1
|
Kittelson KS, Junior AG, Fillmore N, da Silva Gomes R. Cardiovascular-kidney-metabolic syndrome - An integrative review. Prog Cardiovasc Dis 2024; 87:26-36. [PMID: 39486671 PMCID: PMC11619311 DOI: 10.1016/j.pcad.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
The American Heart Association recently defined the complex interactions among the cardiovascular, renal, and metabolic systems as CKM syndrome. To promote better patient outcomes, having a more profound understanding of CKM pathophysiology and pursuing holistic preventative and therapy strategies is critical. Despite many gaps in understanding CKM syndrome, this study attempts to elucidate two of these gaps: the new emerging biomarkers for screening and the role of inflammation in its pathophysiology. For this review, an extensive search for specific terms was conducted in the following databases: PubMed, Scopus, Web of Science, and Google Scholar. Studies were first assessed by title, abstract, keywords, and selected for portfolio according to eligibility criteria, which led to 38 studies. They provided background information about CKM syndrome; data suggested that serum uric acid, leptin, aldosterone, bilirubin, soluble neprilysin, lipocalin-type-prostaglandin-D-synthase, and endocan could be valuable biomarkers for CKM screening; and finally, the inflammation role in CKM.
Collapse
Affiliation(s)
- Katiana Simões Kittelson
- Laboratory of Cardiovascular Pharmacology (LaFaC), Faculty of Health Sciences, Federal University of Grande Dourados (UFGD), Dourados, MS, Brazil; Department of Pharmaceutical Sciences, College of Health and Human Sciences, North Dakota State University, Fargo, ND, United States
| | - Arquimedes Gasparotto Junior
- Laboratory of Cardiovascular Pharmacology (LaFaC), Faculty of Health Sciences, Federal University of Grande Dourados (UFGD), Dourados, MS, Brazil
| | - Natasha Fillmore
- Department of Pharmaceutical Sciences, College of Health and Human Sciences, North Dakota State University, Fargo, ND, United States
| | - Roberto da Silva Gomes
- Department of Pharmaceutical Sciences, College of Health and Human Sciences, North Dakota State University, Fargo, ND, United States.
| |
Collapse
|
2
|
Zannad F, Sanyal AJ, Butler J, Ferreira JP, Girerd N, Miller V, Pandey A, Parikh CR, Ratziu V, Younossi ZM, Harrison SA. MASLD and MASH at the crossroads of hepatology trials and cardiorenal metabolic trials. J Intern Med 2024; 296:24-38. [PMID: 38738988 DOI: 10.1111/joim.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Steatotic liver disease (SLD) is a worldwide public health problem, causing considerable morbidity and mortality. Patients with SLD are at increased risk for major adverse cardiovascular (CV) events, type 2 diabetes mellitus and chronic kidney disease. Conversely, patients with cardiometabolic conditions have a high prevalence of SLD. In addition to epidemiological evidence linking many of these conditions, there is evidence of shared pathophysiological processes. In December 2022, a unique multi-stakeholder, multi-specialty meeting, called MOSAIC (Metabolic multi Organ Science Accelerating Innovation in Clinical Trials) was convened to foster collaboration across metabolic, hepatology, nephrology and CV disorders. One of the goals of the meeting was to consider approaches to drug development that would speed regulatory approval of treatments for multiple disorders by combining liver and cardiorenal endpoints within a single study. Non-invasive tests, including biomarkers and imaging, are needed in hepatic and cardiorenal trials. They can be used as trial endpoints, to enrich trial populations, to diagnose and risk stratify patients and to assess treatment efficacy and safety. Although they are used in proof of concept and phase 2 trials, they are often not acceptable for regulatory approval of therapies. The challenge is defining the optimal combination of biomarkers, imaging and morbidity/mortality outcomes and ensuring that they are included in future trials while minimizing the burden on patients, trialists and trial sponsors. This paper provides an overview of some of the wide array of CV, liver and kidney measurements that were discussed at the MOSAIC meeting.
Collapse
Affiliation(s)
- Faiez Zannad
- Université de Lorraine, Inserm Clinical Investigation Center at Institut Lorrain du Coeur et des Vaisseaux, University Hospital of Nancy, Nancy, France
| | - Arun J Sanyal
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas, USA
- University of Mississippi, Jackson, Mississippi, USA
| | - João Pedro Ferreira
- UnIC@RISE, Cardiovascular Research and Development Center, Department Surgery Physiology, University of Porto, Porto, Portugal
- Centre d'Investigations Cliniques Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
- F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre Hospitalier Régional Universitaire de Nancy, Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, Centre d'Investigation Clinique-Plurithématique, CHRU Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Veronica Miller
- Forum for Collaborative Research, Washington, District of Columbia, USA
- University of California Berkeley School of Public Health, Berkeley, California, USA
| | | | - Chirag R Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vlad Ratziu
- Sorbonne Université, Hôpital Pitié-Salpêtrière, Institute for Cardiometabolism and Nutrition, INSERM UMRS, Paris, France
| | | | - Stephen A Harrison
- Visiting Professor of Hepatology Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Ray AK, Shukla A, Yadav A, Kaur U, Singh AK, Mago P, Bhavesh NS, Chaturvedi R, Tandon R, Shalimar, Kumar A, Malik MZ. A Comprehensive Pilot Study to Elucidate the Distinct Gut Microbial Composition and Its Functional Significance in Cardio-Metabolic Disease. Biochem Genet 2024:10.1007/s10528-024-10847-w. [PMID: 38839647 DOI: 10.1007/s10528-024-10847-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Cardio-metabolic disease is a significant global health challenge with increasing prevalence. Recent research underscores the disruption of gut microbial balance as a key factor in disease susceptibility. We aimed to characterize the gut microbiota composition and function in cardio-metabolic disease and healthy controls. For this purpose, we collected stool samples of 18 subjects (12 diseased, 6 healthy) and we performed metagenomics analysis and functional prediction using QIIME2 and PICRUSt. Furthermore, we carried out assessments of microbe-gene interactions, gene ontology, and microbe-disease associations. Our findings revealed distinct microbial patterns in the diseased group, particularly evident in lower taxonomic levels with significant variations in 14 microbial features. The diseased cohort exhibited an enrichment of Lachnospiraceae family, correlating with obesity, insulin resistance, and metabolic disturbances. Conversely, reduced levels of Clostridium, Gemmiger, and Ruminococcus genera indicated a potential inflammatory state, linked to compromised butyrate production and gut permeability. Functional analyses highlighted dysregulated pathways in amino acid metabolism and energy equilibrium, with perturbations correlating with elevated branch-chain amino acid levels-a known contributor to insulin resistance and type 2 diabetes. These findings were consistent across biomarker assessments, microbe-gene associations, and gene ontology analyses, emphasizing the intricate interplay between gut microbial dysbiosis and cardio-metabolic disease progression. In conclusion, our study unveils significant shifts in gut microbial composition and function in cardio-metabolic disease, emphasizing the broader implications of microbial dysregulation. Addressing gut microbial balance emerges as a crucial therapeutic target in managing cardio-metabolic disease burden.
