their daily lives. Beyond the COVID-19 pandemic, these sensors will also prove useful for monitoring inflammation-based diseases such as cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Sharda Kotru
- Department of Integrated Biomedical Engineering and Health Sciences, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Martin Klimuntowski
- Department of Electrical and Computer Engineering, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Hashim Ridha
- School of Interdisciplinary Science, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Zakir Uddin
- School of Rehabilitation Science, McMaster University, 1400 Main St W, Hamilton, ON, L8S 1C7, Canada
| | - Ali A Askhar
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Matiar M R Howlader
- Department of Electrical and Computer Engineering, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| |
Collapse
|
24
|
Raffaele S, Lombardi M, Verderio C, Fumagalli M. TNF Production and Release from Microglia via Extracellular Vesicles: Impact on Brain Functions. Cells 2020; 9:cells9102145. [PMID: 32977412 PMCID: PMC7598215 DOI: 10.3390/cells9102145] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor (TNF) is a pleiotropic cytokine powerfully influencing diverse processes of the central nervous system (CNS) under both physiological and pathological conditions. Here, we analyze current literature describing the molecular processes involved in TNF synthesis and release from microglia, the resident immune cells of the CNS and the main source of this cytokine both in brain development and neurodegenerative diseases. A special attention has been given to the unconventional vesicular pathway of TNF, based on the emerging role of microglia-derived extracellular vesicles (EVs) in the propagation of inflammatory signals and in mediating cell-to-cell communication. Moreover, we describe the contribution of microglial TNF in regulating important CNS functions, including the neuroinflammatory response following brain injury, the neuronal circuit formation and synaptic plasticity, and the processes of myelin damage and repair. Specifically, the available data on the functions mediated by microglial EVs carrying TNF have been scrutinized to gain insights on possible novel therapeutic strategies targeting TNF to foster CNS repair.
Collapse
Affiliation(s)
- Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Marta Lombardi
- CNR Institute of Neuroscience, 20129 Milan, Italy; (M.L.); (C.V.)
| | - Claudia Verderio
- CNR Institute of Neuroscience, 20129 Milan, Italy; (M.L.); (C.V.)
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
- Correspondence: ; Tel.: +39-0250318307
| |
Collapse
|
25
|
Vasilieva E, Gianella S, Freeman ML. Novel Strategies to Combat CMV-Related Cardiovascular Disease. Pathog Immun 2020; 5:240-274. [PMID: 33089035 PMCID: PMC7556413 DOI: 10.20411/pai.v5i1.382] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cytomegalovirus (CMV), a ubiquitous human pathogen that is never cleared from the host, has long been thought to be relatively innocuous in immunocompetent adults, but causes severe complications including blindness, end-organ disease, and death in newborns and in immuno-compromised individuals, such as organ transplant recipients and those suffering from AIDS. Yet even in persons with intact immunity, CMV infection is associated with profound stimulation of immune and inflammatory pathways. Carriers of CMV infection also have an elevated risk of developing cardiovascular complications. In this review, we define the proposed mechanisms of how CMV contributes to cardiovascular disease (CVD), describe current approaches to target CMV, and discuss how these strategies may or may not alleviate cardiovascular complications in those with CMV infection. In addition, we discuss the special situation of CMV coinfection in people with HIV infection receiving antiretroviral therapy, and describe how these 2 viral infections may interact to potentiate CVD in this especially vulnerable population.
Collapse
Affiliation(s)
- Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine; Department of Medicine; Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
26
|
Arsenaki E, Georgakopoulos P, Mitropoulou P, Koutli E, Thomas K, Charakida M, Georgiopoulos G. Cardiovascular Disease in Juvenile Idiopathic Arthritis. Curr Vasc Pharmacol 2020; 18:580-591. [DOI: 10.2174/1570161118666200408121307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 01/21/2023]
Abstract
Juvenile idiopathic arthritis (JIA), is a term used to describe a group of inflammatory disorders
beginning before the age of 16 years. Although for the majority of children remission is achieved
early, those with systemic or polyarticular form of the disease may present persistent symptoms in
adulthood. Considering that there is overlap in the pathogenesis of JIA with adult rheumatic diseases,
concerns have been raised as to whether JIA patients could be at increased cardiovascular (CV) risk in
the long-term. In this review, we summarize evidence for CV involvement in JIA and present data on
CV risk factors and surrogate markers of arterial disease. We also provide information on beneficial and
harmful CV effects of anti-inflammatory medications in the context of JIA and suggest strategies for
CV screening. Overall, patients with systemic forms of JIA demonstrate an adverse lipid profile and
early arterial changes relevant to accelerated arterial disease progression. Although there is paucity of
data on CV outcomes, we recommend a holistic approach in the management of JIA patients, which
includes CV risk factor monitoring and lifestyle modification as well as use, when necessary, of antiinflammatory
therapies with documented CV safety.
Collapse
Affiliation(s)
| | - Panagiotis Georgakopoulos
- National Department of Anatomy, School of Medicine, Faculty of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Evangelia Koutli
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, United Kingdom
| | - Konstantinos Thomas
- Joint Rheumatology Program, Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Marietta Charakida
- School of Biomedical Engineering and Imaging Sciences, King’s College, London, United Kingdom
| | - Georgios Georgiopoulos
- School of Biomedical Engineering and Imaging Sciences, King’s College, London, United Kingdom
| |
Collapse
|
27
|
Skytte MJ, Samkani A, Astrup A, Larsen TM, Frystyk J, Poulsen HE, Henriksen T, Holst JJ, Andersen O, Madsbad S, Haugaard SB, Krarup T, Larsen EL. Effects of a highly controlled carbohydrate-reduced high-protein diet on markers of oxidatively generated nucleic acid modifications and inflammation in weight stable participants with type 2 diabetes; a randomized controlled trial. Scand J Clin Lab Invest 2020; 80:401-407. [PMID: 32374188 DOI: 10.1080/00365513.2020.1759137] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/20/2020] [Accepted: 04/19/2020] [Indexed: 01/04/2023]
Abstract
Carbohydrate-restricted diets are increasingly recognized as options for dietary management of type 2 diabetes mellitus (T2DM). We investigated the effects of a carbohydrate-reduced high-protein (CRHP) and a conventional diabetes (CD) diet on oxidative stress and inflammation in weight stable individuals with T2DM. We hypothesized that the CRHP diet would improve markers of oxidatively generated RNA and DNA modifications as well as inflammatory parameters. Thirty participants with T2DM were randomized to 6 weeks of CRHP or CD dietary treatment (30/50 energy percentage (E%) carbohydrate, 30/17E% protein, 40/33E% fat), followed by a cross-over to the opposite diet for a subsequent 6-week period. All meals were provided during the study and body weight was controlled. Diurnal urine samples were collected after 4 weeks on each diet and oxidatively generated RNA and DNA modifications were measured as 8-oxo-7,8-dihydroguanosine (8-oxoGuo) and 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), respectively. Fasting concentrations of soluble urokinase plasminogen activator receptor, high-sensitivity C-reactive protein, tumor necrosis factor alpha and interleukin-6 were measured before and after 6 weeks of interventions. Compared with the CD diet, the CRHP diet increased 24-hour urinary excretion of 8-oxoGuo by 9.3% (38.6 ± 12.6 vs. 35.3 ± 11.0 nmol/24 h, p = .03), whereas 8-oxodG did not differ between diets (24.0 ± 9.5 vs. 24.8 ± 11.1 nmol/24 h, p = .17). Changes in plasma inflammatory parameters did not differ between CRHP and CD diets, all p ≥ .2. The clinical implications of increased RNA oxidation following a CRHP diet as well as long-term effects of carbohydrate-restriction on markers of oxidatively generated nucleic acid modifications should be a field of future study.
