1
|
Utembe W, Kamng'ona AW. Inhalation exposure to chemicals, microbiota dysbiosis and adverse effects on humans. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:176938. [PMID: 39414049 DOI: 10.1016/j.scitotenv.2024.176938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/21/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
As revealed by culture-independent methodologies, disruption of the normal lung microbiota (LM) configuration (LM dysbiosis) is a potential mediator of adverse effects from inhaled chemicals. LM, which consists of microbiota in the upper and lower respiratory tract, is influenced by various factors, including inter alia environmental exposures. LM dysbiosis has been associated with multiple respiratory pathologies such as asthma, lung cancer, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF). Chemically-induced LM dysbiosis appears to play significant roles in human respiratory diseases, as has been shown for some air pollutants, cigarette smoke and some inhalable chemical antibiotics. Lung microbiota are also linked with the central nervous system (CNS) in the so-called lung-brain axis. Inhaled chemicals that undergo mucociliary clearance may be linked to respiratory conditions through gut microbiota (GM) dysbiosis in the so-called Gut-Lung axis. However, current linkages of various disease states to LM appears to be associative, with causal linkages requiring further studies using more robust approaches, methods and techniques that are different from those applied in studies involving (GM). Most importantly, the sampling techniques determine the level of risk of cross contamination. Furthermore, the development of continuous or semi-continuous systems designed to replicate the lung microbiome will go a long way to further LM dysbiosis studies. These challenges notwithstanding, the preponderance of evidence points to the significant role of LM-mediated chemical toxicity in human disease and conditions.
Collapse
Affiliation(s)
- W Utembe
- Toxicology and Biochemistry Department, National Institute for Occupational Health, National Health Laboratory Services, Johannesburg 2000, South Africa; Environmental Health Division, School of Public Health and Family Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - A W Kamng'ona
- School of Life Sciences and Allied Health Professions, Kamuzu University of Health Sciences, Blantyre Campus, Mahatma Gandhi Road, Blantyre 312224, Malawi
| |
Collapse
|
2
|
Godos J, Romano GL, Gozzo L, Laudani S, Paladino N, Dominguez Azpíroz I, Martínez López NM, Giampieri F, Quiles JL, Battino M, Galvano F, Drago F, Grosso G. Resveratrol and vascular health: evidence from clinical studies and mechanisms of actions related to its metabolites produced by gut microbiota. Front Pharmacol 2024; 15:1368949. [PMID: 38562461 PMCID: PMC10982351 DOI: 10.3389/fphar.2024.1368949] [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: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Cardiovascular diseases are among the leading causes of mortality worldwide, with dietary factors being the main risk contributors. Diets rich in bioactive compounds, such as (poly)phenols, have been shown to potentially exert positive effects on vascular health. Among them, resveratrol has gained particular attention due to its potential antioxidant and anti-inflammatory action. Nevertheless, the results in humans are conflicting possibly due to interindividual different responses. The gut microbiota, a complex microbial community that inhabits the gastrointestinal tract, has been called out as potentially responsible for modulating the biological activities of phenolic metabolites in humans. The present review aims to summarize the main findings from clinical trials on the effects of resveratrol interventions on endothelial and vascular outcomes and review potential mechanisms interesting the role of gut microbiota on the metabolism of this molecule and its cardioprotective metabolites. The findings from randomized controlled trials show contrasting results on the effects of resveratrol supplementation and vascular biomarkers without dose-dependent effect. In particular, studies in which resveratrol was integrated using food sources, i.e., red wine, reported significant effects although the resveratrol content was, on average, much lower compared to tablet supplementation, while other studies with often extreme resveratrol supplementation resulted in null findings. The results from experimental studies suggest that resveratrol exerts cardioprotective effects through the modulation of various antioxidant, anti-inflammatory, and anti-hypertensive pathways, and microbiota composition. Recent studies on resveratrol-derived metabolites, such as piceatannol, have demonstrated its effects on biomarkers of vascular health. Moreover, resveratrol itself has been shown to improve the gut microbiota composition toward an anti-inflammatory profile. Considering the contrasting findings from clinical studies, future research exploring the bidirectional link between resveratrol metabolism and gut microbiota as well as the mediating effect of gut microbiota in resveratrol effect on cardiovascular health is warranted.
Collapse
Affiliation(s)
- Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Lucia Gozzo
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-S. Marco”, Catania, Italy
| | - Samuele Laudani
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nadia Paladino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Irma Dominguez Azpíroz
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
- Universidade Internacional do Cuanza, Cuito, Angola
- Universidad de La Romana, La Romana, Dominican Republic
| | - Nohora Milena Martínez López
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
- Universidad Internacional Iberoamericana, Campeche, Mexico
- Fundación Universitaria Internacional de Colombia, Bogotá, Colombia
| | - Francesca Giampieri
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
- Department of Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - José L. Quiles
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, Parque Tecnologico de la Salud, Granada, Spain
- Research and Development Functional Food Centre (CIDAF), Health Science Technological Park, Granada, Spain
| | - Maurizio Battino
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
- Department of Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
- International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, Catania, Italy
| |
Collapse
|
3
|
Yuan L, Li Y, Chen M, Xue L, Wang J, Ding Y, Gu Q, Zhang J, Zhao H, Xie X, Wu Q. Therapeutic applications of gut microbes in cardiometabolic diseases: current state and perspectives. Appl Microbiol Biotechnol 2024; 108:156. [PMID: 38244075 PMCID: PMC10799778 DOI: 10.1007/s00253-024-13007-7] [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: 09/06/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Cardiometabolic disease (CMD) encompasses a range of diseases such as hypertension, atherosclerosis, heart failure, obesity, and type 2 diabetes. Recent findings about CMD's interaction with gut microbiota have broadened our understanding of how diet and nutrition drive microbes to influence CMD. However, the translation of basic research into the clinic has not been smooth, and dietary nutrition and probiotic supplementation have yet to show significant evidence of the therapeutic benefits of CMD. In addition, the published reviews do not suggest the core microbiota or metabolite classes that influence CMD, and systematically elucidate the causal relationship between host disease phenotypes-microbiome. The aim of this review is to highlight the complex interaction of the gut microbiota and their metabolites with CMD progression and to further centralize and conceptualize the mechanisms of action between microbial and host disease phenotypes. We also discuss the potential of targeting modulations of gut microbes and metabolites as new targets for prevention and treatment of CMD, including the use of emerging technologies such as fecal microbiota transplantation and nanomedicine. KEY POINTS: • To highlight the complex interaction of the gut microbiota and their metabolites with CMD progression and to further centralize and conceptualize the mechanisms of action between microbial and host disease phenotypes. • We also discuss the potential of targeting modulations of gut microbes and metabolites as new targets for prevention and treatment of CMD, including the use of emerging technologies such as FMT and nanomedicine. • Our study provides insight into identification-specific microbiomes and metabolites involved in CMD, and microbial-host changes and physiological factors as disease phenotypes develop, which will help to map the microbiome individually and capture pathogenic mechanisms as a whole.
Collapse
Affiliation(s)
- Lin Yuan
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Ding
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Science & Engineering, Jinan University, Guangzhou, 510632, China
| | - Qihui Gu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Hui Zhao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| |
Collapse
|
4
|
Cui H, Han S, Dai Y, Xie W, Zheng R, Sun Y, Xia X, Deng X, Cao Y, Zhang M, Shang H. Gut microbiota and integrative traditional Chinese and western medicine in prevention and treatment of heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154885. [PMID: 37302262 DOI: 10.1016/j.phymed.2023.154885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Heart failure (HF) is the terminal stage of multiple cardiovascular diseases, with high mortality and morbidity. More and more studies have proved that gut microbiota may play a role in the process of HF, which is expected to become a new therapeutic target. The combination of traditional Chinese and Western medicine has vast therapeutic potential of complementation against HF. PURPOSE This manuscript expounds on the research progress of mechanisms of gut microbiota participating in the occurrence and prognosis of HF and the role of integrative traditional Chinese and Western medicine from 1987 to 2022. The combination of traditional Chinese and Western medicine in the prevention and treatment of HF from the perspective of gut microbiota has been discussed. METHODS Studies focusing on the effects and their mechanisms of gut microbiota in HF and the role of integrative traditional Chinese and Western medicine were identified and summarized, including contributions from February 1987 until August 2022. The investigation was carried out in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. We searched PubMed, Embase, Cochrane Library, CNKI, Wanfang, and VIP databases up to April 2023 by using the relevant keywords and operators. RESULTS A total of 34 articles were finally included in this review.16 RCTs and 13 basic researches, and 3 clinical research studies involving 7 relevant outcome indicators(cardiac function evaluation index, changes in gut microbiota, inflammatory factors, metabolites of gut microbiota, serum nutritional index protein, quality of life score, intestinal permeability and all-cause mortality). Compared with healthy controls, serum TNF-α and TMAO levels were significantly higher in patients with heart failure [MD = 5.77, 95%CI(4.97, 6.56), p < 0.0001; SMD = 1.92, 95%CI(1.70, 2.14), p < 0.0001]. Escherichia coli and Thick-walled bacteria increased significantly [SMD = -0.99, 95%CI(-1.38, -0.61), p < 0.0001, SMD = 2.58, 95%CI(2.23, 2.93), p < 0.0001];The number of bacteroides and lactobacillus decreased [SMD = -2.29, 95%CI(-2.54, -2.04), p < 0.0001; SMD = -1.55, 95%CI(-1.8, -1.3), p < 0.0001]. There was no difference in bifidobacterium [SMD = 0.16, 95%CI(-0.22, 0.54), p = 0.42]. In the published literature, it is not difficult to see that most of the results are studied and proved based on animal experiments or clinical trials, involving the cellular level, while the mechanism and mode of action of the molecular biology of traditional Chinese medicine are less elaborated, which is related to the characteristics of multi-components and multi-targets of traditional Chinese medicine. The above are the shortcomings of published literature, which can also be the direction of future research. CONCLUSION Heart failure patients have decreased beneficial bacteria such as Bacillus mimics and Lactobacillus in the intestinal flora and increased harmful flora like thick-walled flora. And increase the inflammatory response of the body and the expression of trimethylamine oxide (TMAO) in the serum. And The prevention and treatment of integrative traditional Chinese and Western medicine against heart failure based on gut microbiota and its metabolites is a promising research direction.