Collapse
Affiliation(s)
- Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| | - Avaneesh Shukla
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Alka Yadav
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Urvinder Kaur
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Payal Mago
- Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, New Delhi, India
- Campus of Open Learning, University of Delhi, New Delhi, India
| | - Neel Sarovar Bhavesh
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Shalimar
- Department of Gastroenterology, All India Institute of Medical Science, New Delhi, India
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Institute of Bioinformatics, International Technology Park, Whitefield, Bangalore, India
| | - Md Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait.
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
4
|
Di Fiore V, Cappelli F, Del Punta L, De Biase N, Armenia S, Maremmani D, Lomonaco T, Biagini D, Lenzi A, Mazzola M, Tricò D, Masi S, Mengozzi A, Pugliese NR. Novel Techniques, Biomarkers and Molecular Targets to Address Cardiometabolic Diseases. J Clin Med 2024; 13:2883. [PMID: 38792427 PMCID: PMC11122330 DOI: 10.3390/jcm13102883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/01/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Cardiometabolic diseases (CMDs) are interrelated and multifactorial conditions, including arterial hypertension, type 2 diabetes, heart failure, coronary artery disease, and stroke. Due to the burden of cardiovascular morbidity and mortality associated with CMDs' increasing prevalence, there is a critical need for novel diagnostic and therapeutic strategies in their management. In clinical practice, innovative methods such as epicardial adipose tissue evaluation, ventricular-arterial coupling, and exercise tolerance studies could help to elucidate the multifaceted mechanisms associated with CMDs. Similarly, epigenetic changes involving noncoding RNAs, chromatin modulation, and cellular senescence could represent both novel biomarkers and targets for CMDs. Despite the promising data available, significant challenges remain in translating basic research findings into clinical practice, highlighting the need for further investigation into the complex pathophysiology underlying CMDs.
Collapse
Affiliation(s)
- Valerio Di Fiore
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Federica Cappelli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Lavinia Del Punta
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Nicolò De Biase
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Silvia Armenia
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Davide Maremmani
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy; (T.L.)
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy; (T.L.)
| | - Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy; (T.L.)
| | - Matteo Mazzola
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Nicola Riccardo Pugliese
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| |
Collapse
|
5
|
Fang Z, Jia S, Mou X, Li Z, Hu T, Tu Y, Zhao J, Zhang T, Lin W, Lu Y, Feng C, Xia S. Shared genetic architecture and causal relationship between liver and heart disease. iScience 2024; 27:109431. [PMID: 38523778 PMCID: PMC10959668 DOI: 10.1016/j.isci.2024.109431] [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: 10/13/2023] [Revised: 01/08/2024] [Accepted: 03/04/2024] [Indexed: 03/26/2024] Open
Abstract
This study investigates the relationship and genetic mechanisms of liver and heart diseases, focusing on the liver-heart axis (LHA) as a fundamental biological basis. Through genome-wide association study analysis, we explore shared genes and pathways related to LHA. Shared genetic factors are found in 8 out of 20 pairs, indicating genetic correlations. The analysis reveals 53 loci with pleiotropic effects, including 8 loci exhibiting shared causality across multiple traits. Based on SNP-p level tissue-specific multi-marker analysis of genomic annotation (MAGMA) analysis demonstrates significant enrichment of pleiotropy in liver and heart diseases within different cardiovascular tissues and female reproductive appendages. Gene-specific MAGMA analysis identifies 343 pleiotropic genes associated with various traits; these genes show tissue-specific enrichment primarily in the liver, cardiovascular system, and other tissues. Shared risk loci between immune cells and both liver and cardiovascular diseases are also discovered. Mendelian randomization analyses provide support for causal relationships among the investigated trait pairs.
Collapse
Affiliation(s)
- Ziyi Fang
- Department of Gastroenterology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Sixiang Jia
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Xuanting Mou
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Zhe Li
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Tianli Hu
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Yiting Tu
- Department of Orthopedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianqiang Zhao
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Tianlong Zhang
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Wenting Lin
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Yile Lu
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Chao Feng
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Shudong Xia
- Department of Cardiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| |
Collapse
|
6
|
Komaru Y, Ning L, Lama C, Suresh A, Kefaloyianni E, Miller MJ, Herrlich A. Sterile kidney tissue injury induces neutrophil swarming in lung alveolar capillaries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582396. [PMID: 38464306 PMCID: PMC10925262 DOI: 10.1101/2024.02.27.582396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Sterile tissue injury, such as by acute kidney injury, is common in the clinic and frequently associated with respiratory compromise and hypoxemia. We previously described signaling components released by the injured kidney that drive a remote inflammatory response in the lung. How this caused the resultant hypoxemia remained unclear. Here, we report that sterile kidney tissue injury induces rapid intravascular "neutrophil train" formation in lung capillaries, a novel form of neutrophil swarming. Rapid swarming is enhanced by decreased deformability of circulating neutrophils that impedes their lung capillary passage. Classical lung monocytes are required for neutrophil train formation and release CXCL2 to attract and retain stiffened neutrophils in lung capillaries which reduces capillary perfusion. We thus discovered a novel feature of kidney-lung crosstalk after sterile kidney tissue injury, capillary perfusion deficits that lead to reduced oxygenation despite proper alveolar function and ventilation, unlike in infectious inflammatory lung processes, such as bacterial pneumonia.
Collapse
|
7
|
Wang Z, Zhou Y, Xiao X, Liu A, Wang S, Preston RJS, Zaytseva YY, He G, Xiao W, Hennig B, Deng P. Inflammation and cardiometabolic diseases induced by persistent organic pollutants and nutritional interventions: Effects of multi-organ interactions. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 339:122756. [PMID: 37844865 PMCID: PMC10842216 DOI: 10.1016/j.envpol.2023.122756] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
The development and outcome of inflammatory diseases are associated with genetic and lifestyle factors, which include chemical and nonchemical stressors. Persistent organic pollutants (POPs) are major groups of chemical stressors. For example, dioxin-like polychlorinated biphenyls (PCBs), per- and polyfluoroalkyl substances (PFASs), and polybrominated diphenyl ethers (PBDEs) are closely associated with the incidence of inflammatory diseases. The pathology of environmental chemical-mediated inflammatory diseases is complex and may involve disturbances in multiple organs, including the gut, liver, brain, vascular tissues, and immune systems. Recent studies suggested that diet-derived nutrients (e.g., phytochemicals, vitamins, unsaturated fatty acids, dietary fibers) could modulate environmental insults and affect disease development, progression, and outcome. In this article, mechanisms of environmental pollutant-induced inflammation and cardiometabolic diseases are reviewed, focusing on multi-organ interplays and highlighting recent advances in nutritional strategies to improve the outcome of cardiometabolic diseases associated with environmental exposures. In addition, advanced system biology approaches are discussed, which present unique opportunities to unveil the complex interactions among multiple organs and to fuel the development of precision intervention strategies in exposed individuals.