Collapse
Affiliation(s)
- Mads Juul Skytte
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Amirsalar Samkani
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Meinert Larsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jan Frystyk
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henrik Enghusen Poulsen
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Trine Henriksen
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ove Andersen
- Clinical Research Centre and the Emergency Department, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Steen Bendix Haugaard
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Thure Krarup
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Emil List Larsen
- Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| |
Collapse
|
28
|
Tarasiuk E, Bonda TA, Dziemidowicz M, Winnicka MM, Bernaczyk P, Kamiński KA. The effect of interleukin 6 deficiency on myocardial signal transduction pathways activation induced by bacterial lipopolysaccharide in young and old mice. Adv Med Sci 2020; 65:386-393. [PMID: 32693349 DOI: 10.1016/j.advms.2020.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/13/2020] [Accepted: 06/20/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE Exaggerated release of proinflammatory mediators during sepsis contributes to inadequate vasodilatation and depressed myocardial contractility, which lead to development of shock and circulatory collapse. The aim of the study was to evaluate the effect of IL-6 and aging on activation of intracellular signaling pathways in the myocardium induced by bacterial lipopolysaccharide (LPS) administration. MATERIAL/METHODS LPS was injected intraperitoneally to male 3- and 24-month old mice with systemic IL-6 gene knock-out (IL-6KO) and the reference strain (WT). LPS was given intraperitoneally in single low (0.1 mg/kg) or high (10 mg/kg) dose, or in two doses (0.1 + 10 mg/kg) with 24-h delay. The expression and phosphorylation of STAT3, ERK1/2, Akt1/2/3 proteins in the left ventricular myocardium was evaluated after 24 h using Western blotting. RESULTS Low LPS dose induced higher STAT3 phosphorylation only in old IL-6KO mice, not affecting ERK1/2 and Akt1/2/3 phosphorylation in any group. High LPS dose upregulated STAT3 phosphorylation similarly in all groups, reduced ERK1/2 expression in young WT mice and upregulated Akt1/2/3 expression and phosphorylation in young IL-6KO mice. Pretreatment with low LPS dose attenuated phosphorylation of STAT3 in both old groups and phosphorylation of Akt1/2/3 in young IL-6KO group. Two-dose approach also significantly potentiated ERK1/2 phosphorylation in both old groups. CONCLUSIONS Obtained results show that IL-6 deficiency alters the activity of intracellular signaling pathways: JAK/STAT in old and Akt in young LPS-treated mice. This may indicate that lack of IL-6 attenuates Akt-related cytoprotective effect of pretreatment with low LPS dose in young but not in aged animals.
Collapse
Affiliation(s)
- Ewa Tarasiuk
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| | - Tomasz A Bonda
- Department of General and Experimental Pathology, Medical University of Bialystok, Bialystok, Poland
| | - Magdalena Dziemidowicz
- Department of General and Experimental Pathology, Medical University of Bialystok, Bialystok, Poland
| | - Maria M Winnicka
- Department of General and Experimental Pathology, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Bernaczyk
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Karol A Kamiński
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland; Department of Population Medicine and Civilization Diseases Prevention, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
29
|
Millett C, Harder J, Locascio J, Shanahan M, Santone G, Fichorova R, Corrigan A, Baecher-Allan C, Burdick K. TNF-α and its soluble receptors mediate the relationship between prior severe mood episodes and cognitive dysfunction in euthymic bipolar disorder. Brain Behav Immun 2020; 88:403-410. [PMID: 32272224 PMCID: PMC8577222 DOI: 10.1016/j.bbi.2020.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Bipolar disorder (BD) is one of the most disabling mental health conditions in the world. Symptoms of cognitive impairment in BD contribute directly to occupational and social deficiencies and are very difficult to treat. Converging evidence suggests that BD patients have increased peripheral markers of inflammation. The hypothesis of neuroprogression in BD postulates that cognitive deficits develop over the course of the illness and are influenced by prior severe mood episodes, leading to wear-and-tear on the brain- however, there exists a paucity of data statistically testing a mediating role of immune molecules in cognitive dysfunction in BD. METHODS This is a cross-sectional study. We measured serum levels of tumor necrosis factor alpha (TNF-α), and soluble (s) TNF receptors one and two (sTNF-R1 and sTNF-R2) in 219 euthymic BD patients and 52 Healthy Controls (HCs). Structural equation modeling (SEM) was used for the primary purpose of assessing whether TNF markers (measured by the multiple indicators TNF-α, sTNF-R1 and sTNF-R2) mediate the effect or number of prior severe mood episodes (number of prior psychiatric hospitalizations) on cognitive performance. RESULTS BD and HC groups did not differ on circulating levels of TNF molecules in the present study. However, we found higher sTNF-R1 concentration in 'late-stage' BD illness (>1 prior psychiatric hospitalization) compared to those in early stage illness. In the subsequent SEM, we found that the model fits the data acceptably (Chi-square = 49.2, p = 0.3), and had a 'close fit' (RMSEA = 0.02, PCLOSE = 0.9). Holding covariates constant (age, sex, premorbid IQ, education, and race), we found that the standardized indirect effect was significant, p = 0.015, 90%CI [-0.07, -0.01], indicating that the estimated model was consistent with peripheral TNF markers partially mediating a causal effect of severe mood episodes on executive function. CONCLUSIONS Our results indicate that circulating levels of TNF molecules partially mediate the relationship between prior severe mood episodes and executive function in BD. These results may implicate TNF variables in the neuroprogressive course of BD and could point to novel interventions for cognition.