Collapse
Affiliation(s)
- Herong Cui
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yanan Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wei Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rui Zheng
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaofeng Xia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaopeng Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yaru Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mei Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
5
|
Kipka H, Schaflinger R, Tomasi R, Pogoda K, Mannell H. The Effects of the Levosimendan Metabolites OR-1855 and OR-1896 on Endothelial Pro-Inflammatory Responses. Biomedicines 2023; 11:biomedicines11030918. [PMID: 36979897 PMCID: PMC10045601 DOI: 10.3390/biomedicines11030918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
The calcium sensitizer levosimendan is used for the treatment of acute decompensated heart failure. A small portion (4–7%) of levosimendan is metabolized to the pharmacologically active metabolite OR-1896 via the inactive intermediate OR-1855. In addition, levosimendan has been shown to exert positive effects on the endothelium in vitro antagonizing vascular dysfunction and inflammation. However, the function of the levosimendan metabolites within this context is still unknown. In this study, we thus investigated the impact of the metabolites OR-1896 and OR-1855 on endothelial inflammatory processes in vitro. We observed a reduction of IL-1β-dependent endothelial adhesion molecule ICAM-1 and VCAM-1 as well as interleukin (IL) -6 expression upon levosimendan treatment but not after treatment with OR-1855 or OR-1896, as assessed by western blotting, flow cytometry, and qRT-PCR. Instead, the metabolites impaired IL-1β-induced ROS formation via inactivation of the MAPK p38, ERK1/2, and JNK. Our results suggest that the levosimendan metabolites OR-1896 and OR-1855 have certain anti-inflammatory properties, partly other than levosimendan. Importantly, they additionally show that the intermediate metabolite OR-1855 does, in fact, have pharmacological effects in the endothelium. This is interesting, as the metabolites are responsible for the long-term therapeutic effects of levosimendan, and heart failure is associated with vascular dysfunction and inflammation.
Collapse
Affiliation(s)
- Hannah Kipka
- Doctoral Program Clinical Pharmacy, University Hospital, LMU Munich, 81377 Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, 82152 Planegg, Germany
| | - Rebecca Schaflinger
- Doctoral Program Clinical Pharmacy, University Hospital, LMU Munich, 81377 Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, 82152 Planegg, Germany
| | - Roland Tomasi
- Department of Anaesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Kristin Pogoda
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, 82152 Planegg, Germany
- Physiology, Institute for Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
| | - Hanna Mannell
- Doctoral Program Clinical Pharmacy, University Hospital, LMU Munich, 81377 Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, 82152 Planegg, Germany
- Physiology, Institute for Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
- Correspondence:
| |
Collapse
|
6
|
Yuzefpolskaya M, Bohn B, Ladanyi A, Khoruts A, Colombo PC, Demmer RT. Oral and gut microbiome alterations in heart failure: Epidemiology, pathogenesis and response to advanced heart failure therapies. J Heart Lung Transplant 2023; 42:291-300. [PMID: 36586790 DOI: 10.1016/j.healun.2022.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/18/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Despite significant advances in therapies, heart failure (HF) remains a progressive disease that, once advanced, is associated with significant death and disability. Cardiac replacement therapies with left ventricular assist device (LVAD) and heart transplantation (HT) are the only treatment options for advanced HF, while lifesaving they can also be lifespan limiting due to the associated complications. Systemic inflammation is mechanistically important in HF pathophysiology and progression. However, directly targeting inflammation in HF has not been beneficial thus far. These failed attempts at therapeutics might be related to our limited understanding of the factors that cause inflammation in HF, and, therefore, to our inability to investigate these triggers in interventional studies. Observational studies have consistently demonstrated associations between alterations in the digestive (gut and oral) microbiome, inflammation and HF risk and progression. Additionally, recent data indicate that these microbial perturbations persist following LVAD and HT, along with residual inflammation and oxidative stress. Furthermore, there is rising recognition of the critical contribution of the microbiome to the metabolism of immunosuppressive drugs after HT. Cumulatively, these findings might posit a mechanistic link between microbiome alterations, systemic inflammation, and adverse outcomes in HF patients before and after cardiac replacement therapies. This review (1) provides an update on available data linking changes in digestive tract microbiota, inflammation, and oxidative stress, to HF pathogenesis and progression; (2) describes evolution of these relationships following LVAD and HT; and (3) outlines present and future intervention strategies that can manipulate the microbiome and possibly modify HF disease trajectory.
Collapse
Affiliation(s)
- Melana Yuzefpolskaya
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York.
| | - Bruno Bohn
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Annamaria Ladanyi
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine University of Minnesota, Minneapolis, Minnesota
| | - Paolo C Colombo
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota; Division of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
7
|
Epigenetics and Gut Microbiota Crosstalk: A potential Factor in Pathogenesis of Cardiovascular Disorders. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120798. [PMID: 36551003 PMCID: PMC9774431 DOI: 10.3390/bioengineering9120798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of mortality, morbidity, and "sudden death" globally. Environmental and lifestyle factors play important roles in CVD susceptibility, but the link between environmental factors and genetics is not fully established. Epigenetic influence during CVDs is becoming more evident as its direct involvement has been reported. The discovery of epigenetic mechanisms, such as DNA methylation and histone modification, suggested that external factors could alter gene expression to modulate human health. These external factors also influence our gut microbiota (GM), which participates in multiple metabolic processes in our body. Evidence suggests a high association of GM with CVDs. Although the exact mechanism remains unclear, the influence of GM over the epigenetic mechanisms could be one potential pathway in CVD etiology. Both epigenetics and GM are dynamic processes and vary with age and environment. Changes in the composition of GM have been found to underlie the pathogenesis of metabolic diseases via modulating epigenetic changes in the form of DNA methylation, histone modifications, and regulation of non-coding RNAs. Several metabolites produced by the GM, including short-chain fatty acids, folates, biotin, and trimethylamine-N-oxide, have the potential to regulate epigenetics, apart from playing a vital role in normal physiological processes. The role of GM and epigenetics in CVDs are promising areas of research, and important insights in the field of early diagnosis and therapeutic approaches might appear soon.
Collapse
|
8
|
Association Between Depressive Symptoms and Cardiac Structure and Function in a Peruvian Population. Glob Heart 2022; 17:78. [DOI: 10.5334/gh.981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
|
9
|
Koniari I, Artopoulou E, Velissaris D, Ainslie M, Mplani V, Karavasili G, Kounis N, Tsigkas G. Biomarkers in the clinical management of patients with atrial fibrillation and heart failure. J Geriatr Cardiol 2021; 18:908-951. [PMID: 34908928 PMCID: PMC8648548 DOI: 10.11909/j.issn.1671-5411.2021.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Atrial fibrillation (AF) and heart failure (HF) are two cardiovascular diseases with an increasing prevalence worldwide. These conditions share common pathophysiologiesand frequently co-exit. In fact, the occurrence of either condition can 'cause' the development of the other, creating a new patient group that demands different management strategies to that if they occur in isolation. Regardless of the temproral association of the two conditions, their presence is linked with adverse cardiovascular outcomes, increased rate of hospitalizations, and increased economic burden on healthcare systems. The use of low-cost, easily accessible and applicable biomarkers may hasten the correct diagnosis and the effective treatment of AF and HF. Both AF and HF effect multiple physiological pathways and thus a great number of biomarkers can be measured that potentially give the clinician important diagnostic and prognostic information. These will then guide patient centred therapeutic management. The current biomarkers that offer potential for guiding therapy, focus on the physiological pathways of miRNA, myocardial stretch and injury, oxidative stress, inflammation, fibrosis, coagulation and renal impairment. Each of these has different utility in current clinincal practice.
Collapse
Affiliation(s)
- Ioanna Koniari
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Eleni Artopoulou
- Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | | | - Mark Ainslie
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
- Division of Cardiovascular Sciences, University of Manchester
| | - Virginia Mplani
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Georgia Karavasili
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Nicholas Kounis
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Grigorios Tsigkas
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| |
Collapse
|
10
|
Chen Y, Pu W, Maswikiti E, Tao P, Li X, Wang D, Gu B, Yu Y, Gao L, Zhao C, Chen H. Intestinal congestion and reperfusion injury: damage caused to the intestinal tract and distal organs. Biosci Rep 2021; 41:BSR20211560. [PMID: 34369557 PMCID: PMC8421592 DOI: 10.1042/bsr20211560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022] Open
Abstract
In clinical practice, intestinal autologous diseases, ailments and organ transplants can cause severe congestive damage to the intestinal tract. However, after the etiological factor is gotten rid of and blood flow is free without any hinderance, further damage to the intestinal wall often occurs, causing other related organ dysfunctions. This ultimately results in intestinal congestion reperfusion injury (ICRI). When the structure and function of the intestine are destroyed, bacteria, metabolites and endotoxins in the intestinal tract perfuse and enter the portal vein through the already compromised intestinal mucosa, to the other organs via the liver. Nevertheless, this gives rise to further aggravation of the injury, and reperfusion injury syndrome occurs. ICRI is a very common complication encountered by clinicians, and its harm is more severe and serious as compared with that caused by ischemia-reperfusion. Quite a few number of studies on ICRI have been reported to date. The exact mechanism of the injury is still idiopathic, and effective treatment strategies are still limited. Based on recent studies, this article is aimed at reviewing the destruction, damage mechanisms resulting from ICRI to the intestinal anatomical sites and distant organs. It is geared towards providing new ideas for the prevention and therapeutic approaches of ICRI.