Collapse
Affiliation(s)
- Zhongmin Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China; Irish Centre for Vascular Biology, School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Ireland
| | - Yixuan Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xia Xiao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Aowen Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Shengnan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Ireland
| | - Yekaterina Y Zaytseva
- Superfund Research Center, University of Kentucky, Lexington, KY, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Guangzhao He
- Department of Pharmacy, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu, China
| | - Wenjin Xiao
- Department of Endocrinology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington, KY, USA; Department of Animal and Food Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA
| | - Pan Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
8
|
Tang F, Liu D, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H, Peng C. Targeting endothelial cells with golden spice curcumin: A promising therapy for cardiometabolic multimorbidity. Pharmacol Res 2023; 197:106953. [PMID: 37804925 DOI: 10.1016/j.phrs.2023.106953] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic multimorbidity (CMM) is an increasingly significant global public health concern. It encompasses the coexistence of multiple cardiometabolic diseases, including hypertension, stroke, heart disease, atherosclerosis, and T2DM. A crucial component to the development of CMM is the disruption of endothelial homeostasis. Therefore, therapies targeting endothelial cells through multi-targeted and multi-pathway approaches hold promise for preventing and treatment of CMM. Curcumin, a widely used dietary supplement derived from the golden spice Carcuma longa, has demonstrated remarkable potential in treatment of CMM through its interaction with endothelial cells. Numerous studies have identified various molecular targets of curcumin (such as NF-κB/PI3K/AKT, MAPK/NF-κB/IL-1β, HO-1, NOs, VEGF, ICAM-1 and ROS). These findings highlight the efficacy of curcumin as a therapeutic agent against CMM through the regulation of endothelial function. It is worth noting that there is a close relationship between the progression of CMM and endothelial damage, characterized by oxidative stress, inflammation, abnormal NO bioavailability and cell adhesion. This paper provides a comprehensive review of curcumin, including its availability, pharmacokinetics, pharmaceutics, and therapeutic application in treatment of CMM, as well as the challenges and future prospects for its clinical translation. In summary, curcumin shows promise as a potential treatment option for CMM, particularly due to its ability to target endothelial cells. It represents a novel and natural lead compound that may offer significant therapeutic benefits in the management of CMM.
Collapse
Affiliation(s)
- Fei Tang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dong Liu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
9
|
Muendlein A, Heinzle C, Leiherer A, Brandtner EM, Geiger K, Gaenger S, Fraunberger P, Mader A, Saely CH, Drexel H. Circulating glypican-4 is a new predictor of all-cause mortality in patients with heart failure. Clin Biochem 2023; 121-122:110675. [PMID: 37844682 DOI: 10.1016/j.clinbiochem.2023.110675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Heart failure confers a high burden of morbidity and mortality. However, risk prediction in heart failure patients still is limited. Blood-based biomarkers hold promise to improve clinical risk assessment. Recently we have identified circulating glypican-4 (GPC4) as a significant predictor of mortality in coronary angiography patients and patients with peripheral artery disease. The impact of serum GPC4 on mortality in patients with heart failure is unknown and is addressed in this prospective cohort study. METHODS We prospectively recorded all-cause mortality in 288 patients with heart failure. GPC4 levels were measured using an enzyme-linked immunosorbent assay at baseline. RESULTS During the 24-month follow-up period, 28.1% (n = 81) of the patients died. Serum GPC4 significantly predicted all-cause mortality (hazard ratio (HR) per doublingof GPC4 = 3.57 [2.31-5.53]; P < 0.001). Subgroup analysis showed that GPC4 was significantly associated with all-cause mortality in patients with reduced ejection fraction (HR per doubling = 3.25 [1.75-6.04]; P < 0.001) as well as in those with preserved ejection fraction (HR per doubling = 3.07 [1.22-7.70]; P = 0.017). The association between serum GPC4 and all-cause mortality remained significant in multivariable Cox regression analysis correcting for traditional risk factors (P = 0.035). Results from C-statistics indicated an additional prognostic value of GPC4 relative to NT-proBNP for the prediction of two-year all-cause mortality (P = 0.030). CONCLUSION Circulating GPC4 independently predicts all-cause mortality in patients with heart failure.
Collapse
Affiliation(s)
- Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.
| | - Christine Heinzle
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Medical Central Laboratories, Feldkirch, Austria
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Medical Central Laboratories, Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Eva Maria Brandtner
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Kathrin Geiger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Medical Central Laboratories, Feldkirch, Austria
| | - Stella Gaenger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | | | - Arthur Mader
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Department of Internal Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Christoph H Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Department of Internal Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Vorarlberger Landeskrankenhausbetriebsgesellschaft, Feldkirch, Austria; Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
10
|
Weber JE, Ahmadi M, Boldt LH, Eckardt KU, Edelmann F, Gerhardt H, Grittner U, Haubold K, Hübner N, Kollmus-Heege J, Landmesser U, Leistner DM, Mai K, Müller DN, Nolte CH, Pieske B, Piper SK, Rattan S, Rauch G, Schmidt S, Schmidt-Ott KM, Schönrath K, Schulz-Menger J, Schweizerhof O, Siegerink B, Spranger J, Ramachandran VS, Witzenrath M, Endres M, Pischon T. Protocol of the Berlin Long-term Observation of Vascular Events (BeLOVE): a prospective cohort study with deep phenotyping and long-term follow up of cardiovascular high-risk patients. BMJ Open 2023; 13:e076415. [PMID: 37907297 PMCID: PMC10618970 DOI: 10.1136/bmjopen-2023-076415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/22/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION The Berlin Long-term Observation of Vascular Events is a prospective cohort study that aims to improve prediction and disease-overarching mechanistic understanding of cardiovascular (CV) disease progression by comprehensively investigating a high-risk patient population with different organ manifestations. METHODS AND ANALYSIS A total of 8000 adult patients will be recruited who have either suffered an acute CV event (CVE) requiring hospitalisation or who have not experienced a recent acute CVE but are at high CV risk. An initial study examination is performed during the acute treatment phase of the index CVE or after inclusion into the chronic high risk arm. Deep phenotyping is then performed after ~90 days and includes assessments of the patient's medical history, health status and behaviour, cardiovascular, nutritional, metabolic, and anthropometric parameters, and patient-related outcome measures. Biospecimens are collected for analyses including 'OMICs' technologies (e.g., genomics, metabolomics, proteomics). Subcohorts undergo MRI of the brain, heart, lung and kidney, as well as more comprehensive metabolic, neurological and CV examinations. All participants are followed up for up to 10 years to assess clinical outcomes, primarily major adverse CVEs and patient-reported (value-based) outcomes. State-of-the-art clinical research methods, as well as emerging techniques from systems medicine and artificial intelligence, will be used to identify associations between patient characteristics, longitudinal changes and outcomes. ETHICS AND DISSEMINATION The study was approved by the Charité-Universitätsmedizin Berlin ethics committee (EA1/066/17). The results of the study will be disseminated through international peer-reviewed publications and congress presentations. STUDY REGISTRATION First study phase: Approved WHO primary register: German Clinical Trials Register: https://drks.de/search/de/trial/DRKS00016852; WHO International Clinical Registry Platform: http://apps.who.int/trialsearch/Trial2.aspx?TrialID=DRKS00016852. Recruitment started on July 18, 2017.Second study phase: Approved WHO primary register: German Clinical Trials Register DRKS00023323, date of registration: November 4, 2020, URL: http://www.drks.de/ DRKS00023323. Recruitment started on January 1, 2021.