Collapse
Affiliation(s)
- C.E. Millett
- Mood and Psychosis Research Program, Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA,Department of Psychiatry, Harvard Medical School, Boston, MA
| | - J. Harder
- Mood and Psychosis Research Program, Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA,Department of Psychiatry, Harvard Medical School, Boston, MA
| | - J.J. Locascio
- Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital and Harvard Medical School
| | - M. Shanahan
- Mood and Psychosis Research Program, Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA,Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital and Harvard Medical School
| | - G. Santone
- Laboratory of Genital Tract Biology, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - R. Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - A. Corrigan
- Mood and Psychosis Research Program, Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA
| | - C. Baecher-Allan
- Ann Romney Center for Neurologic Disease, Harvard Medical School, Boston, MA 02115 USA,Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - K.E. Burdick
- Mood and Psychosis Research Program, Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA,Department of Psychiatry, Harvard Medical School, Boston, MA
| |
Collapse
|
30
|
Wu NC, Wang JJ. Ibudilast, a Phosphodiesterase Inhibitor and Toll-Like Receptor-4 Antagonist, Improves Hemorrhagic Shock and Reperfusion-Induced Left Ventricular Dysfunction by Reducing Myocardial Tumor Necrosis Factor α. Transplant Proc 2020; 52:1869-1874. [PMID: 32631581 DOI: 10.1016/j.transproceed.2020.02.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/09/2020] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Rapid loss of blood volume causes ischemic injury to myocardial cells and impairs cardiac function. Subsequent reperfusion, although necessary to revitalize stunned tissues, can induce production of reactive oxygen species and inflammation, causing further tissue damages and weakening cardiac function. Ibudilast, a Toll-like receptor-4 (TLR4) antagonist and an inhibitor of phosphodiesterase-4, possesses antioxidative and anti-inflammatory capacities. In this study, we aim to examine the protective efficacy of ibudilast against hemorrhagic shock and reperfusion (HSR)-induced myocardial injury and cardiac dysfunction. METHODS Studies were conducted on male Sprague-Dawley rats in 3 groups: sham-operated, HSR with, and HSR without pretreatment of ibudilast. Hemorrhagic shock was induced by withdrawing blood from the femoral artery until the mean aortic pressure dropped to around 40 mm Hg; reperfusion was conducted by replenishing blood after 120 minutes of hemorrhagic shock, and the observation continued for another 240 minutes. The left ventricular (LV) contractility, diastolic suction capacity, and ventricular stiffness were evaluated using simultaneous LV pressure, and volume was recorded during a temporary inferior vena cava constriction at the end of reperfusion. Ibudilast (10 mg/kg) was administered intraperitoneally 3 days and 20 minutes prior to HSR. Serum creatine kinase myocardial band (CK-MB) was examined at the end of both HSR, and serum CK-MB, myocardial TLR4 protein expression, and malondialdehyde (MDA) and tumor necrosis factor (TNF)-α levels at the end of reperfusion. RESULTS HSR induced an increase in serum CK-MB. Subsequent reperfusion further increased serum CK-MB, upregulated myocardial TLR4 protein expression, and increased tissue levels MDA and TNF-α vs the sham (P < .05). HSR reduced LV contractility, prolonged LV relaxation time, and increased LV diastolic stiffness. Ibudilast pretreatment attenuated HSR-induced TLR4 protein expression, reduced myocardial MDA and TNF-α levels, and protected against cardiac dysfunction. CONCLUSIONS Ibudilast pretreatment reduced myocardial TLR4 expression, decreased MDA and TNF-α levels, and protected against HSR-induced decrease in LV contractility, prolonged LV relaxation time, and increased diastolic stiffness.
Collapse
Affiliation(s)
- Nan-Chun Wu
- Division of Cardiovascular Surgery, Chi-Mei Foundation Hospital, Tainan, Taiwan; Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Jiun-Jr Wang
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
31
|
Sasserath T, Rumsey JW, McAleer CW, Bridges LR, Long CJ, Elbrecht D, Schuler F, Roth A, Bertinetti‐LaPatki C, Shuler ML, Hickman JJ. Differential Monocyte Actuation in a Three-Organ Functional Innate Immune System-on-a-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000323. [PMID: 32670763 PMCID: PMC7341107 DOI: 10.1002/advs.202000323] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/09/2020] [Indexed: 05/24/2023]
Abstract
A functional, human, multiorgan, pumpless, immune system-on-a-chip featuring recirculating THP-1 immune cells with cardiomyocytes, skeletal muscle, and liver in separate compartments in a serum-free medium is developed. This in vitro platform can emulate both a targeted immune response to tissue-specific damage, and holistic proinflammatory immune response to proinflammatory compound exposure. The targeted response features fluorescently labeled THP-1 monocytes selectively infiltrating into an amiodarone-damaged cardiac module and changes in contractile force measurements without immune-activated damage to the other organ modules. In contrast to the targeted immune response, general proinflammatory treatment of immune human-on-a-chip systems with lipopolysaccharide (LPS) and interferon-γ (IFN-γ) causes nonselective damage to cells in all three-organ compartments. Biomarker analysis indicates upregulation of the proinflammation cytokines TNF-α, IL-6, IL-10, MIP-1, MCP-1, and RANTES in response to LPS + IFN-γ treatment indicative of the M1 macrophage phenotype, whereas amiodarone treatment only leads to an increase in the restorative cytokine IL-6 which is a marker for the M2 phenotype. This system can be used as an alternative to humanized animal models to determine direct immunological effects of biological therapeutics including monoclonal antibodies, vaccines, and gene therapies, and the indirect effects caused by cytokine release from target tissues in response to a drug's pharmacokinetics (PK)/pharmacodynamics (PD) profile.
Collapse
Affiliation(s)
- Trevor Sasserath
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | - John W. Rumsey
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | | | | | | | - Daniel Elbrecht
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | - Franz Schuler
- Hoffmann‐La RochePharmaceuticals DivisionBldg 73, Rm 117bBasel4070Switzerland
| | - Adrian Roth
- Hoffmann‐La RochePharmaceuticals DivisionBldg 73, Rm 117bBasel4070Switzerland
| | | | | | - James J. Hickman
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
- NanoScience Technology Center, University of Central Florida12424 Research Parkway, Suite 400OrlandoFL32826USA
| |
Collapse
|
32
|
Saji N, Francis N, Schwarz LJ, Blanchard CL, Santhakumar AB. The Antioxidant and Anti-Inflammatory Properties of Rice Bran Phenolic Extracts. Foods 2020; 9:foods9060829. [PMID: 32599964 PMCID: PMC7353521 DOI: 10.3390/foods9060829] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022] Open
Abstract
Oxidative stress and inflammation are known to be linked to the development of chronic inflammatory conditions, such as type 2 diabetes and cardiovascular disease. Dietary polyphenols have been demonstrated to contain potent bioactivity against specific inflammatory pathways. Rice bran (RB), a by-product generated during the rice milling process, is normally used in animal feed or discarded due to its rancidity. However, RB is known to be abundant in bioactive polyphenols including phenolic acids. This study investigates the antioxidant and anti-inflammatory effects of RB phenolic extracts (25, 50, 100, and 250 µg/mL) on RAW264.7 mouse macrophage cells stimulated with hydrogen peroxide and lipopolysaccharide. Biomarkers of oxidative stress and inflammation such as malondialdehyde (MDA), intracellular reactive oxygen species, nitric oxide and pro-inflammatory cytokines such as interleukin-6 (IL-6), monocyte chemoattractant protein 1 (MCP-1), interleukin-10 (IL-10), tumor necrosis factor-α (TNF-α), interleukin-12, p70 (IL-12p70), and interferon-γ (IFN-γ) were measured in vitro. Treatment with RB extracts significantly decreased the production of MDA, intracellular reactive oxygen species, nitric oxide and pro-inflammatory cytokines (IL-6, IL-12p70, and IFN-γ) when compared to the control. It is proposed that RB phenolic extracts, via their metal chelating properties and free radical scavenging activity, target pathways of oxidative stress and inflammation resulting in the alleviation of vascular inflammatory mediators.
Collapse
Affiliation(s)
- Nancy Saji
- Australian Research Council (ARC) Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; (N.S.); (N.F.); (L.J.S.); (C.L.B.)
- School of Biomedical Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia
| | - Nidhish Francis
- Australian Research Council (ARC) Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; (N.S.); (N.F.); (L.J.S.); (C.L.B.)
- School of Animal and Veterinary Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia
| | - Lachlan J. Schwarz
- Australian Research Council (ARC) Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; (N.S.); (N.F.); (L.J.S.); (C.L.B.)