Collapse
Affiliation(s)
- Yajing Chen
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- Department of Pediatric Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Weigao Pu
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Ewetse Paul Maswikiti
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Pengxian Tao
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xuemei Li
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dengfeng Wang
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Baohong Gu
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yang Yu
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Lei Gao
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chengji Zhao
- Department of Pediatric Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Hao Chen
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Chia YC, Kieneker LM, van Hassel G, Binnenmars SH, Nolte IM, van Zanden JJ, van der Meer P, Navis G, Voors AA, Bakker SJL, De Borst MH, Eisenga MF. Interleukin 6 and Development of Heart Failure With Preserved Ejection Fraction in the General Population. J Am Heart Assoc 2021; 10:e018549. [PMID: 33998283 PMCID: PMC8483531 DOI: 10.1161/jaha.120.018549] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background The cause of heart failure with preserved ejection fraction (HFpEF) is poorly understood, and specific therapies are lacking. Previous studies suggested that inflammation plays a role in the development of HFpEF. Herein, we aimed to investigate in community-dwelling individuals whether a higher plasma interleukin 6 (IL-6) level is associated with an increased risk of developing new-onset heart failure (HF) over time, and specifically HFpEF. Methods and Results We performed a case-cohort study based on the PREVEND (Prevention of Renal and Vascular End-Stage Disease) study, a prospective general population-based cohort study. We included 961 participants, comprising 200 participants who developed HF and a random group of 761 controls. HF with reduced ejection fraction or HFpEF was defined on the basis of the left ventricular ejection fraction of ≤40% or >40%, respectively. In Cox proportional hazard regression analyses, IL-6 levels were statistically significantly associated with the development of HF (hazard ratio [HR], 1.28; 95% CI, 1.02-1.61; P=0.03) after adjustment for key risk factors. Specifically, IL-6 levels were significantly associated with the development of HFpEF (HR, 1.59; 95% CI, 1.16-2.19; P=0.004), whereas the association with HF with reduced ejection fraction was nonsignificant (HR, 1.05; 95% CI, 0.75-1.47; P=0.77). In sensitivity analyses, defining HFpEF as left ventricular ejection fraction ≥50%, IL-6 levels were also significantly associated with the development of HFpEF (HR, 1.47; 95% CI, 1.04-2.06; P=0.03) after adjustment for key risk factors. Conclusions IL-6 is associated with new-onset HFpEF in community-dwelling individuals, independent of potential confounders. Our findings warrant further research to investigate whether IL-6 might be a novel treatment target to prevent HFpEF.
Collapse
Affiliation(s)
- Yook Chin Chia
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands.,Department of Medical Sciences School of Medical and Life Sciences Sunway University Bandar Sunway Selangor Malaysia
| | - Lyanne M Kieneker
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Gaston van Hassel
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - S Heleen Binnenmars
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Ilja M Nolte
- Department of Epidemiology University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Jelmer J van Zanden
- Certe Department of Clinical Chemistry Martini Hospital Groningen Netherlands
| | - Peter van der Meer
- Department of Cardiology University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Gerjan Navis
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Adriaan A Voors
- Department of Cardiology University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Stephan J L Bakker
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Martin H De Borst
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| | - Michele F Eisenga
- Division of Nephrology Department of Internal Medicine University of GroningenUniversity Medical Center Groningen Groningen the Netherlands
| |
Collapse
|
12
|
Zhang Y, Wang Y, Ke B, Du J. TMAO: how gut microbiota contributes to heart failure. Transl Res 2021; 228:109-125. [PMID: 32841736 DOI: 10.1016/j.trsl.2020.08.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/04/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022]
Abstract
An increasing amount of evidence reveals that the gut microbiota is involved in the pathogenesis and progression of various cardiovascular diseases. In patients with heart failure (HF), splanchnic hypoperfusion causes ischemia and intestinal edema, allowing bacterial translocation and bacterial metabolites to enter the blood circulation via an impaired intestinal barrier. This results in local and systemic inflammatory responses. Gut microbe-derived metabolites are implicated in the pathology of multiple diseases, including HF. These landmark findings suggest that gut microbiota influences the host's metabolic health, either directly or indirectly by producing several metabolites. In this review, we mainly discuss a newly identified gut microbiota-dependent metabolite, trimethylamine N-oxide (TMAO), which appears to participate in the pathologic processes of HF and can serve as an early warning marker to identify individuals who are at the risk of disease progression. We also discuss the potential of the gut-TMAO-HF axis as a new target for HF treatment and highlight the current controversies and potentially new and exciting directions for future research.
Collapse
Affiliation(s)
- Yixin Zhang
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Bingbing Ke
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Beijing, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| |
Collapse
|
13
|
Ausoni S, Calamelli S, Saccà S, Azzarello G. How progressive cancer endangers the heart: an intriguing and underestimated problem. Cancer Metastasis Rev 2021; 39:535-552. [PMID: 32152913 DOI: 10.1007/s10555-020-09869-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since it came into being as a discipline, cardio-oncology has focused on the prevention and treatment of cardiotoxicity induced by antitumor chemotherapy and radiotherapy. Over time, it has been proved that even more detrimental is the direct effect generated by cancer cells that release pro-cachectic factors in the bloodstream. Secreted molecules target different organs at a distance, including the heart. Inflammatory and neuronal modulators released by the tumor bulk, either as free molecules or through exosomes, contribute to the pathogenesis of cardiac disease. Progressive cancer causes cachexia and severe cardiac muscle wasting accompanied by cardiomyocyte atrophy, tissue fibrosis, and several functional impairments up to heart failure. The molecular mechanisms responsible for such a cardiac muscle wasting have been partially elucidated in animal models, but minimally investigated in humans, although severe cardiac dysfunction exacerbates global cachexia and hampers efficient anti-cancer treatments. This review provides an overview of cancer-induced structural cardiac and functional damage, drawing on both clinical and scientific research. We start by looking at the pathophysiological mechanisms and evolving epidemiology and go on to discuss prevention, diagnosis, and a multimodal policy of intervention aimed at providing overall prognosis and global care for patients. Despite much interest in the cardiotoxicity of cancer therapies, the direct tumor effect on the heart remains poorly explored. There is still a lack of diagnostic criteria for the identification of the early stages of cardiac disease in cancer patients, while the possibilities that there are for effective prevention are largely underestimated. Research on innovative therapies has claimed considerable advances in preclinical studies, but none of the molecular targets suitable for clinical application has been approved for therapy. These issues are critically discussed here.
Collapse
Affiliation(s)
- Simonetta Ausoni
- Department of Biomedical Sciences, University of Padua, Padova, Italy.
| | - Sara Calamelli
- Department of Cardiology, Local Health Unit 3 Serenissima, Mirano Hospital, Mirano, Venice, Italy
| | - Salvatore Saccà
- Department of Cardiology, Local Health Unit 3 Serenissima, Mirano Hospital, Mirano, Venice, Italy
| | - Giuseppe Azzarello
- Department of Medical Oncology, Local Health Unit 3 Serenissima, Mirano Hospital, Mirano, Venice, Italy.
| |
Collapse
|
14
|
Abstract
Fecal microbial community changes are associated with numerous disease states, including cardiovascular disease (CVD). However, such data are merely associative. A causal contribution for gut microbiota in CVD has been further supported by a multitude of more direct experimental evidence. Indeed, gut microbiota transplantation studies, specific gut microbiota-dependent pathways, and downstream metabolites have all been shown to influence host metabolism and CVD, sometimes through specific identified host receptors. Multiple metaorganismal pathways (involving both microbe and host) both impact CVD in animal models and show striking clinical associations in human studies. For example, trimethylamine N-oxide and, more recently, phenylacetylglutamine are gut microbiota-dependent metabolites whose blood levels are associated with incident CVD risks in large-scale clinical studies. Importantly, a causal link to CVD for these and other specific gut microbial metabolites/pathways has been shown through numerous mechanistic animal model studies. Phenylacetylglutamine, for example, was recently shown to promote adverse cardiovascular phenotypes in the host via interaction with multiple ARs (adrenergic receptors)-a class of key receptors that regulate cardiovascular homeostasis. In this review, we summarize recent advances of microbiome research in CVD and related cardiometabolic phenotypes that have helped to move the field forward from associative to causative results. We focus on microbiota and metaorganismal compounds/pathways, with specific attention paid to short-chain fatty acids, secondary bile acids, trimethylamine N-oxide, and phenylacetylglutamine. We also discuss novel therapeutic strategies for directly targeting the gut microbiome to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Marco Witkowski
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Center for Microbiome and Human Health (M.W., S.L.H.), Cleveland Clinic, OH
| | - Taylor L Weeks
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Department of Cardiovascular Medicine, Heart and Vascular Institute (S.L.H.), Cleveland Clinic, OH
| | - Stanley L Hazen
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Center for Microbiome and Human Health (M.W., S.L.H.), Cleveland Clinic, OH
| |
Collapse
|
15
|
Affiliation(s)
- Jonas BergstrÖm
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Huddinge University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Bengt Lindholm
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Huddinge University Hospital, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
16
|
Woodrow G. Extracellular Water Expansion: Part of the Malnutrition– Inflammation–Atherosclerosis Syndrome? Perit Dial Int 2020. [DOI: 10.1177/089686080602600508] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
17
|
Abstract
Advances in our understanding of how the gut microbiota contributes to human health and diseases have expanded our insight into how microbial composition and function affect the human host. Heart failure is associated with splanchnic circulation congestion, leading to bowel wall oedema and impaired intestinal barrier function. This situation is thought to heighten the overall inflammatory state via increased bacterial translocation and the presence of bacterial products in the systemic blood circulation. Several metabolites produced by gut microorganisms from dietary metabolism have been linked to pathologies such as atherosclerosis, hypertension, heart failure, chronic kidney disease, obesity, and type 2 diabetes mellitus. These findings suggest that the gut microbiome functions like an endocrine organ by generating bioactive metabolites that can directly or indirectly affect host physiology. In this Review, we discuss several newly discovered gut microbial metabolic pathways, including the production of trimethylamine and trimethylamine N-oxide, short-chain fatty acids, and secondary bile acids, that seem to participate in the development and progression of cardiovascular diseases, including heart failure. We also discuss the gut microbiome as a novel therapeutic target for the treatment of cardiovascular disease, and potential strategies for targeting intestinal microbial processes.