Collapse
Affiliation(s)
- Joachim E Weber
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Michael Ahmadi
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Leif-Hendrik Boldt
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Kai-Uwe Eckardt
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Frank Edelmann
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Holger Gerhardt
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ulrike Grittner
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Kathrin Haubold
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Hübner
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Jil Kollmus-Heege
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Ulf Landmesser
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Department for Cardiology, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - David M Leistner
- Department of Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Knut Mai
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Endocrinology and Metabolism, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Dominik N Müller
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Christian H Nolte
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Burkert Pieske
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Sophie K Piper
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Institute of Medical Informatics, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Simrit Rattan
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Geraldine Rauch
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Sein Schmidt
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Kai M Schmidt-Ott
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Katharina Schönrath
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Jeanette Schulz-Menger
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Oliver Schweizerhof
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Bob Siegerink
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Joachim Spranger
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Endocrinology and Metabolism, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Cardiovascular Research (CCR), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Vasan S Ramachandran
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- Sections of Preventive Medicine and Epidemiology, and Cardiovascular Medicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Martin Witzenrath
- Division of Pulmonary Inflammation, and Department of Infectious Diseases and Respiratory Medicine, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Germany
| | - Matthias Endres
- Department of Neurology, Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Berlin, Germany
- ExellenceCluster NeuroCure, Berlin, Germany
| | - Tobias Pischon
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Charité- Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|
11
|
Jiménez-Chávez A, Morales-Rubio R, Sánchez-Gasca E, Rivera-Rosas M, Uribe-Ramírez M, Amador-Muñoz O, Martínez-Domínguez YM, Rosas-Pérez I, Choy EH, Herman DA, Kleinman MT, De Vizcaya-Ruiz A. Subchronic co-exposure to particulate matter and fructose-rich-diet induces insulin resistance in male Sprague Dawley rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 100:104115. [PMID: 37075874 DOI: 10.1016/j.etap.2023.104115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 05/03/2023]
Abstract
Insulin resistance (IR) and metabolic disorders are non-pulmonary adverse effects induced by fine particulate matter (PM2.5) exposure. The worldwide pandemic of high fructose sweeteners and fat rich modern diets, also contribute to IR development. We investigated some of the underlying effects of IR, altered biochemical insulin action and Insulin/AKT pathway biomarkers. Male Sprague Dawley rats were subchronically exposed to filtered air, PM2.5, a fructose rich diet (FRD), or PM2.5 + FRD. Exposure to PM2.5 or FRD alone did not induce metabolic changes. However, PM2.5 + FRD induced leptin release, systemic hyperinsulinemia, and Insulin/AKT dysregulation in insulin-sensitive tissues preceded by altered AT1R levels. Histological damage and increased HOMA-IR were also observed from PM2.5 + FRD co-exposure. Our results indicate that the concomitant exposure to a ubiquitous environmental pollutant, such as PM2.5, and a metabolic disease risk factor, a FRD, can contribute to the metabolic disorder pandemic occurring in highly polluted locations.
Collapse
Affiliation(s)
- Arturo Jiménez-Chávez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México
| | - Russell Morales-Rubio
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México
| | - Eliu Sánchez-Gasca
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México
| | - Mónica Rivera-Rosas
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México
| | - Marisela Uribe-Ramírez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México
| | - Omar Amador-Muñoz
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, México
| | - Y Margarita Martínez-Domínguez
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, México
| | - Irma Rosas-Pérez
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, México
| | - Elizabeth H Choy
- Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California Irvine, Irvine, CA, USA
| | - David A Herman
- Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California Irvine, Irvine, CA, USA
| | - Michael T Kleinman
- Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California Irvine, Irvine, CA, USA
| | - Andrea De Vizcaya-Ruiz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Ciudad de México, México; Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
12
|
Sun JY, Du LJ, Shi XR, Zhang YY, Liu Y, Wang YL, Chen BY, Liu T, Zhu H, Liu Y, Ruan CC, Gan Z, Ying H, Yin Z, Gao PJ, Yan X, Li RG, Duan SZ. An IL-6/STAT3/MR/FGF21 axis mediates heart-liver cross-talk after myocardial infarction. SCIENCE ADVANCES 2023; 9:eade4110. [PMID: 37018396 PMCID: PMC10075967 DOI: 10.1126/sciadv.ade4110] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/07/2023] [Indexed: 06/19/2023]
Abstract
The liver plays a protective role in myocardial infarction (MI). However, very little is known about the mechanisms. Here, we identify mineralocorticoid receptor (MR) as a pivotal nexus that conveys communications between the liver and the heart during MI. Hepatocyte MR deficiency and MR antagonist spironolactone both improve cardiac repair after MI through regulation on hepatic fibroblast growth factor 21 (FGF21), illustrating an MR/FGF21 axis that underlies the liver-to-heart protection against MI. In addition, an upstreaming acute interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) pathway transmits the heart-to-liver signal to suppress MR expression after MI. Hepatocyte Il6 receptor deficiency and Stat3 deficiency both aggravate cardiac injury through their regulation on the MR/FGF21 axis. Therefore, we have unveiled an IL-6/STAT3/MR/FGF21 signaling axis that mediates heart-liver cross-talk during MI. Targeting the signaling axis and the cross-talk could provide new strategies to treat MI and heart failure.
Collapse
Affiliation(s)
- Jian-Yong Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Xue-Rui Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yu-Yao Zhang
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yong-Li Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Hong Zhu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Chemistry and Biomedicine Innovation Center (ChemBIC), Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing 210061, China
| | - Hao Ying
- CAS Key Laboratory of Nutrition, Metabolism, and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai 519000, Guangdong, China
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, Guangdong, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
- Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai, China
| |
Collapse
|
13
|
Filosa A, Sawamiphak S. Heart development and regeneration-a multi-organ effort. FEBS J 2023; 290:913-930. [PMID: 34894086 DOI: 10.1111/febs.16319] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/22/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Development of the heart, from early morphogenesis to functional maturation, as well as maintenance of its homeostasis are tasks requiring collaborative efforts of cardiac tissue and different extra-cardiac organ systems. The brain, lymphoid organs, and gut are among the interaction partners that can communicate with the heart through a wide array of paracrine signals acting at local or systemic level. Disturbance of cardiac homeostasis following ischemic injury also needs immediate response from these distant organs. Our hearts replace dead muscles with non-contractile fibrotic scars. We have learned from animal models capable of scarless repair that regenerative capability of the heart does not depend only on competency of the myocardium and cardiac-intrinsic factors but also on long-range molecular signals originating in other parts of the body. Here, we provide an overview of inter-organ signals that take part in development and regeneration of the heart. We highlight recent findings and remaining questions. Finally, we discuss the potential of inter-organ modulatory approaches for possible therapeutic use.