- School of Agricultural and Wine Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia
| | - Christopher L. Blanchard
- Australian Research Council (ARC) Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; (N.S.); (N.F.); (L.J.S.); (C.L.B.)
- School of Biomedical Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia
| | - Abishek B. Santhakumar
- Australian Research Council (ARC) Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia; (N.S.); (N.F.); (L.J.S.); (C.L.B.)
- School of Biomedical Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia
- Correspondence: ; Tel.: +61-2-6933-2678
| |
Collapse
|
33
|
Tyerman Z, Mehaffey JH, Hawkins RB, Dahl J, Narahari A, Chancellor WZ, Yount K, Yarboro LT, Teman NR, Ailawadi G. History of Serious Mental Illness Is a Predictor of Morbidity and Mortality in Cardiac Surgery. Ann Thorac Surg 2020; 111:109-116. [PMID: 32544450 DOI: 10.1016/j.athoracsur.2020.04.118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 03/18/2020] [Accepted: 04/24/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Serious mental illness (SMI), defined as a mental disorder causing functional impairment, affects 9.8 million Americans. SMI correlates with earlier onset, more extensive cardiac disease, and reduced life expectancy by 25 years. The impact of SMI on patients undergoing cardiac surgery has not been extensively studied. We hypothesized that patients with SMI have worse cardiac surgery outcomes. METHODS Using our institution's Society of Thoracic Surgeons database of 16,781 cardiac operations (2002-2017), a total of 1445 (8.7%) patients with SMI were identified and stratified into anxiety, mood disorders, and psychosis. The risk-adjusted impact on morbidity and mortality were evaluated using multivariable regression. RESULTS Patients with SMI were more often female patients, were younger, and had more comorbid disease. SMI patients were more likely to have had previous cardiac surgery and require urgent or emergent procedures (both P < .05). Among specific SMI diagnoses, patients with psychosis had worse outcomes compared with the general population, with higher operative mortality (9.1% vs 4.2%; P = .001), major morbidity (30.4% vs 15.8%; P < .0001), and cost ($50,211 vs $38,820; P < .001). After multivariable risk adjustment, SMI and psychosis remained independently associated with composite mortality and major morbidity (odds ratio, 1.21; P = .012; and odds ratio, 1.68; P = .003, respectively). CONCLUSIONS SMI is independently associated with morbidity and mortality after cardiac surgery. SMI patients, especially the subset with psychosis, are complicated, high-risk, and resource-consuming. Refined strategies to reduce postoperative complications and improve care coordination are necessary in this population.
Collapse
Affiliation(s)
- Zachary Tyerman
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - J Hunter Mehaffey
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Robert B Hawkins
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Jolian Dahl
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Adishesh Narahari
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - William Z Chancellor
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Kenan Yount
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Leora T Yarboro
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Nicholas R Teman
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Gorav Ailawadi
- Division of Thoracic and Cardiovascular Surgery, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
34
|
Imani MM, Sadeghi M, Khazaie H, Emami M, Sadeghi Bahmani D, Brand S. Serum and Plasma Tumor Necrosis Factor Alpha Levels in Individuals with Obstructive Sleep Apnea Syndrome: A Meta-Analysis and Meta-Regression. Life (Basel) 2020; 10:E87. [PMID: 32545460 PMCID: PMC7345342 DOI: 10.3390/life10060087] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Obstructive sleep apnea syndrome (OSAS) is associated with a variety of inflammatory factors. Specifically, proinflammatory cytokines appear to be associated with the pathogenesis of OSAS. METHODS For the present meta-analysis and meta-regression on serum and plasma tumor necrosis factor alpha (TNF-α) levels in individuals with and without OSAS, we performed a systematic search without any restrictions of the PubMed/Medline, Scopus, Cochrane Library, and Web of Science databases to find relevant articles published up to February 1, 2020. RESULTS Fifty-five (adults: 29 studies on serum and 17 studies on plasma; children: 4 studies on serum and 5 studies on plasma) were included and analyzed. Always compared to age-matched healthy controls, the pooled MDs were as follows: adults, serum: 10.22 pg/mL (95% CI = 8.86, 11.58; p < 0.00001); adults, plasma: 5.90 pg/mL (95% CI = 4.00, 7.80; p < 0.00001); children, serum: 0.21 pg/mL (95% CI = 0.05, 0.37; p = 0.01); children, plasma: 5.90 pg/mL (95% CI = 4.00, 7.80; p < 0.00001). CONCLUSIONS Compared to healthy and age-matched controls, adult individuals with OSAS had significantly higher serum/plasma TNF-α levels. For children with OSAS, significantly higher levels were observed for TNF-α in serum but not in plasma.
Collapse
Affiliation(s)
- Mohammad Moslem Imani
- Department of Orthodontics, Kermanshah University of Medical Sciences, 6713954658 Kermanshah, Iran;
| | - Masoud Sadeghi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, 6714415185 Kermanshah, Iran;
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, 6719851115 Kermanshah, Iran; (H.K.); (D.S.B.)
| | - Mehrdad Emami
- Students Research Committee, Kermanshah University of Medical Sciences, 6715847141 Kermanshah, Iran;
| | - Dena Sadeghi Bahmani
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, 6719851115 Kermanshah, Iran; (H.K.); (D.S.B.)
- Center for Affective, Stress and Sleep Disorders, University of Basel, Psychiatric Clinics, 4001 Basel, Switzerland
- Substance Abuse Prevention Research Center, Kermanshah University of Medical Sciences, 6715847141 Kermanshah, Iran
- Departments of Physical Therapy, University of Alabama at Birmingham, Birmingham, AL 35209, USA
| | - Serge Brand
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, 6719851115 Kermanshah, Iran; (H.K.); (D.S.B.)
- Center for Affective, Stress and Sleep Disorders, University of Basel, Psychiatric Clinics, 4001 Basel, Switzerland
- Substance Abuse Prevention Research Center, Kermanshah University of Medical Sciences, 6715847141 Kermanshah, Iran
- Department of Sport, Exercise and Health, Division of Sport Science and Psychosocial Health, University of Basel, 4052 Basel, Switzerland
- School of Medicine, Tehran University of Medical Sciences, 1416753955 Tehran, Iran
| |
Collapse
|
35
|
Maione AS, Pilato CA, Casella M, Gasperetti A, Stadiotti I, Pompilio G, Sommariva E. Fibrosis in Arrhythmogenic Cardiomyopathy: The Phantom Thread in the Fibro-Adipose Tissue. Front Physiol 2020; 11:279. [PMID: 32317983 PMCID: PMC7147329 DOI: 10.3389/fphys.2020.00279] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/12/2020] [Indexed: 12/22/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart disorder, predisposing to malignant ventricular arrhythmias leading to sudden cardiac death, particularly in young and athletic patients. Pathological features include a progressive loss of myocardium with fibrous or fibro-fatty substitution. During the last few decades, different clinical aspects of ACM have been well investigated but still little is known about the molecular mechanisms that underlie ACM pathogenesis, leading to these phenotypes. In about 50% of ACM patients, a genetic mutation, predominantly in genes that encode for desmosomal proteins, has been identified. However, the mutation-associated mechanisms, causing the observed cardiac phenotype are not always clear. Until now, the attention has been principally focused on the study of molecular mechanisms that lead to a prominent myocardium adipose substitution, an uncommon marker for a cardiac disease, thus often recognized as hallmark of ACM. Nonetheless, based on Task Force Criteria for the diagnosis of ACM, cardiomyocytes death associated with fibrous replacement of the ventricular free wall must be considered the main tissue feature in ACM patients. For this reason, it urges to investigate ACM cardiac fibrosis. In this review, we give an overview on the cellular effectors, possible triggers, and molecular mechanisms that could be responsible for the ventricular fibrotic remodeling in ACM patients.