Collapse
Affiliation(s)
- W H Wilson Tang
- Center for Microbiome & Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA. .,Department for Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA. .,Center for Clinical Genomics, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA. .,Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA. .,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA.
| | - Daniel Y Li
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| | - Stanley L Hazen
- Center for Microbiome & Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department for Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
18
|
Bartekova M, Radosinska J, Jelemensky M, Dhalla NS. Role of cytokines and inflammation in heart function during health and disease. Heart Fail Rev 2019; 23:733-758. [PMID: 29862462 DOI: 10.1007/s10741-018-9716-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
By virtue of their actions on NF-κB, an inflammatory nuclear transcription factor, various cytokines have been documented to play important regulatory roles in determining cardiac function under both physiological and pathophysiological conditions. Several cytokines including TNF-α, TGF-β, and different interleukins such as IL-1 IL-4, IL-6, IL-8, and IL-18 are involved in the development of various inflammatory cardiac pathologies, namely ischemic heart disease, myocardial infarction, heart failure, and cardiomyopathies. In ischemia-related pathologies, most of the cytokines are released into the circulation and serve as biological markers of inflammation. Furthermore, there is an evidence of their direct role in the pathogenesis of ischemic injury, suggesting cytokines as potential targets for the development of some anti-ischemic therapies. On the other hand, certain cytokines such as IL-2, IL-4, IL-6, IL-8, and IL-10 are involved in the post-ischemic tissue repair and thus are considered to exert beneficial effects on cardiac function. Conflicting reports regarding the role of some cytokines in inducing cardiac dysfunction in heart failure and different types of cardiomyopathies seem to be due to differences in the nature, duration, and degree of heart disease as well as the concentrations of some cytokines in the circulation. In spite of extensive research work in this field of investigation, no satisfactory anti-cytokine therapy for improving cardiac function in any type of heart disease is available in the literature.
Collapse
Affiliation(s)
- Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Jana Radosinska
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic.,Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Marek Jelemensky
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, 351 Tache Avenue, Winnipeg, MB, R2H 2A6, Canada. .,Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
19
|
Wrobel CA, Drazner MH, Ayers CR, Pham DD, La Hoz RM, Grodin JL, Garg S, Mammen PPA, Morlend RM, Araj F, Amin AA, Cornwell WK, Thibodeau JT. Delayed febrile response with bloodstream infections in patients with continuous-flow left ventricular assist devices. J Investig Med 2019; 67:653-658. [PMID: 30696751 DOI: 10.1136/jim-2018-000893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2018] [Indexed: 11/03/2022]
Abstract
Bloodstream infections (BSIs) are common in patients with continuous-flow left ventricular assist devices (CF-LVADs). Whether CF-LVADs modulate the febrile response to BSIs is unknown. We retrospectively compared the febrile response to BSIs in patients with heart failure (HF) with CF-LVADs versus a control population of patients with HF receiving inotropic infusions. BSIs were adjudicated using the Centers for Disease Control and Prevention and the National Healthcare Safety Network criteria. Febrile status (temperature ≥38°C, 100.4 °F), temperature at presentation with BSI, and the highest temperature within 72 hours (Tmax) were collected. We observed 59 BSIs in LVAD patients and 45 BSIs in controls. LVAD patients were more likely to be afebrile and to have a lower temperature at presentation than control (88% vs 58%, p=0.002, and 37°C ±0.7 vs 37.7°C ±1.0, p=0.0009, respectively). By 72 hours, the difference in afebrile status diminished (53% vs 44%, p=0.42), and the Tmax was similar between the LVAD and control groups (37.9°C±0.9 vs 38.2°C±0.8, respectively, p=0.10). In conclusion, at presentation with a BSI, the vast majority of CF-LVAD patients were afebrile, an event which occurred at a higher frequency when compared with patients with advanced HF on chronic inotropes via an indwelling venous catheter. These data alert clinicians to have a very low threshold to obtain blood cultures in CF-LVAD patients even in the absence of fever. Further study is needed to determine whether a delayed or diminished febrile response represents another pathophysiological consequence of CF-LVADs.
Collapse
Affiliation(s)
- Christopher A Wrobel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mark H Drazner
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Colby R Ayers
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David D Pham
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ricardo M La Hoz
- Department of Internal Medicine, Division of Infectious Disease, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Justin L Grodin
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sonia Garg
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Pradeep P A Mammen
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert M Morlend
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Faris Araj
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Alpesh A Amin
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - William K Cornwell
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jennifer T Thibodeau
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
20
|
Verschure DO, Lutter R, van Eck-Smit BLF, Somsen GA, Verberne HJ. Myocardial 123I-mIBG scintigraphy in relation to markers of inflammation and long-term clinical outcome in patients with stable chronic heart failure. J Nucl Cardiol 2018; 25:845-853. [PMID: 27858345 PMCID: PMC5966475 DOI: 10.1007/s12350-016-0697-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/14/2016] [Indexed: 12/14/2022]
Abstract
AIM Chronic heart failure (CHF) results in both increased cardiac sympathetic activity and myocardial inflammation. The aim of this study was to identify the relationship between severity of heart failure (i.e., NT-proBNP and LVEF), cardiac sympathetic activity (123I-mIBG scintigraphy), and measures of inflammation in subjects with stable, optimally treated CHF. In addition, the predictive value for cardiac events (i.e., ventricular arrhythmia, progression of CHF and cardiac death) of 123I-mIBG parameters and these inflammatory markers was evaluated. MATERIALS AND METHODS Fifty-five CHF patients (age 66.3 ± 8.0 years, 78% male, LVEF 22.4 ± 6.3) referred for cardiac 123I-mIBG imaging were included. At 15 minutes (early) and 4 hours (late) after i.v. administration of 123I-mIBG (185 MBq), planar images were acquired. Early Heart/Mediastinum (H/M) ratio, late H/M ratio, and 123I-mIBG washout (WO) were calculated. NT-proBNP and markers of inflammation (i.e., C-reactive protein (CRP), IL-1β, IL-6, IL-8, IL-10, IL-12p40, tumor necrosis factor-α (TNF-α), soluble (s)E-selectin, myeloperoxidase (MPO), plasminogen activator inhibitor-1 (PAI-1), tPA, tumor necrosis factor receptor (TNFR) 1 and 2, and interferon (IFN) α and β) were measured in blood plasma samples, taken just before 123I-mIBG administration. RESULTS Mean early H/M ratio was 2.12 ± 0.39, late H/M ratio was 1.84 ± 0.40, and 123I-mIBG WO was 13.0 ± 10.9. LVEF was the only independent predictor of late H/M ratio (adjusted R 2 = 0.100, p = 0.011). NT-proBNP was an independent predictor of 123I-mIBG WO (adjusted R 2 = 0.090, p = 0.015). CRP, IL12p40, TNF-α, sE-selectin, MPO, PAI-1, tPA, and TNFR2 were not related to late H/M ratio and 123I-mIBG WO. During a median follow-up of 34 months (2-58 months), 13 patients experienced a cardiac event [ventricular arrhythmia (4), progression of CHF (4), and cardiac death (5)]. Univariate Cox regression analysis showed that the risk of a cardiac event was associated with CRP (HR 1.047 [1.013-1.081]), NT-proBNP (HR 1.141 [1.011-1.288]), MPO (HR 0.998 [0.996-1.000]), and late H/M ratio (HR 0.182 [0.035-0.946]). Multivariate Cox regression analysis showed that only CRP, NT-proBNP, MPO, and IL-12p40 were predictors of a cardiac event. CONCLUSION Inflammation and cardiac sympathetic activity seem not to be related in stable CHF patients. This is corroborated by the finding that they both provide prognostic information in this specific CHF population. The current findings should be regarded as insightful but preliminary.
Collapse
Affiliation(s)
- Derk O Verschure
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands.
- Department of Cardiology, Zaans Medical Center, Zaandam, The Netherlands.
| | - René Lutter
- Departments of Respiratory Medicine and Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Berthe L F van Eck-Smit
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| | - G Aernout Somsen
- Cardiology Centers of the Netherlands, Amsterdam, The Netherlands
| | - Hein J Verberne
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, P.O. Box 22700, 1100 DE, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Senthong V, Hudec T, Neale S, Wu Y, Hazen SL, Tang WHW. Relation of Red Cell Distribution Width to Left Ventricular End-Diastolic Pressure and Mortality in Patients With and Without Heart Failure. Am J Cardiol 2017; 119:1421-1427. [PMID: 28285713 DOI: 10.1016/j.amjcard.2017.01.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 01/06/2023]
Abstract
Higher red cell distribution width (RDW) has been associated with poor prognosis in patients with heart failure (HF). RDW is also closely associated with iron deficiency. However, the mechanism underlying this association is unclear. The relation between left ventricular end-diastolic pressure (LVEDP) and RDW has not been studied, especially in those without HF. We examined the relation between LVEDP and RDW in 1,084 consecutive stable patients who underwent elective coronary angiography. We observed that 38% had high LVEDP (>16 mm Hg) and 29% had history of HF. The median RDW was 13.4%, which was higher with increasing LVEDP (p <0.0001) and significantly higher in patients with HF (p <0.0001). Baseline RDW were independently associated with high LVEDP even after multivariable logistic regression analysis (adjusted odds ratio [OR] per unit change 1.14, 95% confidence interval [CI] 1.0 to 1.29, p = 0.044). Interestingly, result were stronger in non-HF cohort (adjusted OR per unit change 1.37, 95% CI 1.13 to 1.67, p = 0.001). In addition, elevated (third vs first tertiles) RDW levels were independently a predictor of high LVEDP and were associated with a 4.8-fold increased 5-year mortality risk (adjusted hazard ratio 4.11, 95% CI 2.12 to 7.96, p <0.0001), even with the addition of B-type natriuretic peptide to the model (adjusted OR for LVEDP 2.25, 95% CI 1.0 to 5.05, p = 0.05; adjusted hazard ratio for mortality 3.79, 95% CI 1.033 to 13.89, p = 0.044, respectively). In conclusion, high RDW levels were observed in patients with or without HF and independently associated with high LVEDP and with mortality.