Collapse
Affiliation(s)
- Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Suphansa Sawamiphak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany
| |
Collapse
|
14
|
Zaidi M, Kim SM, Mathew M, Korkmaz F, Sultana F, Miyashita S, Gumerova AA, Frolinger T, Moldavski O, Barak O, Pallapati A, Rojekar S, Caminis J, Ginzburg Y, Ryu V, Davies TF, Lizneva D, Rosen CJ, Yuen T. Bone circuitry and interorgan skeletal crosstalk. eLife 2023; 12:83142. [PMID: 36656634 PMCID: PMC9851618 DOI: 10.7554/elife.83142] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
The past decade has seen significant advances in our understanding of skeletal homeostasis and the mechanisms that mediate the loss of bone integrity in disease. Recent breakthroughs have arisen mainly from identifying disease-causing mutations and modeling human bone disease in rodents, in essence, highlighting the integrative nature of skeletal physiology. It has become increasingly clear that bone cells, osteoblasts, osteoclasts, and osteocytes, communicate and regulate the fate of each other through RANK/RANKL/OPG, liver X receptors (LXRs), EphirinB2-EphB4 signaling, sphingolipids, and other membrane-associated proteins, such as semaphorins. Mounting evidence also showed that critical developmental pathways, namely, bone morphogenetic protein (BMP), NOTCH, and WNT, interact each other and play an important role in postnatal bone remodeling. The skeleton communicates not only with closely situated organs, such as bone marrow, muscle, and fat, but also with remote vital organs, such as the kidney, liver, and brain. The metabolic effect of bone-derived osteocalcin highlights a possible role of skeleton in energy homeostasis. Furthermore, studies using genetically modified rodent models disrupting the reciprocal relationship with tropic pituitary hormone and effector hormone have unraveled an independent role of pituitary hormone in skeletal remodeling beyond the role of regulating target endocrine glands. The cytokine-mediated skeletal actions and the evidence of local production of certain pituitary hormones by bone marrow-derived cells displays a unique endocrine-immune-skeletal connection. Here, we discuss recently elucidated mechanisms controlling the remodeling of bone, communication of bone cells with cells of other lineages, crosstalk between bone and vital organs, as well as opportunities for treating diseases of the skeleton.
Collapse
Affiliation(s)
- Mone Zaidi
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Se-Min Kim
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Mehr Mathew
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Funda Korkmaz
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Farhath Sultana
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Sari Miyashita
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Anisa Azatovna Gumerova
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Tal Frolinger
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Ofer Moldavski
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Orly Barak
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Anusha Pallapati
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Satish Rojekar
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - John Caminis
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Yelena Ginzburg
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Vitaly Ryu
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Terry F Davies
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Daria Lizneva
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | | | - Tony Yuen
- The Mount Sinai Bone Program, Departments of Pharmacological Sciences and of Medicine, and Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| |
Collapse
|
15
|
Lewis JE, Reginald McDaniel H, Woolger JM, Khan SA. The Characterization of the Th1/Th2 Ratio in Moderate-Severe Alzheimer's Disease Patients and Its Response to an Aloe Polymannose-Based Dietary Supplement. J Alzheimers Dis 2023; 96:1723-1737. [PMID: 38007658 DOI: 10.3233/jad-230659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a leading killer of Americans, imparting a tremendous societal toll. Relationships between immune function and inflammation with cognition are well-established in AD, but the Th1/Th2 ratio of immune function is unknown. Describing the Th1/Th2 ratio and its relationship with cognition may shed light on the disease's clinical context. How the Th1/Th2 ratio responds to dietary supplementation is another unknown question in this population. OBJECTIVE The objectives of the study were to: 1) characterize the Th1/Th2 ratio according to IL-2/IL-10, IFN-γ/IL-10, IL-2/IL-4, IFN-γ/IL-4, IL-2/TNF-α, and IFN-γ/TNF-α in subjects with moderate-to-severe AD and in comparison to healthy adults; 2) investigate the effect of an aloe polymannose multinutrient complex (APMC) dietary supplement on the Th1/Th2 ratios over 12 months; and 3) compare the changes in the Th1/Th2 ratios with the changes in cognition from baseline to 12 months. METHODS Subjects consumed 2.5 g of the APMC four times per day for 12 months, and they were assessed on cognition and cytokines at baseline and 12 months. RESULTS The Th1/Th2 ratios in AD patients were significantly higher than the healthy controls, and five of the six ratios decreased from baseline to 12 months follow-up (other than IL-2/TNF-α). Several significant relationships were noted between the changes in Th1/Th2 ratios with cognitive assessments. CONCLUSIONS Our results showed an overall rebalancing of the Th1/Th2 ratio in response to APMC, these changes were related to improved cognition in subjects with moderate-to-severe AD, and the APMC supplement was safely tolerated.
Collapse
Affiliation(s)
- John E Lewis
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Judi M Woolger
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sher Ali Khan
- Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
16
|
Ashton KJ, Kiessling CJ, Thompson JLM, Aziz AY, Thomas WG, Headrick JP, Reichelt ME. Early cardiac aging linked to impaired stress-resistance and transcriptional control of stress response, quality control and mitochondrial pathways. Exp Gerontol 2023; 171:112011. [PMID: 36347360 DOI: 10.1016/j.exger.2022.112011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 10/18/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Phenotypic and transcriptomic evidence of early cardiac aging, and associated mechanisms, were investigated in young to middle-aged male mice (C57Bl/6; ages 8, 16, 32, 48 wks). Left ventricular gene expression (profiled via Illumina MouseWG-6 BeadChips), contractile and coronary function, and stress-resistance were assessed in Langendorff perfused hearts under normoxic conditions and following ischemic insult (20 min global ischemia-45 min reperfusion; I-R). Baseline or normoxic contractile function was unaltered by age, while cardiac and coronary 'reserves' (during β-adrenoceptor stimulation; 1 μM isoproterenol) declined by 48 wks. Resistance to I-R injury fell from 16 to 32 wks. Age-dependent transcriptional changes In un-stressed hearts were limited to 104 genes (>1.3-fold; 0.05 FDR), supporting: up-regulated innate defenses (glutathione and xenobiotic metabolism, chemotaxis, interleukins) and catecholamine secretion; and down-regulated extracellular matrix (ECM), growth factor and survival (PI3K/Akt) signaling. In stressed (post-ischemic) myocardium, ∼15-times as many genes (1528) were age-dependent, grouped into 6 clusters (>1.3-fold change; 0.05 FDR): most changing from 16 wks (45 % up/44 % down), a further 5 % declining from 32 wks. Major age-dependent Biological Processes in I-R hearts reveal: declining ATP metabolism, oxidative phosphorylation, cardiac contraction and morphogenesis, phospholipid metabolism and calcineurin signaling; increasing proteolysis and negative control of MAPK; and mixed changes in nuclear transport and angiogenic genes. Pathway analysis supports reductions in: autophagy, stress response, ER protein processing, mRNA surveillance and ribosome/translation genes; with later falls in mitochondrial biogenesis, oxidative phosphorylation and proteasome genes in I-R hearts. Summarizing, early cardiac aging is evident from 16 to 32 wks in male mice, characterized by: declining cardiovascular reserve and stress-resistance, transcriptomic evidence of constitutive stress and altered catecholamine and survival/growth signaling in healthy hearts; and declining stress response, quality control, mitochondrial energy metabolism and cardiac modeling processes in stressed hearts. These very early changes, potentially key substrate for advanced aging, may inform approaches to healthy aging and cardioprotection in the adult heart.