Collapse
Affiliation(s)
- Angela Serena Maione
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Chiara Assunta Pilato
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michela Casella
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Alessio Gasperetti
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milan, Italy
- University Heart Center, Zurich University Hospital, Zurich, Switzerland
| | - Ilaria Stadiotti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| |
Collapse
|
36
|
Cytokine and Cancer Biomarkers Detection: The Dawn of Electrochemical Paper-Based Biosensor. SENSORS 2020; 20:s20071854. [PMID: 32230808 PMCID: PMC7180619 DOI: 10.3390/s20071854] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
Although the established ELISA-based sensing platforms have many benefits, the importance of cytokine and cancer biomarkers detection for point-of-care diagnostics has propelled the search for more specific, sensitive, simple, accessible, yet economical sensor. Paper-based biosensor holds promise for future in-situ applications and can provide rapid analysis and data without the need to conduct in a laboratory. Electrochemical detection plays a vital role in interpreting results obtained from qualitative assessment to quantitative determination. In this review, various factors affecting the design of an electrochemical paper-based biosensor are highlighted and discussed in depth. Different detection methods, along with the latest development in utilizing them in cytokine and cancer biomarkers detection, are reviewed. Lastly, the fabrication of portable electrochemical paper-based biosensor is ideal in deliberating positive societal implications in developing countries with limited resources and accessibility to healthcare services.
Collapse
|
37
|
Abstract
Alpha-1 antitrypsin (AAT) protects the lung by inhibiting neutrophil proteinases, but AAT has many other non-proteolytic functions that are anti-inflammatory, antiviral and homeostatic. Approximately 1 in 1600 to 1 in 5000 people have the homozygous Z mutation, which causes AAT misfolding, accumulation in (predominantly) liver cells and low circulating levels of AAT, leading to AAT deficiency (AATD). AATD is classically a disease of neutrophilic inflammation, with an aggressive and damaging innate immune response contributing to emphysema and other pathologies. AATD is one of the most common genetic disorders but considerably under-recognised. Most patients are diagnosed later in life, by which time they may have accumulated significant lung, liver and multisystem damage. Disease presentation is heterogeneous and not fully explained by deficiency levels alone or exposure to cigarette smoking. This suggests other factors influence AATD-associated pathological processes. Aging itself is associated with organ dysfunction, including emphysema and airflow obstruction, inflammation, altered immune cell responses (termed immunosenescence) and a loss of proteostasis. Many of these processes are present in AATD but at an earlier age and more advanced stage compared with chronological aging alone. Augmentation therapy does not completely abrogate the manifold disease processes present in AATD. New approaches are needed. There is emerging evidence that both age- and AATD-related disease processes are amenable to correction by targeting proteostasis, autophagy, immunosenescence and epigenetic factors. This review explores the impact of the aging process on AATD presentation and discusses novel therapeutic strategies to mitigate low levels of AAT or misfolded AAT in an aging host.
Collapse
|
38
|
Toczylowski K, Hirnle T, Harasiuk D, Zabielski P, Lewczuk A, Dmitruk I, Ksiazek M, Sulik A, Gorski J, Chabowski A, Baranowski M. Plasma concentration and expression of adipokines in epicardial and subcutaneous adipose tissue are associated with impaired left ventricular filling pattern. J Transl Med 2019; 17:310. [PMID: 31533725 PMCID: PMC6751580 DOI: 10.1186/s12967-019-2060-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/06/2019] [Indexed: 12/11/2022] Open
Abstract
Background Adipokines in serum derive mainly from subcutaneous and visceral adipose tissues. Epicardial adipose tissue (EAT), being a relatively small but unique fat depot, probably does not make an important contribution to systemic concentrations of adipokines. However, proximity of EAT to cardiac muscle and coronary arteries allows cells and proteins to penetrate between tissues. It is hypothesized that overexpression of proinflammatory cytokines in EAT plays an important role in pathophysiology of the heart. The aim of the study was to analyze the relationship between echocardiographic heart parameters and adipokines in plasma, epicardial, and subcutaneous fat in patients with obesity and type 2 diabetes mellitus (T2DM). Additionally, we evaluate proinflammatory properties of EAT by comparing that depot with subcutaneous adipose tissue. Methods The study included 55 male individuals diagnosed with coronary artery disease (CAD) who underwent planned coronary artery bypass graft. Plasma concentrations of leptin, adiponectin, resistin, visfatin, apelin, IL-6, and TNF-α, as well as their mRNA and protein expressions in EAT and subcutaneous adipose tissue (SAT) were determined. Results Obesity and diabetes were associated with increased leptin and decreased adiponectin plasma levels, higher protein expression of leptin and IL-6 in SAT, and higher visfatin protein expression in EAT. Impaired left ventricular (LV) diastolic function was associated with increased plasma concentrations of leptin, resistin, IL-6, and adiponectin, as well as with increased expressions of resistin, apelin, and adiponectin in SAT, and leptin in EAT. Conclusions Obesity and T2DM in individuals with CAD have a limited effect on adipokines. Expression of adipokines in EAT and SAT is linked to certain heart parameters, however diastolic dysfunction of the LV is strongly associated with circulating adipokines.
Collapse
Affiliation(s)
- Kacper Toczylowski
- Department of Pediatric Infectious Diseases, Medical University of Bialystok, Waszyngtona 17, 15-274, Bialystok, Poland
| | - Tomasz Hirnle
- Department of Cardiac Surgery, Medical University of Bialystok, Sklodowskiej 24A, 15-276, Bialystok, Poland
| | - Dorota Harasiuk
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Piotr Zabielski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Anna Lewczuk
- Department of Cardiac Surgery, Medical University of Bialystok, Sklodowskiej 24A, 15-276, Bialystok, Poland
| | - Iwona Dmitruk
- Department of Cardiac Surgery, Medical University of Bialystok, Sklodowskiej 24A, 15-276, Bialystok, Poland
| | - Monika Ksiazek
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Artur Sulik
- Department of Pediatric Infectious Diseases, Medical University of Bialystok, Waszyngtona 17, 15-274, Bialystok, Poland
| | - Jan Gorski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Marcin Baranowski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland.
| |
Collapse
|
39
|
Abstract
Chemokines are small secreted proteins with chemoattractant properties that play a key role in inflammation. One such chemokine, Stromal cell-derived factor-1 (SDF-1) also known as CXCL12, and its receptor, CXCR4, are expressed and functional in cardiac myocytes. SDF-1 both stimulates and enhances the cellular signal which attracts potentially beneficial stem cells for tissue repair within the ischemic heart. Paradoxically however, this chemokine is known to act in concert with the inflammatory cytokines of the innate immune response which contributes to cellular injury through the recruitment of inflammatory cells during ischemia. In the present study, we have demonstrated that SDF-1 has dose dependent effects on freshly isolated cardiomyocytes. Using Tunnel and caspase 3-activation assays, we have demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations (pathological concentrations) induced apoptosis. Furthermore, ELISA data demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations upregulated TNF-α protein expression which directly correlated with subsequent apoptosis. There was a significant reduction in SDF-1 mediated apoptosis when TNF-α expression was neutralized which suggests that SDF-1 mediated apoptosis is TNF-α-dependent. The fact that certain stimuli are capable of driving cardiomyocytes into apoptosis indicates that these cells are susceptible to clinically relevant apoptotic triggers. Our findings suggest that the elevated SDF-1 levels seen in a variety of clinical conditions, including ischemic myocardial infarction, may either directly or indirectly contribute to cardiac cell death via a TNF-α mediated pathway. This highlights the importance of this receptor/ligand in regulating the cardiomyocyte response to stress conditions.