Collapse
Affiliation(s)
- Vichai Senthong
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Ohio; Queen Sirikit Heart Center of the Northeast, Department of Medicine, Faculty of Medicine, Khon Kaen University, Thailand
| | - Timothy Hudec
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Ohio
| | - Sarah Neale
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Yuping Wu
- Department of Mathematics, Cleveland State University, Cleveland, Ohio
| | - Stanley L Hazen
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Ohio; Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Ohio; Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
22
|
Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ Res 2017; 118:1786-807. [PMID: 27230642 DOI: 10.1161/circresaha.115.306885] [Citation(s) in RCA: 414] [Impact Index Per Article: 59.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/16/2016] [Indexed: 02/07/2023]
Abstract
Obesity is causally linked with the development of cardiovascular disorders. Accumulating evidence indicates that cardiovascular disease is the collateral damage of obesity-driven adipose tissue dysfunction that promotes a chronic inflammatory state within the organism. Adipose tissues secrete bioactive substances, referred to as adipokines, which largely function as modulators of inflammation. The microenvironment of adipose tissue will affect the adipokine secretome, having actions on remote tissues. Obesity typically leads to the upregulation of proinflammatory adipokines and the downregulation of anti-inflammatory adipokines, thereby contributing to the pathogenesis of cardiovascular diseases. In this review, we focus on the microenvironment of adipose tissue and how it influences cardiovascular disorders, including atherosclerosis and ischemic heart diseases, through the systemic actions of adipokines.
Collapse
Affiliation(s)
- José J Fuster
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.).
| | - Noriyuki Ouchi
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.)
| | - Noyan Gokce
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.)
| | - Kenneth Walsh
- From the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (J.J.F., N.G., K.W.); and Department of Molecular Cardiology, Nagoya University School of Medicine, Nagoya, Japan (N.O.).
| |
Collapse
|
23
|
Affiliation(s)
- Stefan D. Anker
- Clinical Cardiology, National Heart Lung Institute, Imperial College School of Medicine, London, UK
- Franz Volhard Klinik (Charité, Campus Berlin-Buch), Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| |
Collapse
|
24
|
Mirhafez SR, Avan A, Tajfard M, Mohammadi S, Moohebati M, Fallah A, Ghazavi H, Savadi H, Ghayour Mobarhan M. Relationship between serum cytokines receptors and matrix metalloproteinase 9 levels and coronary artery disease. J Clin Lab Anal 2016; 31. [PMID: 27910999 DOI: 10.1002/jcla.22100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/29/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The prevalence of coronary artery disease (CAD) is increasing globally, supporting the need for the identification of novel biomarkers. Therefore in the present study, we have explored the association of SIL2A, SIL6R, STNFRI, STNFRII, and MMP9 in CAD patients. METHODS Twenty one patients with angiographically defined CAD with more than 50% occlusion, at least, in one coronary artery and twenty healthy subjects (n=20) without the history of cardiovascular symptoms were enrolled. Demographic and biochemical analysis (e.g. Total Cholesterol (TC), Triglyceride (TG), and HDL-C) were measured in all the subjects. The level of cytokines receptor (SIL2A, SIL6R, SIL6R, STNFRI, STNFRII, and matrix metallopeptidase 9 (MMP9) were evaluated. RESULTS Our results showed the higher level of MMP9 in patients group compared to the control subjects, while no significant differences were detected for other cytokines. In particular the level of MMP9 was significantly (P=.015) increased from 181.16 ng/mL (95%CI: 112.1-199.2) to 192.0 ng/mL (95%CI: 181.5-265.2). Moreover, the sensitivity and specificity of MMP9 were 95.45% and 45%, respectively, as detected by receiver operating characteristic (ROC) curves. CONCLUSION We demonstrate the significant correlation of MMP-9 with CAD with sensitivity of 95.45%, suggesting its role as a biomarker in CAD patients. Further studies in larger population - preferably multicenter setting - are warranted to explore the functional role of this marker in coronary artery disease.
Collapse
Affiliation(s)
- Seyed Reza Mirhafez
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Tajfard
- Department of Health Education and Health Promotion, School of Health, Mashhad University of Medical Science, Mashhad, Iran.,Management and Social Determinants of Health Research Center, School of Health, Mashhad University of Medical Science, Mashhad, Iran
| | - Shabnam Mohammadi
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.,Neurogenic Inflammation Research Centre, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Moohebati
- Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Fallah
- Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Ghazavi
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Savadi
- Department of Medicine, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Biochemistry and Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Wang JF, Ren X, DeAngelis J, Min J, Zhang Y, Hampton TG, Amende I, Morgan JP. Differential Patterns of Cocaine-Induced Organ Toxicity in Murine Heart versus Liver. Exp Biol Med (Maywood) 2016; 226:52-60. [PMID: 11368239 DOI: 10.1177/153537020122600108] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
To determine cocaine's toxicity in different organs, BALB/c mice were intraperitoneally injected daily for 15 days with either saline or cocaine: 10 mg/kg, 30 mg/kg, or 60 mg/kg. Cardiac function, hepatic pathophysiology, heart and liver apoptosis, and tumor necrosis factor (TNF-α) levels were analyzed. After administration of cocaine, cardiac function decreased. Inflammatory cell infiltration and eosinophilic contraction bands were visible in the hearts of mice treated with 60mg/kg cocaine. Moreover, histopathology demonstrated that cocaine caused hepatic necrosis. TdT-mediated dUTP nick end-labeling (TUNEL) staining and DNA ladder analysis indicated that cocaine caused apoptosis in both the heart and liver. Moreover, immunoassay showed that TNF-α levels significantly increased in the heart and liver with cocaine administration. However, our RT-PCR study showed that there was no significant difference in either the heart or liver in the levels of mRNA for TNF-α between cocaine-treated and saline control mice. The present study demonstrated that cocaine is toxic to multiple organs, and at low dose can induce hepatic damage without gross pathological injury to the heart. The results suggest that the liver is more sensitive than the heart to cocaine toxicity, and induction of apoptosis or TNF-α elevation may be a common mechanism responsible for cocaines toxicity.
Collapse
Affiliation(s)
- J F Wang
- The Charles A. Dana Research Institute and Harvard-Thorndike Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Ocampo V, Haaland D, Legault K, Mittoo S, Aitken E. Successful treatment of recurrent pleural and pericardial effusions with tocilizumab in a patient with systemic lupus erythematous. BMJ Case Rep 2016; 2016:bcr-2016-215423. [PMID: 27503940 DOI: 10.1136/bcr-2016-215423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A 22-year-old Caucasian man presented to hospital with pleuritic chest pain. He had had a history of a sun-sensitive rash a year prior. Workup revealed normal cardiac enzymes and chest X-ray. However, electrocardiogram revealed ST elevation and PR depression, and echocardiogram revealed a slight pericardial effusion without other findings. A diagnosis of pericarditis was made. Subsequently, he was found to be positive for antinuclear antibodies (ANAs), as well as antibodies to SSA, SSB and double-stranded DNA; C3 was low, and C4 was undetectable. A diagnosis of systemic lupus erythematosus was made. The patient initially responded to high-dose ibuprofen. One month later, he developed a new pericardial effusion, this time with concomitant massive left-sided pleural effusion, requiring three separate thoracenteses draining a total of 6 L of pleural fluid. The recurrent effusion failed to respond to high-dose corticosteroid treatment. Owing to the severity and rapidity of the recurrence of pleural and pericardial effusion, intravenous tocilizumab was administered. The patient had excellent clinical and radiographic improvement. This case shows that tocilizumab may have a role in the treatment of intractable pleuropericardial effusion and other forms of lupus-associated serositis.
Collapse
Affiliation(s)
- Vanessa Ocampo
- Internal Medicine Program, McMaster University, Hamilton, Ontario, Canada
| | - Derek Haaland
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada The Waterside Clinic, Barrie, Ontario, Canada
| | - K Legault
- Division of Rheumatology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Shika Mittoo
- Division of Rheumatology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
27
|
Jandali B, Tang WHW, Husni E. Heart Failure and Inflammatory Arthritis: the Relationship of Systemic Inflammation. CURRENT CARDIOVASCULAR RISK REPORTS 2016. [DOI: 10.1007/s12170-016-0497-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
28
|
Emerging importance of chemokine receptor CXCR3 and its ligands in cardiovascular diseases. Clin Sci (Lond) 2016; 130:463-78. [DOI: 10.1042/cs20150666] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The CXC chemokines, CXCL4, -9, -10, -11, CXCL4L1, and the CC chemokine CCL21, activate CXC chemokine receptor 3 (CXCR3), a cell-surface G protein-coupled receptor expressed mainly by Th1 cells, cytotoxic T (Tc) cells and NK cells that have a key role in immunity and inflammation. However, CXCR3 is also expressed by vascular smooth muscle and endothelial cells, and appears to be important in controlling physiological vascular function. In the last decade, evidence from pre-clinical and clinical studies has revealed the participation of CXCR3 and its ligands in multiple cardiovascular diseases (CVDs) of different aetiologies including atherosclerosis, hypertension, cardiac hypertrophy and heart failure, as well as in heart transplant rejection and transplant coronary artery disease (CAD). CXCR3 ligands have also proven to be valid biomarkers for the development of heart failure and left ventricular dysfunction, suggesting an underlining pathophysiological relation between levels of these chemokines and the development of adverse cardiac remodelling. The observation that several of the above-mentioned chemokines exert biological actions independent of CXCR3 provides both opportunities and challenges for developing effective drug strategies. In this review, we provide evidence to support our contention that CXCR3 and its ligands actively participate in the development and progression of CVDs, and may additionally have utility as diagnostic and prognostic biomarkers.