Collapse
Affiliation(s)
- Kevin J Ashton
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Can J Kiessling
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Jamie-Lee M Thompson
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Aliah Y Aziz
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - John P Headrick
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
17
|
Systematic Review and Meta-Analysis of the Usefulness of Epicardial Fat Thickness as a Non-Invasive Marker of the Presence and Severity of Nonalcoholic Fatty Liver Disease. Biomedicines 2022; 10:biomedicines10092204. [PMID: 36140303 PMCID: PMC9496452 DOI: 10.3390/biomedicines10092204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/12/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
We performed a systematic review and meta-analysis to assess the association between epicardial fat thickness (EFT) and nonalcoholic fatty liver disease (NAFLD). This systematic review was conducted in accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analysis (PRISMA) and was based on a registered protocol (CRD 4201809 5493). We searched Medline and Embase until December 2021 for studies reporting on the association between EFT and NAFLD. Qualitative reviews, meta-analyses and meta-regressions were performed to explore this association. Effect sizes are reported as standardized mean differences. We included 12 studies, comprising 3610 individuals. EFT was evaluated with trans-thoracic echocardiography in nine studies, two studies using cardiac computed tomography and one study using magnetic resonance imaging (MRI). The presence of NAFLD was evaluated using transabdominal liver ultrasound in nine studies. Other studies used histology, magnetic resonance spectroscopy and MRI-derived proton density fat fraction. Liver biopsy was performed to assess the severity of NAFLD in four studies. The random-effects meta-analysis indicated that, as compared to control patients with lean livers, patients with NAFLD displayed significantly higher EFT (standardized mean difference 0.61, 95% confidence interval: 0.47−0.75, p < 0.0001, I2 = 72%). EFT was further significantly higher in patients with severe liver steatosis versus patients with mild−moderate liver steatosis (standardized mean difference 1.21 95% confidence interval: 0.26−2.16, p < 0.001, I2 S = 96%). Through the meta-regression analysis, we found that patients with increasingly higher blood levels of aspartate aminotransferase displayed an increasingly higher depth of association. The current meta-analysis suggests that EFT may represent a useful surrogate for assessing the presence and severity of NAFLD in a non-invasive manner.
Collapse
|
18
|
Zhernakova DV, Sinha T, Andreu-Sánchez S, Prins JR, Kurilshikov A, Balder JW, Sanna S, Franke L, Kuivenhoven JA, Zhernakova A, Fu J. Age-dependent sex differences in cardiometabolic risk factors. NATURE CARDIOVASCULAR RESEARCH 2022; 1:844-854. [PMID: 39196077 PMCID: PMC11357998 DOI: 10.1038/s44161-022-00131-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 08/05/2022] [Indexed: 08/29/2024]
Abstract
Cardiometabolic diseases (CMDs) are a major cause of mortality worldwide, yet men and women present remarkable differences in disease prognosis, onset and manifestation. Here we characterize how sex differences in cardiometabolic risk factors vary with age by examining 45 phenotypes and 6 lifestyle factors in 146,021 participants of the Dutch population cohort Lifelines. We show that sex differences are present in 71% of the studied phenotypes. For 31% of these phenotypes, the phenotypic difference between sexes is dependent on age. CMD risk factors show various patterns of age-related sex differences, ranging from no difference for phenotypes such as body mass index (BMI) to strong age-modified sex differences for lipid levels. We also identify lifestyle factors that influence phenotypes in a sex- and age-dependent manner. These results highlight the importance of taking age into account when studying sex differences in CMDs.
Collapse
Affiliation(s)
- Daria V Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
- Laboratory of Genomic Diversity, Center for Computer Technologies, ITMO University, Saint Petersburg, Russia.
| | - Trishla Sinha
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sergio Andreu-Sánchez
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan-Willem Balder
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Serena Sanna
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Istituto di Ricerca Genetica e Biomedica (IRGB) del Consiglio Nazionale delle Ricerche (CNR), Monserrato, Italy
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan A Kuivenhoven
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
19
|
Hernandez L, Laucyte-Cibulskiene A, Ward LJ, Kautzky-Willer A, Herrero MT, Norris CM, Raparelli V, Pilote L, Stenvinkel P, Kublickiene K. Gender dimension in cardio-pulmonary continuum. Front Cardiovasc Med 2022; 9:916194. [PMID: 36003909 PMCID: PMC9393639 DOI: 10.3389/fcvm.2022.916194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Cardio-pulmonary diseases, which were once regarded as a man's illness, have been one of the leading causes of morbidity and mortality for both men and women in many countries in recent years. Both gender and sex influence the functional and structural changes in the human body and therefore play an important role in disease clinical manifestation, treatment choice, and/or response to treatment and prognosis of health outcomes. The gender dimension integrates sex and gender analysis in health sciences and medical research, however, it is still relatively overlooked suggesting the need for empowerment in the medical research community. Latest advances in the field of cardiovascular research have provided supportive evidence that the application of biological variables of sex has led to the understanding that heart disease in females may have different pathophysiology compared to males, particularly in younger adults. It has also resulted in new diagnostic techniques and a better understanding of symptomatology, while gender analysis has informed more appropriate risk stratification and prevention strategies. The existing knowledge in the pulmonary field shows the higher prevalence of pulmonary disorders among females, however, the role of gender as a socio-cultural construct has yet to be explored for the implementation of targeted interventions. The purpose of this review is to introduce the concept of gender dimension and its importance for the cardiopulmonary continuum with a focus on shared pathophysiology and disease presentation in addition to interrelation with chronic kidney disease. The review presents basic knowledge of what gender dimension means, and the application of sex and gender aspects in cardiovascular medicine with a specific focus on early pulmonary development, pulmonary hypertension, and chronic obstructive pulmonary disease (COPD). Early vascular aging and inflammation have been presented as a potential pathophysiological link, with further interactions between the cardiopulmonary continuum and chronic kidney disease. Finally, implications for potential future research have been provided to increase the impact of gender dimension on research excellence that would add value to everybody, foster toward precision medicine and ultimately improve human health.