Collapse
|
40
|
Al-Sawalha NA, Al-Filali MS, Alzoubi KH, Khabour OF. Effect of Prenatal Waterpipe Tobacco Smoke Exposure on Cardiac Biomarkers in Adult Offspring Rats. J Cardiovasc Pharmacol Ther 2019; 24:567-574. [DOI: 10.1177/1074248419849424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: The prevalence of waterpipe tobacco smoke (WTS) consumption is increased among pregnant woman. Prenatal cigarette smoke exposure increased the risk of developing cardiovascular diseases in offspring. The current study examined the effect of prenatal WTS exposure on inflammatory profile, oxidative stress, and cardiac biomarkers in adult offspring rats. Methods: Female rats received WTS (2 hours per day) or fresh air 1 day prior to mating and throughout the pregnancy period. The body and heart masses were measured in male offspring rats. The level of oxidative stress biomarkers, nitrate, inflammatory mediators (interleukin 6 [IL-6], tumor necrosis factor alpha [TNF-α]), and gene expression of protein kinase C epsilon, angiotensin 2 receptor one, and transforming growth factor beta1 were measured in cardiac tissue homogenates of 13-week-old male offspring rats. Results: Prenatal WTS exposure reduced body weight and increased heart to body weight ratio ( P < .05). Prenatal WTS exposure did not affect oxidative stress biomarkers (superoxide dismutase, glutathione peroxidase, and thiobarbituric acid reactive substances) but significantly increased catalase activity and nitrate level ( P < .05) in cardiac tissue of adult male offspring rats. In addition, prenatal exposure to WTS did not affect cardiac level of TNF-α and IL-6 as well as the gene expression of different cardiac modulators in adult male offspring rats ( P > .05). Conclusion: Prenatal WTS exposure has detrimental consequences on adult offspring rats by increasing the ratio of heart to body mass, increasing the catalase activity and nitrate level in cardiac tissue of adult male offspring rats.
Collapse
Affiliation(s)
- Nour A. Al-Sawalha
- Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Maria S. Al-Filali
- Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H. Alzoubi
- Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F. Khabour
- Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
41
|
The cytokine balance during CABG surgery with and without cardiopulmonary bypass. EGYPTIAN JOURNAL OF ANAESTHESIA 2019. [DOI: 10.1016/j.egja.2010.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
42
|
Lutkemeyer DDS, Do Amaral MA, Ishida Assunção NH, Marques Tejada NF, Saraiva Camara NO. Obesidade: uma abordagem inflamatória e microbiana. HU REVISTA 2019. [DOI: 10.34019/1982-8047.2018.v44.13963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A obesidade está intimamente ligada ao estado inflamatório, sendo considerada uma patologia metabólica complexa. Dietas hipercalóricas alteram a composição da microbiota intestinal, sendo a mudança da proporção de bactérias dos filos Bacteroidetes e Firmicutes uma das consequências mais conhecidas. Essa mudança determina a produção de metabólitos específicos do sistema imune, induzindo estado inflamatório responsável pelo agravamento de uma série de doenças. A dieta hipercalórica representa um fator de risco para a obesidade e para o diabetes mellitus, doenças interligadas pelo conceito de lipotoxicidade, e o estado inflamatório também contribui para o aparecimento e para a progressão de doenças cardiovasculares. Com esse artigo, objetivamos estudar a obesidade pela perspectiva imunológica e microbiológica, abordando as consequências de dietas hipercalóricas sobre o estado inflamatório e a sobre a microbiota. Ademais, associar a mudança no microbioma a doenças prevalentes como o diabetes mellitus e as doenças cardiovasculares, apontando abordagens terapêuticas potenciais.
Collapse
|
43
|
Dyball D, Evans S, Boos CJ, Stevelink SAM, Fear NT. The association between PTSD and cardiovascular disease and its risk factors in male veterans of the Iraq/Afghanistan conflicts: a systematic review. Int Rev Psychiatry 2019; 31:34-48. [PMID: 31041877 DOI: 10.1080/09540261.2019.1580686] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Military personnel with Post-Traumatic Stress Disorder (PTSD) can experience high levels of mental and physical health comorbidity, potentially indicating a high level of functional impairment that can impact on both military readiness and later ill-health. There is strong evidence to implicate PTSD as a contributory factor to Cardiovascular Disease (CVD) among serving personnel and veterans. This systematic review focusses on the association between PTSD and cardiovascular disease/risk factors in male, military serving and ex-serving personnel who served in the Iraq/Afghanistan conflicts. PUBMED, MEDLINE, PILOTS, EMBASE, PSYCINFO, and PSYCARTICLES were searched using PRISMA guidelines. Three hundred and forty-three records were identified, of which 20 articles were selected. PTSD was positively associated with the development of CVD, specifically circulatory diseases, including hypertension. PTSD was also positively associated with the following risk factors: elevated heart rate, tobacco use, dyslipidaemia, and obesity. Conflicting data is presented regarding heart rate variability and inflammatory markers. Future studies would benefit from a standardized methodological approach to investigating PTSD and physical health manifestations. It is suggested that clinicians offer health advice for CVD at an earlier age for ex-/serving personnel with PTSD.