Collapse
|
29
|
Colombo PC, Doran AC, Onat D, Wong KY, Ahmad M, Sabbah HN, Demmer RT. Venous congestion, endothelial and neurohormonal activation in acute decompensated heart failure: cause or effect? Curr Heart Fail Rep 2016; 12:215-22. [PMID: 25740404 DOI: 10.1007/s11897-015-0254-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Venous congestion and endothelial and neurohormonal activation are known to occur in acute decompensated heart failure (ADHF), yet the temporal role of these processes in the pathophysiology of decompensation is not fully understood. Conventional wisdom presumes congestion to be a consequence of worsening cardiovascular function; however, the biomechanically driven effects of venous congestion are biologically plausible contributors to ADHF that remain largely unexplored in vivo. Recent experimental evidence from human models suggests that fluid accumulation and venous congestion are not simply consequences of poor cardiovascular function, but rather are fundamental pro-oxidant, pro-inflammatory, and hemodynamic stimuli that contribute to acute decompensation. The latest advances in the monitoring of volume status using implantable devices allow for the detection of venous congestion before symptoms arise. This may ultimately lead to improved treatment strategies including not only diuretics, but also specific, adjuvant interventions to counteract endothelial and neurohormonal activation during early preclinical decompensation.
Collapse
Affiliation(s)
- Paolo C Colombo
- Division of Cardiology, College of Physicians & Surgeons, Department of Medicine, Columbia University, 622 West 168th Street, PH 12-134, New York, NY, 10032, USA,
| | | | | | | | | | | | | |
Collapse
|
30
|
Gouweleeuw L, Naudé PJW, Rots M, DeJongste MJL, Eisel ULM, Schoemaker RG. The role of neutrophil gelatinase associated lipocalin (NGAL) as biological constituent linking depression and cardiovascular disease. Brain Behav Immun 2015; 46:23-32. [PMID: 25576802 DOI: 10.1016/j.bbi.2014.12.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/11/2014] [Accepted: 12/23/2014] [Indexed: 12/20/2022] Open
Abstract
Depression is more common in patients with cardiovascular disease than in the general population. Conversely, depression is a risk factor for developing cardiovascular disease. Comorbidity of these two pathologies worsens prognosis. Several mechanisms have been indicated in the link between cardiovascular disease and depression, including inflammation. Systemic inflammation can have long-lasting effects on the central nervous system, which could be associated with depression. NGAL is an inflammatory marker and elevated plasma levels are associated with both cardiovascular disease and depression. While patients with depression show elevated NGAL levels, in patients with comorbid heart failure, NGAL levels are significantly higher and associated with depression scores. Systemic inflammation evokes NGAL expression in the brain. This is considered a proinflammatory effect as it is involved in microglia activation and reactive astrocytosis. Animal studies support a direct link between NGAL and depression/anxiety associated behavior. In this review we focus on the role of NGAL in linking depression and cardiovascular disease.
Collapse
Affiliation(s)
- L Gouweleeuw
- Department of Molecular Neurobiology, University of Groningen, Groningen, The Netherlands
| | - P J W Naudé
- Department of Molecular Neurobiology, University of Groningen, Groningen, The Netherlands; Department of Neurology and Alzheimer Research Center, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - M Rots
- Department of Molecular Neurobiology, University of Groningen, Groningen, The Netherlands
| | - M J L DeJongste
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - U L M Eisel
- Department of Molecular Neurobiology, University of Groningen, Groningen, The Netherlands
| | - R G Schoemaker
- Department of Molecular Neurobiology, University of Groningen, Groningen, The Netherlands; Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Abstract
The failing human heart is a bustling network of intra- and inter-cellular signals and related processes attempting to coordinate a repair mechanism for the injured or diseased myocardium. While our understanding of signaling by mode of cytokines is well understood on a systemic level, we are only now coming to elucidate the role of cytokines in cardiac self-regulation. An increasing number of studies are showing now that cardiomyocytes themselves have not only the ability but also the mandate to produce signals, and play direct roles in how these signals are interpreted. One of the families of cytokines employed by distressed cardiac tissue are chemokines. By regulating the movement of pro-inflammatory cell types to sites of injury, we see now how the myocardium responds to stress. Herein we review the participation of these inflammatory mediators and explore the delicate balance between their protective roles and damaging functions.
Collapse
Affiliation(s)
- Andrew A Jarrah
- Department of Medicine, Division of Cardiovascular Research Center, Mount Sinai School of Medicine, 1 Gustave L Levy Place, Box 1030, New York, NY 10029, USA
| | | |
Collapse
|
32
|
Tham YK, Bernardo BC, Ooi JYY, Weeks KL, McMullen JR. Pathophysiology of cardiac hypertrophy and heart failure: signaling pathways and novel therapeutic targets. Arch Toxicol 2015; 89:1401-38. [DOI: 10.1007/s00204-015-1477-x] [Citation(s) in RCA: 371] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 12/18/2022]
|
33
|
Yucel H, Kaya H, Zorlu A, Yıldırımlı K, Sancakdar E, Gunes H, Kurt R, Ozgul U, Turgut O, Yilmaz M. Cancer antigen 125 levels and increased risk of new-onset atrial fibrillation. Herz 2014; 40 Suppl 2:119-24. [DOI: 10.1007/s00059-014-4148-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/12/2014] [Accepted: 08/15/2014] [Indexed: 12/12/2022]
|
34
|
Maier-Moore JS, Horton CG, Mathews SA, Confer AW, Lawrence C, Pan Z, Coggeshall KM, Farris AD. Interleukin-6 deficiency corrects nephritis, lymphocyte abnormalities, and secondary Sjögren's syndrome features in lupus-prone Sle1.Yaa mice. Arthritis Rheumatol 2014; 66:2521-31. [PMID: 24891301 PMCID: PMC4146640 DOI: 10.1002/art.38716] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/15/2014] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To assess disease features in Sle1.Yaa mice with genetic interleukin-6 (IL-6) deficiency. METHODS Sera and tissues were collected from C57BL/6 (B6), Sle1.Yaa, and Sle1.Yaa.IL-6(-/-) mice and analyzed for various features of disease. Using serum samples, autoantibody specificities were determined by enzyme-linked immunosorbent assay (ELISA) and indirect immunofluorescence, cytokine production was analyzed by Luminex and ELISA, and levels of blood urea nitrogen were determined by ELISA. Renal, lung, and salivary gland tissue sections were evaluated for pathologic changes. Lymphocyte phenotypes, including CD4+ T cell cytokine production, and those of follicular and extrafollicular T helper subsets, germinal center B cells, and plasma cells, were determined using flow cytometry. RESULTS IL-6 deficiency not only ameliorated autoantibody production and renal disease in this model, but also effectively reduced inflammation of lungs and salivary glands. Furthermore, IL-6 deficiency abrogated differentiation of Th1 and extrafollicular T helper cells, germinal center B cells, and plasma cells in the spleen and eliminated renal T cells with IL-17, interferon-γ, and IL-21 production potential. CONCLUSION Our findings highlight IL-6-mediated T cell aberrations in Yaa-driven autoimmunity and support the concept of therapeutic IL-6/IL-6 receptor blockade in systemic lupus erythematosus and Sjögren's syndrome by impairing the production of autoantibodies and lymphocytic infiltration of the kidneys, lungs, and salivary glands.
Collapse
Affiliation(s)
- Jacen S. Maier-Moore
- Department of Clinical Laboratory Sciences, University of Texas at El Paso, El Paso, Texas
- The Department of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, 73014
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | - Christopher G. Horton
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
- The Department of Microbiology and Immunology, OUHSC, Oklahoma City, OK, 73014
| | - Shirley A. Mathews
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | - Anthony W. Confer
- The Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, 74078
| | - Christina Lawrence
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | - Zijian Pan
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | | | - A. Darise Farris
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
- The Department of Microbiology and Immunology, OUHSC, Oklahoma City, OK, 73014
| |
Collapse
|
35
|
|
36
|
Does intraoperative ulinastatin improve postoperative clinical outcomes in patients undergoing cardiac surgery: a meta-analysis of randomized controlled trials. BIOMED RESEARCH INTERNATIONAL 2014; 2014:630835. [PMID: 24734237 PMCID: PMC3964764 DOI: 10.1155/2014/630835] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 11/04/2013] [Accepted: 12/02/2013] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The systematic meta-analysis of randomized controlled trials (RCTs) evaluated the effects of intraoperative ulinastatin on early-postoperative recovery in patients undergoing cardiac surgery. METHODS RCTs comparing intraoperative ulinastatin with placebo in cardiac surgery were searched through PubMed, Cochrane databases, Medline, SinoMed, and the China National Knowledge Infrastructure (1966 to May 20th, 2013). The primary endpoints included hospital mortality, postoperative complication rate, length of stay in intensive care unit, and extubation time. The physiological and biochemical parameters illustrating postoperative cardiac and pulmonary function as well as inflammation response were considered as secondary endpoints. RESULTS Fifteen RCTs (509 patients) met the inclusion criteria. Ulinastatin did not affect hospital mortality, postoperative complication rate, or ICU length of stay but reduced extubation time. Ulinastatin also increased the oxygenation index on postoperative day 1 and reduced the plasma level of cardiac troponin-I. Additionally, ulinastatin inhibited the increased level of tumor necrosis factor-alpha, polymorphonuclear neutrophil elastase, interleukin-6, and interleukin-8 associated with cardiac surgery. CONCLUSION Ulinastatin may be of value for the inhibition of postoperative increased inflammatory agents and most likely provided pulmonary protective effects in cardiac surgery. However, larger adequately powered RCTs are required to define the clinical effect of ulinastatin on postoperative outcomes in cardiac surgery.