Collapse
Affiliation(s)
- Leah Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Agne Laucyte-Cibulskiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Nephrology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Liam J. Ward
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maria-Trinidad Herrero
- Clinical and Experimental Neuroscience, Institutes for Aging Research and Bio-Health Research of Murcia, School of Medicine, University of Murcia, Murcia, Spain
| | - Colleen M. Norris
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
- Cardiovascular and Stroke Strategic Clinical Network, Alberta Health Services, Edmonton, AB, Canada
| | - Valeria Raparelli
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Louise Pilote
- Division of Clinical Epidemiology, Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
20
|
Hofherr A, Williams J, Gan LM, Söderberg M, Hansen PBL, Woollard KJ. Targeting inflammation for the treatment of Diabetic Kidney Disease: a five-compartment mechanistic model. BMC Nephrol 2022; 23:208. [PMID: 35698028 PMCID: PMC9190142 DOI: 10.1186/s12882-022-02794-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/20/2022] [Indexed: 12/25/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of kidney failure worldwide. Mortality and morbidity associated with DKD are increasing with the global prevalence of type 2 diabetes. Chronic, sub-clinical, non-resolving inflammation contributes to the pathophysiology of renal and cardiovascular disease associated with diabetes. Inflammatory biomarkers correlate with poor renal outcomes and mortality in patients with DKD. Targeting chronic inflammation may therefore offer a route to novel therapeutics for DKD. However, the DKD patient population is highly heterogeneous, with varying etiology, presentation and disease progression. This heterogeneity is a challenge for clinical trials of novel anti-inflammatory therapies. Here, we present a conceptual model of how chronic inflammation affects kidney function in five compartments: immune cell recruitment and activation; filtration; resorption and secretion; extracellular matrix regulation; and perfusion. We believe that the rigorous alignment of pathophysiological insights, appropriate animal models and pathology-specific biomarkers may facilitate a mechanism-based shift from recruiting ‘all comers’ with DKD to stratification of patients based on the principal compartments of inflammatory disease activity.
Collapse
Affiliation(s)
- Alexis Hofherr
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden. .,Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Julie Williams
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolic, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, UK
| | - Li-Ming Gan
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolism, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine, Department of Cardiology, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Magnus Söderberg
- Cardiovascular, Renal and Metabolic Safety, Clinical Pharmacology and Safety Sciences, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Pernille B L Hansen
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolic, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, UK.,Wallenberg Center for Molecular and Translational Medicine, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kevin J Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolic, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, UK. .,Centre for Inflammatory Disease, Imperial College London, London, UK.
| |
Collapse
|
21
|
Kikuchi T, Matsuura K, Shimizu T. In vitro circulation model driven by tissue-engineered dome-shaped cardiac tissue. Biofabrication 2022; 14. [PMID: 35688123 DOI: 10.1088/1758-5090/ac77c1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 06/10/2022] [Indexed: 11/11/2022]
Abstract
The heart is an essential organ for animals and humans. With the increased availability of pluripotent stem cells, the use of three-dimensional cardiac tissues consisting of cultured cardiomyocytes in in vitro drug evaluation has been widely studied. Several models have been proposed for the realization of the pump function, which is the original function of the heart. However, there are no models that simulate the human circulatory system using cultured cardiac tissue. This study shows that a dome-shaped cardiac tissue fabricated using the cell sheet stacking technique can achieve a heart-like pump function and circulate culture medium, there by mimicking the human circulatory system. Firstly, human induced pluripotent stem cells were differentiated into autonomously beating cardiomyocytes, and cardiomyocyte cell sheets were created using temperature-responsive culture dishes. A cardiomyocyte sheet and a human dermal fibroblast sheet were stacked using a cell sheet manipulator. This two-layered cell sheet was then inflated to create a dome-shaped cardiac tissue with a base diameter of 8 mm. The volume of the dome-shaped cardiac tissue changed according to the autonomous beating. The stroke volume increased with the culture period and reached 21 ± 8.9 μL (n = 6) on day 21. It also responded to β-stimulant and extracellular calcium concentrations. Internal pressure fluctuations were also recorded under isovolumetric conditions by dedicated culture devices. The peak heights of pulsatile pressure were 0.33 ± 0.048 mmHg (n = 3) under a basal pressure of 0.5 mmHg on day 19. When the tissue was connected to a flow path that had check valves applied, it drove a directional flow with an average flow rate of approximately 1 μL/s. Furthermore, pressure-volume (P-V) diagrams were created from the simultaneous measurement of changes in pressure and volume under three conditions of fluidic resistance. In conclusion, this cardiac model can potentially be used for biological pumps that drive multi-organ chips and for more accurate in vitro drug evaluation using P-V diagrams.
Collapse
Affiliation(s)
- Tetsutaro Kikuchi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, JAPAN
| | - Katsuhisa Matsuura
- Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, JAPAN
| | - Tatsuya Shimizu
- Institute of Advanced BioMedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjyuku-ku,, Tokyo, 162-8666, JAPAN
| |
Collapse
|
22
|
Sumida K, Han Z, Chiu CY, Mims TS, Bajwa A, Demmer RT, Datta S, Kovesdy CP, Pierre JF. Circulating Microbiota in Cardiometabolic Disease. Front Cell Infect Microbiol 2022; 12:892232. [PMID: 35592652 PMCID: PMC9110890 DOI: 10.3389/fcimb.2022.892232] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022] Open
Abstract
The rapid expansion of microbiota research has significantly advanced our understanding of the complex interactions between gut microbiota and cardiovascular, metabolic, and renal system regulation. Low-grade chronic inflammation has long been implicated as one of the key mechanisms underlying cardiometabolic disease risk and progression, even before the insights provided by gut microbiota research in the past decade. Microbial translocation into the bloodstream can occur via different routes, including through the oral and/or intestinal mucosa, and may contribute to chronic inflammation in cardiometabolic disease. Among several gut-derived products identifiable in the systemic circulation, bacterial endotoxins and metabolites have been extensively studied, however recent advances in microbial DNA sequencing have further allowed us to identify highly diverse communities of microorganisms in the bloodstream from an -omics standpoint, which is termed "circulating microbiota." While detecting microorganisms in the bloodstream was historically considered as an indication of infection, evidence on the circulating microbiota is continually accumulating in various patient populations without clinical signs of infection and even in otherwise healthy individuals. Moreover, both quantitative and compositional alterations of the circulating microbiota have recently been implicated in the pathogenesis of chronic inflammatory conditions, potentially through their immunostimulatory, atherogenic, and cardiotoxic properties. In this mini review, we aim to provide recent evidence on the characteristics and roles of circulating microbiota in several cardiometabolic diseases, such as type 2 diabetes, cardiovascular disease, and chronic kidney disease, with highlights of our emerging findings on circulating microbiota in patients with end-stage kidney disease undergoing hemodialysis.