Collapse
Affiliation(s)
- Daniel Dyball
- a King's Centre for Military Health Research, Psychological Medicine , King's College London , London , UK.,b ADVANCE study, Academic Department of Military Rehabilitation , Defence Medical Rehabilitation Centre Stanford Hall , Loughborough , UK
| | - Sarah Evans
- a King's Centre for Military Health Research, Psychological Medicine , King's College London , London , UK.,b ADVANCE study, Academic Department of Military Rehabilitation , Defence Medical Rehabilitation Centre Stanford Hall , Loughborough , UK
| | - Christopher J Boos
- b ADVANCE study, Academic Department of Military Rehabilitation , Defence Medical Rehabilitation Centre Stanford Hall , Loughborough , UK.,c Department of Postgraduate Medical Education , Bournemouth University , Poole , UK
| | - Sharon A M Stevelink
- a King's Centre for Military Health Research, Psychological Medicine , King's College London , London , UK.,d Department of Psychological Medicine , King's College London , London , UK
| | - Nicola T Fear
- a King's Centre for Military Health Research, Psychological Medicine , King's College London , London , UK.,b ADVANCE study, Academic Department of Military Rehabilitation , Defence Medical Rehabilitation Centre Stanford Hall , Loughborough , UK.,e Academic Department for Military Mental Health , King's College London , London , UK
| |
Collapse
|
44
|
Lamelas P, Schwalm JD, Leong D, Jolly S, Mehta S, Bangdiwala S, Yusuf S. Varying Effects of Body Mass Index and Mortality in Different Risk Groups. Am J Cardiol 2018; 122:1155-1160. [PMID: 30107907 DOI: 10.1016/j.amjcard.2018.06.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 01/04/2023]
Abstract
It is unclear why high body mass index (BMI) is associated with a lower risk of death in patients with cardiovascular disease (CVD). We hypothesized that the impact of higher BMI varies by the patient's baseline risk of death. We analyzed data from 14 prospective studies conducted by us in 50 countries of 170,470 patients: 22.8% without CVD, 29.4% with stable CVD and 47.8% with acute coronary syndromes (ACS). Compared with overweight (BMI 25 to 29.9 kg/m2), normal weight (BMI 20 to 24.9 kg/m2), and underweight (BMI < 20 kg/m2) were associated with higher mortality in all cohorts. Compared with overweight, the risk of death among those with obesity (BMI ≥ 30 kg/m2) varied depending on the population studied: HR (95% confidence interval) in those without CVD, with stable CVD, and with ACS were 1.20 (1.01 to 1.45; p = 0.04), 1.08 (1.02 to 1.15; p = 0.01), and 1.01 (0.93 to 1.10; p = 0.72), respectively. The BMI associated with lowest mortality increased as cohort risk increased (no-CVD 27.2 kg/m2, stable CVD 28.1 kg/m2, and ACS 30.9 kg/m2; p <0.001). Within each cohort, the optimal BMI value was higher in the high-risk subgroup. In the ACS cohort, low-risk patients had an optimal BMI value of 29.1 kg/m2 while in the high-risk group higher BMI was associated with lower risk of death (interaction-p <0.001). In conclusion, between and within cohorts of people without CVD, stable CVD, and ACS, higher BMI was associated with lower risk of death in higher risk groups. The benefits of increased body fat (i.e., increased caloric reserve) may counteract the hazards in high-risk groups, suggesting an alternative explanation of the obesity paradox.
Collapse
Affiliation(s)
- Pablo Lamelas
- The Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada.
| | - J D Schwalm
- The Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Darryl Leong
- The Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Sanjit Jolly
- The Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Shamir Mehta
- The Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Shrikant Bangdiwala
- The Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Salim Yusuf
- The Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| |
Collapse
|
45
|
Léger T, Charrier A, Moreau C, Hininger-Favier I, Mourmoura E, Rigaudière JP, Pitois E, Bouvier D, Sapin V, Pereira B, Azarnoush K, Demaison L. Early sepsis does not stimulate reactive oxygen species production and does not reduce cardiac function despite an increased inflammation status. Physiol Rep 2018; 5:5/13/e13231. [PMID: 28684640 PMCID: PMC5506518 DOI: 10.14814/phy2.13231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/14/2022] Open
Abstract
If it is sustained for several days, sepsis can trigger severe abnormalities of cardiac function which leads to death in 50% of cases. This probably occurs through activation of toll-like receptor-9 by bacterial lipopolysaccharides and overproduction of proinflammatory cytokines such as TNF-α and IL-1β In contrast, early sepsis is characterized by the development of tachycardia. This study aimed at determining the early changes in the cardiac function during sepsis and at finding the mechanism responsible for the observed changes. Sixty male Wistar rats were randomly assigned to two groups, the first one being made septic by cecal ligation and puncture (sepsis group) and the second one being subjected to the same surgery without cecal ligation and puncture (sham-operated group). The cardiac function was assessed in vivo and ex vivo in standard conditions. Several parameters involved in the oxidative stress and inflammation were determined in the plasma and heart. As evidenced by the plasma level of TNF-α and gene expression of IL-1β and TNF-α in the heart, inflammation was developed in the sepsis group. The cardiac function was also slightly stimulated by sepsis in the in vivo and ex vivo situations. This was associated with unchanged levels of oxidative stress, but several parameters indicated a lower cardiac production of reactive oxygen species in the septic group. In conclusion, despite the development of inflammation, early sepsis did not increase reactive oxygen species production and did not reduce myocardial function. The depressant effect of TNF-α and IL-1β on the cardiac function is known to occur at very high concentrations. The influence of low- to moderate-grade inflammation on the myocardial mechanical behavior must thus be revisited.
Collapse
Affiliation(s)
- Thibault Léger
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Alice Charrier
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Clarisse Moreau
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Isabelle Hininger-Favier
- Laboratoire de Bioénergétique Fondamentale et Appliquée, INSERM U1055, Université Joseph Fourier, Grenoble, France
| | - Evangelia Mourmoura
- Laboratoire de Bioénergétique Fondamentale et Appliquée, INSERM U1055, Université Joseph Fourier, Grenoble, France
| | | | - Elodie Pitois
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| | - Damien Bouvier
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Vincent Sapin
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Bruno Pereira
- Department of Clinical Research and Innovation, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Kasra Azarnoush
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France.,Heart Surgery Department, G. Montpied Hospital, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Luc Demaison
- INRA, UMR 1019 Nutrition Humaine, Clermont-Ferrand Cedex 1, France
| |
Collapse
|
46
|
Obad A, Peeran A, Little JI, Haddad GE, Tarzami ST. Alcohol-Mediated Organ Damages: Heart and Brain. Front Pharmacol 2018; 9:81. [PMID: 29487525 PMCID: PMC5816804 DOI: 10.3389/fphar.2018.00081] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022] Open
Abstract
Alcohol is one of the most commonly abused substances in the United States. Chronic consumption of ethanol has been responsible for numerous chronic diseases and conditions globally. The underlying mechanism of liver injury has been studied in depth, however, far fewer studies have examined other organs especially the heart and the central nervous system (CNS). The authors conducted a narrative review on the relationship of alcohol with heart disease and dementia. With that in mind, a complex relationship between inflammation and cardiovascular disease and dementia has been long proposed but inflammatory biomarkers have gained more attention lately. In this review we examine some of the consequences of the altered cytokine regulation that occurs in alcoholics in organs other than the liver. The article reviews the potential role of inflammatory markers such as TNF-α in predicting dementia and/or cardiovascular disease. It was found that TNF-α could promote and accelerate local inflammation and damage through autocrine/paracrine mechanisms. Unraveling the mechanisms linking chronic alcohol consumption with proinflammatory cytokine production and subsequent inflammatory signaling pathways activation in the heart and CNS, is essential to improve our understanding of the disease and hopefully facilitate the development of new remedies.