Collapse
|
37
|
Colombo PC, Onat D, Harxhi A, Demmer RT, Hayashi Y, Jelic S, LeJemtel TH, Bucciarelli L, Kebschull M, Papapanou P, Uriel N, Schmidt AM, Sabbah HN, Jorde UP. Peripheral venous congestion causes inflammation, neurohormonal, and endothelial cell activation. Eur Heart J 2014; 35:448-54. [PMID: 24265434 PMCID: PMC3924182 DOI: 10.1093/eurheartj/eht456] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIMS Volume overload and venous congestion are typically viewed as a consequence of advanced and of acute heart failure (HF) and renal failure (RF) although it is possible that hypervolaemia itself might be a critical intermediate in the pathophysiology of these diseases. This study aimed at elucidating whether peripheral venous congestion is sufficient to promote changes in inflammatory, neurohormonal, and endothelial phenotype similar to those observed in HF and RF. METHODS To experimentally model peripheral venous congestion, we developed a new method (so-called venous stress test) and applied the methodology on 24 healthy subjects (14 men, age 35 ± 2 years). Venous arm pressure was increased to ∼30 mmHg above the baseline level by inflating a tourniquet cuff around the dominant arm (test arm). Blood and endothelial cells (ECs) were sampled from test and control arm (lacking an inflated cuff) before and after 75 min of venous congestion, using angiocatheters and endovascular wires. Magnetic beads coated with EC-specific antibodies were used for EC separation; amplified mRNA was analysed by Affymetrix HG-U133 Plus 2.0 Microarray. RESULTS Plasma interleukin-6 (IL-6), endothelin-1 (ET-1), angiotensin II (AII), vascular cell adhesion molecule-1 (VCAM-1), and chemokine (C-X-C motif) ligand 2 (CXCL2) were significantly increased in the congested arm. A total of 3437 mRNA probe sets were differentially expressed (P < 0.05) in venous ECs before vs. after testing, including ET-1, VCAM-1, and CXCL2. CONCLUSION Peripheral venous congestion causes release of inflammatory mediators, neurohormones, and activation of ECs. Overall, venous congestion mimicked, notable aspects of the phenotype typical of advanced and of acute HF and RF.
Collapse
Affiliation(s)
- Paolo C. Colombo
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA,Corresponding author. Tel: +1 2123052638, Fax: +1 2123057439,
| | - Duygu Onat
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Ante Harxhi
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Ryan T. Demmer
- Department of Epidemiology, Mailman School of Public Health Columbia University, New York, NY, USA
| | - Yacki Hayashi
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Sanja Jelic
- Division of Pulmonary, Columbia University Medical Center, New York, NY, USA
| | - Thierry H. LeJemtel
- Section of Cardiology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - Panos Papapanou
- Department of Periodontology, Columbia University Medical Center, New York, NY, USA
| | - Nir Uriel
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Ann Marie Schmidt
- Division of Endocrinology, Department of Medicine, New York University, New York, NY, USA
| | - Hani N. Sabbah
- Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Ulrich P. Jorde
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
38
|
Statin-Mediated Low-Density Lipoprotein Lowering in Chronic Congestive Heart Failure. Am J Med Sci 2014; 347:14-22. [DOI: 10.1097/maj.0b013e318273514c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
39
|
Yilmaz H, Gürel OM, Çelik HT, Şahiner E, Yildirim ME, Bilgiç MA, Bavbek N, Akcay A. CA 125 levels and left ventricular function in patients with end-stage renal disease on maintenance hemodialysis. Ren Fail 2013; 36:210-6. [DOI: 10.3109/0886022x.2013.859528] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
40
|
Echocardiography, spirometry, and systemic acute-phase inflammatory proteins in smokers with COPD or CHF: an observational study. PLoS One 2013; 8:e80166. [PMID: 24244639 PMCID: PMC3823838 DOI: 10.1371/journal.pone.0080166] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/29/2013] [Indexed: 11/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and chronic heart failure (CHF) may coexist in elderly patients with a history of smoking. Low-grade systemic inflammation induced by smoking may represent the link between these 2 conditions. In this study, we investigated left ventricular dysfunction in patients primarily diagnosed with COPD, and nonreversible airflow limitation in patients primarily diagnosed with CHF. The levels of circulating high-sensitive C-reactive protein (Hs-CRP), pentraxin 3 (PTX3), interleukin-1β (IL-1 β), and soluble type II receptor of IL-1 (sIL-1RII) were also measured as markers of systemic inflammation in these 2 cohorts. Patients aged ≥50 years and with ≥10 pack years of cigarette smoking who presented with a diagnosis of stable COPD (n=70) or stable CHF (n=124) were recruited. All patients underwent echocardiography, N-terminal pro-hormone of brain natriuretic peptide measurements, and post-bronchodilator spirometry. Plasma levels of Hs-CRP, PTX3, IL-1 β, and sIL-1RII were determined by using a sandwich enzyme-linked immuno-sorbent assay in all patients and in 24 healthy smokers (control subjects). Although we were unable to find a single COPD patient with left ventricular dysfunction, we found nonreversible airflow limitation in 34% of patients with CHF. On the other hand, COPD patients had higher plasma levels of Hs-CRP, IL1 β, and sIL-1RII compared with CHF patients and control subjects (p < 0.05). None of the inflammatory biomarkers was different between CHF patients and control subjects. In conclusion, although the COPD patients had no evidence of CHF, up to one third of patients with CHF had airflow limitation, suggesting that routine spirometry is warranted in patients with CHF, whereas echocardiography is not required in well characterized patients with COPD. Only smokers with COPD seem to have evidence of systemic inflammation.
Collapse
|
41
|
Hu CL, Li H, Xia JM, Li X, Zeng X, Liao XX, Zhan H, Jing XL, Dai G. Ulinastatin improved cardiac dysfunction after cardiac arrest in New Zealand rabbits. Am J Emerg Med 2013; 31:768-74. [DOI: 10.1016/j.ajem.2012.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 11/13/2012] [Indexed: 01/25/2023] Open
|
42
|
Colombo PC, Ganda A, Lin J, Onat D, Harxhi A, Iyasere JE, Uriel N, Cotter G. Inflammatory activation: cardiac, renal, and cardio-renal interactions in patients with the cardiorenal syndrome. Heart Fail Rev 2013; 17:177-90. [PMID: 21688186 DOI: 10.1007/s10741-011-9261-3] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although inflammation is a physiologic response designed to protect us from infection, when unchecked and ongoing it may cause substantial harm. Both chronic heart failure (CHF) and chronic kidney disease (CKD) are known to cause elaboration of several pro-inflammatory mediators that can be detected at high concentrations in the tissues and blood stream. The biologic sources driving this chronic inflammatory state in CHF and CKD are not fully established. Traditional sources of inflammation include the heart and the kidneys which produce a wide range of pro-inflammatory cytokines in response to neurohormones and sympathetic activation. However, growing evidence suggests that non-traditional biomechanical mechanisms such as venous and tissue congestion due to volume overload are also important as they stimulate endotoxin absorption from the bowel and peripheral synthesis and release of pro-inflammatory mediators. Both during the chronic phase and, more rapidly, during acute exacerbations of CHF and CKD, inflammation and congestion appear to amplify each other resulting in a downward spiral of worsening cardiac, vascular, and renal functions that may negatively impact patients' outcome. Anti-inflammatory treatment strategies aimed at attenuating end organ damage and improving clinical prognosis in the cardiorenal syndrome have been disappointing to date. A new therapeutic paradigm may be needed, which involves different anti-inflammatory strategies for individual etiologies and stages of CHF and CKD. It may also include specific (short-term) anti-inflammatory treatments that counteract inflammation during the unsettled phases of clinical decompensation. Finally, it will require greater focus on volume overload as an increasingly significant source of systemic inflammation in the cardiorenal syndrome.
Collapse
Affiliation(s)
- Paolo C Colombo
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, College of Physicians and Surgeons, New York, NY, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Massin MM. Relation between red cell distribution width and clinical outcome after surgery for congenital heart disease in children. Pediatr Cardiol 2012; 33:1021-5. [PMID: 22314369 DOI: 10.1007/s00246-012-0220-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 12/19/2011] [Indexed: 01/31/2023]
Abstract
Recent studies have reported a strong association between increased red cell distribution width (RDW) and the risk of adverse outcomes for adults with heart failure. This study investigated the association between preoperative RDW and postoperative clinical outcomes for children with cardiac disease. The relation between preoperative RDW and the length of postoperative stay was tested with 688 consecutive children undergoing surgery for congenital heart disease (CHD). The RDW was significantly higher in patients who died during the postoperative hospital stay (mean, 18.34 ± 4.69 vs 16.12 ± 2.84; p = 0.004). The risk of postoperative death was five times higher for patients with an RDW of 16% or more. In the general study population, RDW correlated with the intensive care unit (ICU) stay (p < 0.0001) and with the total hospital stay in the local population (p < 0.0001). The correlation between RDW and ICU stay was stronger for patients with acyanotic CHD (p < 0.0001) than for those with cyanotic CHD (p = 0.0007), and for the subpopulation of patients with acyanotic CHD and normal hemoglobin level (p < 0.0001) than for anemic patients with acyanotic CHD (p = 0.025). Preoperative RDW is a strong predictor of an adverse outcome in children undergoing surgery for CHD, especially in nonanemic patients, for whom it reflects an underlying inflammatory stress.
Collapse
Affiliation(s)
- Martial M Massin
- Division of Pediatric Cardiology, Queen Fabiola Children's University Hospital (HUDERF), Free University of Brussels (ULB), Avenue J. J. Crocq, 15, B-1020 Brussels, Belgium.
| |
Collapse
|
44
|
Bravo Tobar I, Parra F, Nello Pérez C, Rodríguez-Bonfante C, Useche F, Bonfante-Cabarcas R. Prevalence of Trypanosoma cruzi antibodies and inflammatory markers in uncompensated heart failure. Rev Soc Bras Med Trop 2012; 44:691-6. [PMID: 22231242 DOI: 10.1590/s0037-86822011000600008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 07/28/2010] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Heart failure (HF) represents the final stage of chronic chagasic cardiomyopathy (CChC). The diagnosis of CChC is based on the demonstration of anti-Trypanosoma cruzi antibodies (aTcAg) and clinical and epidemiological data. In Venezuela, there are no data about the prevalence of chagasic HF. The aim of this study was to determine the epidemiological, clinical, and inflammatory risk factors associated with seronegative or seropositive HF patients. METHODS We performed a cross-sectional study in the Venezuelan central-west states among a healthy rural population and in patients admitted to the emergency room with uncompensated HF. RESULTS The seroprevalence rates of Trypanosoma cruzi antibodies were 11.2% and 40.1% in the healthy population and in HF patients, respectively. Seropositivity in healthy individuals was associated with age, knowledge on triatomine vectors, and having seen wild reservoirs in the house; in HF patients, with contact with the vector and previous clinical diagnosis of Chagas' disease; and in both groups taken together, with age, knowledge on triatomines, and HF. Seropositive patients had prolonged QRS, decreased ejection fraction, and high serum magnesium, all significant as compared with HF seronegative cases. Left atrium enlargement and ventricular hypertrophy were most frequently observed in HF seronegative patients. CRP, IL6, ILβ1, IL2, and FNTα were elevated in 94.5%, 48%, 17.8%, 13.7%, and 6.9% of HF patients, respectively, but only IL2 levels were associated with chagasic HF. CONCLUSIONS There is a high prevalence of aTcAg in HF patients from the central-west region of Venezuela, and their epidemiological, clinical, and inflammatory features are discreetly different as compared with those of seronegative cases.