Collapse
Affiliation(s)
| | | | | | | | - Sima T. Tarzami
- Department of Physiology and Biophysics, Howard University, Washington, DC, United States
| |
Collapse
|
47
|
Azhari Z, Ismail MD, Zuhdi ASM, Md Sari N, Zainal Abidin I, Wan Ahmad WA. Association between body mass index and outcomes after percutaneous coronary intervention in multiethnic South East Asian population: a retrospective analysis of the Malaysian National Cardiovascular Disease Database-Percutaneous Coronary Intervention (NCVD-PCI) registry. BMJ Open 2017; 7:e017794. [PMID: 29127228 PMCID: PMC5695449 DOI: 10.1136/bmjopen-2017-017794] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE To examine the relationship between body mass index (BMI) and outcomes after percutaneous coronary intervention (PCI) in a multiethnic South East Asian population. SETTING Fifteen participating cardiology centres contributed to the Malaysian National Cardiovascular Disease Database-Percutaneous Coronary Intervention (NCVD-PCI) registry. PARTICIPANTS 28 742 patients from the NCVD-PCI registry who had their first PCI between January 2007 and December 2014 were included. Those without their BMI recorded or BMI <11 kg/m2 or >70 kg/m2 were excluded. MAIN OUTCOME MEASURES In-hospital death, major adverse cardiovascular events (MACEs), vascular complications between different BMI groups were examined. Multivariable-adjusted HRs for 1-year mortality after PCI among the BMI groups were also calculated. RESULTS The patients were divided into four groups; underweight (BMI <18.5 kg/m2), normal BMI (BMI 18.5 to <23 kg/m2), overweight (BMI 23 to <27.5 kg/m2) and obese (BMI ≥27.5 kg/m2). Comparison of their baseline characteristics showed that the obese group was younger, had lower prevalence of smoking but higher prevalence of diabetes, hypertension and dyslipidemia. There was no difference found in terms of in-hospital death, MACE and vascular complications after PCI. Multivariable Cox proportional hazard regression analysis showed that compared with normal BMI group the underweight group had a non-significant difference (HR 1.02, p=0.952), while the overweight group had significantly lower risk of 1-year mortality (HR 0.71, p=0.005). The obese group also showed lower HR but this was non-significant (HR 0.78, p=0.056). CONCLUSIONS Using Asian-specific BMI cut-off points, the overweight group in our study population was independently associated with lower risk of 1-year mortality after PCI compared with the normal BMI group.
Collapse
Affiliation(s)
- Zaid Azhari
- Division of Cardiology, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Muhammad Dzafir Ismail
- Division of Cardiology, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Ahmad Syadi Mahmood Zuhdi
- Division of Cardiology, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Norashikin Md Sari
- Division of Cardiology, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Imran Zainal Abidin
- Division of Cardiology, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Wan Azman Wan Ahmad
- Division of Cardiology, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
48
|
Guo XF, Li JM, Tang J, Li D. Effects of resveratrol supplementation on risk factors of non-communicable diseases: A meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2017; 58:3016-3029. [PMID: 28933578 DOI: 10.1080/10408398.2017.1349076] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The results of randomized controlled trials (RCTs) investigating resveratrol supplementation on risk factors of non-communicable diseases (NCDs) have been inconsistent. The present meta-analysis aimed to quantitatively evaluate the effects of resveratrol intervention on risk factors of NCDs. PubMed and Scopus databases were searched up to June 2017. Weighted mean differences were calculated for net changes in risk factors of NCDs by using a random-effects model. Pre-specified subgroup and univariate meta-regression analyses were carried out to explore the sources of heterogeneity. Twenty-nine studies (30 treatment arms) with 1069 participants were identified. Resveratrol supplementation significantly reduced the concentrations of fasting glucose (-4.77 mg/dL; 95% CI: -9.33 to -0.21 mg/dL; P = 0.040), total cholesterol (TC) (-9.75 mg/dL; 95% CI: -17.04 to -2.46 mg/dL; P = 0.009), and C-reactive protein (CRP) (-0.81 mg/L; 95% CI: -1.42 to -0.21 mg/L; P = 0.009). Resveratrol intervention exerted significant reductions in systolic blood pressure (SBP) and diastolic blood pressure (DBP) in subjects with type 2 diabetes mellitus (T2DM). Subgroup analysis also showed that the trials with resveratrol intervention ≥3 months significantly reduced the low-density lipoprotein cholesterol (LDL-C), DBP, and glycated hemoglobin (HbA1c) values. The results did not support that resveratrol intervention had favorable effects in altering high-density lipoprotein cholesterol (HDL-C), triglyceride (TAG), and homeostasis model assessment of insulin resistance (HOMA-IR). The present study provides substantial evidence that resveratrol supplementation has favorable effects on several risk factors of NCDs.
Collapse
Affiliation(s)
- Xiao-Fei Guo
- a Department of Food Science and Nutrition , Zhejiang University , Hangzhou , China
| | - Jiao-Mei Li
- a Department of Food Science and Nutrition , Zhejiang University , Hangzhou , China
| | - Jun Tang
- a Department of Food Science and Nutrition , Zhejiang University , Hangzhou , China
| | - Duo Li
- a Department of Food Science and Nutrition , Zhejiang University , Hangzhou , China.,b Institute of Nutrition and Health , Qingdao University , Qingdao , China
| |
Collapse
|
49
|
Carnevali L, Montano N, Statello R, Sgoifo A. Rodent models of depression-cardiovascular comorbidity: Bridging the known to the new. Neurosci Biobehav Rev 2017; 76:144-153. [DOI: 10.1016/j.neubiorev.2016.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/27/2016] [Accepted: 11/09/2016] [Indexed: 12/22/2022]
|
50
|
Li X, Yuan H, Li Q, Sun Z, Liu J, Wu W. Effects of ERK1/2 signal pathway on cardiomyocyte during glucose lowering. J Recept Signal Transduct Res 2017; 37:365-369. [PMID: 28425329 DOI: 10.1080/10799893.2017.1286675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE This study investigated whether the extracellular signal-regulated kinase 1/2 (ERK1/2) signal pathway affects cardiomyocyte apoptosis and the expression of tumor necrosis factor (TNF-α) at different glucose-lowering rates. METHODS Cardiomyocytes of Wistar neonate rats were maintained in a medium supplemented with 25 mmol/L glucosamine for 72 h. Then the medium was changed to different concentrations of glucosamine, and all cells were divided into five groups. The survival rate of cardiomyocyte was measured using the Cell Counting Kit-8; cardiomyocyte apoptosis was measured using the flow cytometry instrument and laser confocal microscope; TNF-α was measured using the enzyme-linked immunosorbent assay; and ERK1/2 protein and phosphorylation were measured using the Western blot. Cardiomyocyte apoptosis and TNF-α were measured again after adding U0126. RESULTS As the glucose-lowering rate increased, the survival rate of cardiomyocytes increased in group B and decreased in groups C, D, and E. The TNF-α concentration increased in groups B, C, and D and decreased in group E. After 24 h, the apoptosis rate decreased in group B and increased in groups C, D, and E. The expression of p-ERK1/2 increased in groups B, D, and E, and was the lowest in group C. After adding U0126, the survival rate of cardiomyocyte in all groups increased and TNF-α concentration decreased. CONCLUSIONS The influence of glucose-lowering rate on cardiomyocyte apoptosis and TNF-α was caused by the p-ERK1/2 pathway. During the glucose-lowering course, the p-ERK1/2 pathway promoted cardiomyocyte apoptosis, and TNF-α secretion was related to not only osmotic pressure but also ERK1/2 signal pathway activation.
Collapse
Affiliation(s)
- Xin Li
- a Department of Endocrinology , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Hui Yuan
- b Department of Endocrinology , Daqing Oil Field General Hospital , Daqing , China
| | - Qian Li
- a Department of Endocrinology , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Zhenjie Sun
- a Department of Endocrinology , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Jiajia Liu
- a Department of Endocrinology , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Weihua Wu
- a Department of Endocrinology , The First Affiliated Hospital of Harbin Medical University , Harbin , China
| |
Collapse
|