Collapse
Affiliation(s)
- Iván Bravo Tobar
- Unidade de Bioquímica, Decanato de Ciencias de la Salud, Universidade Centro-Occidental Lisandro Alvarado Barquisimeto, Estado Lara, Venezuela
| | | | | | | | | | | |
Collapse
|
45
|
Xin W, Wei W, Li X. Effects of fish oil supplementation on inflammatory markers in chronic heart failure: a meta-analysis of randomized controlled trials. BMC Cardiovasc Disord 2012; 12:77. [PMID: 22994912 PMCID: PMC3507701 DOI: 10.1186/1471-2261-12-77] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 09/18/2012] [Indexed: 11/10/2022] Open
Abstract
Background Effects of fish oil on systematic inflammation in chronic heart failure remain unclear. In this meta-analysis, we aimed to evaluate the influence of fish oil supplementation on circulating levels of inflammatory markers in patients with chronic heart failure. Methods Human randomized controlled trials, which compared the effects of fish oil supplementation with placebo in patients with chronic heart failure, were identified by systematic search of Medline, Embase, Cochrane’s library and references cited in related reviews and studies up to November 2011. Outcome measures comprised the changes of circulating inflammatory markers. Meta-analysis was performed with the fixed-effect model or random-effect model according to the heterogeneity. Results A total of seven trials with eight study arms were included. The pooled results indicated circulating levels of tumor necrosis factor α (SMD = -0.62, 95% CI -1.08 to -0.16, p = 0.009), interleukin 1 (SMD = -1.24, 95% CI -1.56 to -0.91, p < 0.001) and interleukin 6 (SMD = -0.81, 95% CI -1.48 to -0.14, p = 0.02) were significantly decreased after fish oil supplementation; however, high sensitivity C reactive protein, soluble intracellular adhesion molecular 1 and vascular cell adhesion molecular 1 were not significantly affected. Meta-regression and subgroup analysis results suggested the difference in dose of fish oil and follow-up duration might influence the effects of fish oil on tumor necrosis factor α and interleukin 6. Greater reduction of these two markers might be achieved in patients taking fish oil of a higher dose (over 1000 mg/day) or for a longer duration (over 4 months). Conclusions Limited evidence suggests anti-inflammation may be a potential mechanism underlying the beneficial effects of fish oil for chronic heart failure. Further large-scale and adequately powered clinical trials are needed to confirm these effects.
Collapse
Affiliation(s)
- Wei Xin
- First Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, Peoples Republic of China
| | | | | |
Collapse
|
46
|
Consoli C, Gatta L, Iellamo F, Molinari F, Rosano GMC, Marlier LNJL. Severity of left ventricular dysfunction in heart failure patients affects the degree of serum-induced cardiomyocyte apoptosis. Importance of inflammatory response and metabolism. Int J Cardiol 2012; 167:2859-66. [PMID: 22882964 DOI: 10.1016/j.ijcard.2012.07.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 07/10/2012] [Accepted: 07/21/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND/OBJECTIVES In heart failure pro-inflammatory cytokines contribute to cardiomyocytes loss by apoptosis and play a role in the remodelling of the extracellular matrix (ECM). Myocardial injury recruits endothelial progenitor cells (EPCs) to the site of damage and stimulates their differentiation, contributing to myocardial tissue repair. We investigated if the severity of left ventricular dysfunction in heart failure patients (HF) may influence the ability of serum to induce cardiomyocytes death and whether this effect is affected by inflammation and intracellular oxidative stress pathways. METHODS Adult murine cardiomyocytes HL-5 were incubated with 2% human serum from patients with heart failure (NYHA classes I to IV). Apoptosis was analysed by two different methods. TNF-α, IL-1β, IL-6, matrix metalloproteinase 1 (MMP-1) and tissue inhibitor of metalloproteinases 1 (TIMP-1) were measured in sera from patients. RESULTS Cytokine levels were higher in sera from moderate-severe CHF compared to that of patients with mild CHF. Levels of CD117(+) (c-Kit(+)) cells and EPCs were significantly lower in blood from moderate-severe HF patients. Serum from HF patients induced a significantly higher ROS production involving p38 MAPK signalling and apoptosis in cardiomyocytes. NAC treatment prevented serum-induced oxidative effects. The increase of AMPK phosphorylation showed an involvement of FFA β-oxidation during apoptotic stress. CONCLUSIONS All these alterations could be used as predictive factors of worsening in heart failure and culture of cardiomyocytes could be employed to test pharmacological effects.
Collapse
Affiliation(s)
- Claudia Consoli
- Centre for Clinical & Basic Research, Cardiovascular Research Unit, Department. of Medical Sciences, IRCCS San Raffaele Pisana, Rome, Italy
| | | | | | | | | | | |
Collapse
|
47
|
De Gennaro L, Brunetti ND, Montrone D, De Rosa F, Cuculo A, Di Biase M. Inflammatory activation and carbohydrate antigen-125 levels in subjects with atrial fibrillation. Eur J Clin Invest 2012; 42:371-5. [PMID: 21913917 DOI: 10.1111/j.1365-2362.2011.02592.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) might be associated with an inflammatory activation and reduced left ventricular (LV) function. Less is known with regard to newly introduced markers of LV dysfunction such as carbohydrate antigen-125 (CA-125) in subjects with AF. The aim of this study was therefore to assess possible associations between AF, inflammatory markers and CA-125. METHODS AND RESULTS Forty-eight consecutive patients with AF and 58 control patients in sinus rhythm were enrolled in this study. Patients with acute heart failure, chronic inflammatory or neoplastic disease were excluded from the study. Circulating levels of hs-C-reactive protein (CRP), interleukin-8 (IL-8), IL-6, soluble IL-2 receptor (sIL-2r), TNF-α and CA-125 were assessed; all patients underwent clinical examination with NYHA class assessment and echocardiography. Patients with AF were characterised by higher levels of IL-8 (180 ± 266 vs. 39 ± 43 pg/mL, P < 0·001), sIL-2r (987 ± 1045 vs. 680 ± 336 U/mL, P < 0·05) and TNF-α (26 ± 25 vs. 9 ± 4 pg/mL, P < 0·001). Patients with AF duration < 6 months had higher levels of CRP (54 ± 73 vs. 12 ± 14 mg/dL, P < 0·05) and IL-8 (251 ± 225 vs. 99 ± 123 pg/mL, P < 0·05) when compared with AF duration > 6 months. CA-125 levels were not statistically different if comparing subjects with AF with controls and AF > 6 months with AF < 6 months. Among patients with AF, CA-125 levels were significantly related to NYHA class, (r = 0·33, P < 0·05) as well as IL-6 levels (r = 0·31, P < 0·05). Results remained statistically significant even after multivariable correction for age, gender and LV ejection fraction. RESULTS AND CONCLUSION AF is characterised by an inflammatory activation. Impaired functional class in AF subjects might be associated with increased CA-125 levels and higher inflammatory markers.
Collapse
|
48
|
Rosner MH, Ronco C, Okusa MD. The Role of Inflammation in the Cardio-Renal Syndrome: A Focus on Cytokines and Inflammatory Mediators. Semin Nephrol 2012; 32:70-8. [DOI: 10.1016/j.semnephrol.2011.11.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
49
|
Association of IL1B polymorphism with left ventricular systolic dysfunction: a relation with the release of interleukin-1β in stress condition. Pharmacogenet Genomics 2011; 21:579-86. [PMID: 21811191 DOI: 10.1097/fpc.0b013e3283493a05] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Proinflammatory cytokines play a major role in the pathomechanisms of heart failure. Besides this, the influence of mental stress on heart failure is poorly documented despite its effects on sympathetic stimulation of interleukin-1β (IL-1β) secretion. We examined whether the polymorphisms of proinflammatory cytokines are predictors of left ventricular systolic dysfunction (LVSD) and if so, whether such associations are related to the secretion of these cytokines, in 572 consecutive patients under mental stress produced by coronary angiography. METHODS We examined IL-1RN (VNTR), IL1A-889 C>T, IL1B-511 C>T, IL6-174 G>C and TNFA-308 G>A, according to LVSD (left ventricular ejection fraction, <40%). Saliva IL-1β, serum tumour necrosis factor-α and C-reactive protein were assayed in basal (T0 and T2, before and after coronary angiography) and stress (T1) conditions. MAIN RESULTS The 42.1% of patients with LVSD had a 1.5-fold higher frequency of IL1B T allele (P<0.001). IL1B-511TT was associated with LVSD (P=0.008) and with a decrease in IL-1β level in saliva at T1 (P=0.013). IL-1β was the highest at T1 (P<0.001) and was associated with left ventricular ejection fraction (P=0.002). The IL1B TT genotype and the C-reactive protein were the two independent predictors of LVSD in multivariate analysis, with an odds ratio of 2.7 (95% confidence interval: 1.3-5.5; P=0.008) and 1.1 (95% confidence interval: 1.1-1.2; P<0.001), respectively. CONCLUSION IL1B was a predictor of LVSD and of the decreased IL-1β response to stress. This suggests that IL1B exerts an influence on LVSD through its effect on IL-1β secretion.
Collapse
|
50
|
Effects of selective H.E.L.P. LDL-apheresis on plasma inflammatory markers concentration in severe dyslipidemia: Implication for anti-inflammatory response. Cytokine 2011; 56:850-4. [DOI: 10.1016/j.cyto.2011.08.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/22/2011] [Accepted: 08/25/2011] [Indexed: 11/18/2022]
|