Collapse
Affiliation(s)
- Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States,*Correspondence: Keiichi Sumida,
| | - Zhongji Han
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chi-Yang Chiu
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Tahliyah S. Mims
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| | - Amandeep Bajwa
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Csaba P. Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States,Nephrology Section, Memphis Veterans Affairs (VA) Medical Center, Memphis, TN, United States
| | - Joseph F. Pierre
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
23
|
Trends in the Prevalence of Cardiometabolic Multimorbidity in the United States, 1999-2018. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19084726. [PMID: 35457593 PMCID: PMC9027860 DOI: 10.3390/ijerph19084726] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/11/2022] [Indexed: 12/18/2022]
Abstract
Cardiometabolic multimorbidity (co-existence of ≥1 cardiometabolic diseases) is increasingly common, while its prevalence in the U.S. is unknown. We utilized data from 10 National Health and Nutrition Examination Survey (NHANES) two-year cycles in U.S. adults from 1999 to 2018. We reported the age-standardized prevalence of cardiometabolic multimorbidity in 2017-2018 and analyzed their trends during 1999-2018 with joinpoint regression models. Stratified analyses were performed according to gender, age, and race/ethnicity. In 2017-2018, the prevalence of cardiometabolic multimorbidity was 14.4% in the U.S., and it was higher among male, older, and non-Hispanic Black people. The three most common patterns were hypertension and diabetes (7.5%); hypertension, diabetes, and CHD (2.2%); and hypertension and CHD (1.8%). During 1999-2018, the prevalence of cardiometabolic multimorbidity in U.S. adults increased significantly, with an averaged two-year cycle percentage change (AAPC) of 3.6 (95% CI: 2.1 to 5.3). The increasing trend was significant for both genders, most age groups except for 60-79 years, and non-Hispanic White people. For common patterns, the trend was increasing for hypertension and diabetes and hypertension, diabetes, and CHD, while it was decreasing for hypertension and CHD. Our findings provide evidence that cardiometabolic multimorbidity has risen as an austere issue of public health in the U.S.
Collapse
|
24
|
Wang J, Yu C, Zhang Y, Huang Y. A Prediction Model for Acute Kidney Injury After Pericardiectomy: An Observational Study. Front Cardiovasc Med 2022; 9:790044. [PMID: 35224038 PMCID: PMC8873385 DOI: 10.3389/fcvm.2022.790044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/19/2022] [Indexed: 12/18/2022] Open
Abstract
Objectives Acute kidney injury is a common complication after pericardiectomy for constrictive pericarditis, which predisposes patients to worse outcomes and high medical costs. We aimed to investigate potential risk factors and consequences and establish a prediction model. Methods We selected patients with constrictive pericarditis receiving isolated pericardiectomy from January 2013 to January 2021. Patients receiving concomittant surgery or repeat percardiectomy, as well as end-stage of renal disease were excluded. Acute kidney injury was diagnosed and classified according to the KDIGO criteria. Clinical features were compared between patients with and without postoperative acute kidney injury. A prediction model was established based on multivariable regression analysis. Results Among two hundred and eleven patients, ninety-five (45.0%) developed postoperative acute kidney injury, with fourty-three (45.3%), twenty-eight (29.5%), and twenty-four (25.3%) in mild, moderate and severe stages, respectively. Twenty-nine (13.7%) patients received hemofiltration. Nine (4.3%) patients died perioperatively and were all in the acute kidney injury (9.5%) group. Eleven (5.2%) patients were considered to have chronic renal dysfunction states at the 6-month postoperative follow-up, and eight (72.7%) of them experienced moderate to severe stages of postoperative acute kidney injury. Univariable analysis showed that patients with acute kidney injury were older (difference 8 years, P < 0.001); had higher body mass index (difference 1.68 kg·m−2, P = 0.002); rates of smoking (OR = 2, P = 0.020), hypertension (OR = 2.83, P = 0.004), and renal dysfunction (OR = 3.58, P = 0.002); higher central venous pressure (difference 3 cm H2O, P < 0.001); and lower cardiac index (difference −0.23 L·min−1·m−2, P < 0.001) than patients without acute kidney injury. Multivariable regression analysis showed that advanced age (OR 1.03, P = 0.003), high body mass index (OR 1.10, P = 0.024), preoperative atrial arrhythmia (OR 3.12, P = 0.041), renal dysfunction (OR 2.70 P = 0.043), high central venous pressure (OR 1.12, P = 0.002), and low cardiac index (OR 0.36, P = 0.009) were associated with a high risk of postoperative acute kidney injury. A nomogram was established based on the regression results. The model showed good model fitness (Hosmer-Lemeshow test P = 0.881), with an area under the curve value of 0.78 (95% CI: 0.71, 0.84, P < 0.001). Conclusion The prediction model may help with early recognition, management, and reduction of acute kidney injury after pericardiectomy.
Collapse
Affiliation(s)
- Jin Wang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chunhua Yu
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Chunhua Yu
| | - Yuelun Zhang
- Department of Biostatistics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuguang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Guo J, Wu J, Wei D, Wang T, Hu Y, Lin Y, Chen M, Yang L, Wen Y, Cai Y, Xu X, Li H, Wu S, Xie X. Association between greenness and dyslipidemia in patients with coronary heart disease: A proteomic approach. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 231:113199. [PMID: 35042090 DOI: 10.1016/j.ecoenv.2022.113199] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/27/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Residential surrounding greenness may be protective of dyslipidemia are often theorized but remain poorly quantified. In particular, the underlying biological mechanisms of blood lipid changes with green spaces remain unclear. METHODS Our observational epidemiology study included a residentially stable sample of 1035 coronary heart disease patients, and proteomics study included 16 participants. Normalized Difference Vegetation Index (NDVI) was used to evaluate residential greenness exposures. Proteomics technology was used to identify plasma greenness-related proteome disturbance, and the pathway analysis was employed to evaluate the potential biological mechanisms of greenness decreasing dyslipidemia risk. RESULT Higher residential surrounding greenness in the 500-m area was associated with lower risks of dyslipidemia (odds ratio (OR) = 0.871, 95% confidence interval (CI): 0.763, 0.994 for per one-quartile NDVI increase). Lymphocytes mediated 18.7% of the association between greenness and dyslipidemia. Greenness related proteins (including PLXDC1, IGFBP2 and LY6D) may regulate the biological functions of lipid metabolism and transport-related proteins (including ADIPOQ and CES1) through a series of biological processes. CONCLUSION People in greener surroundings have a lower risk of dyslipidemia, which may be due to their lower inflammation, stronger lipid transporter activity, and normal cholesterol metabolism.
Collapse
Affiliation(s)
- Jianhui Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Jieyu Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Donghong Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Tinggui Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yuduan Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yawen Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Mingjun Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Le Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yeyin Wen
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yingying Cai
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xingyan Xu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Huanyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| | - Siying Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| | - Xiaoxu Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
26
|
Miura T, Kuno A, Tanaka M. Diabetes modulation of the myocardial infarction- acute kidney injury axis. Am J Physiol Heart Circ Physiol 2022; 322:H394-H405. [PMID: 35089809 DOI: 10.1152/ajpheart.00639.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since there is crosstalk in functions of the heart and kidney, acute or chronic injury in one of the two organs provokes adaptive and/or maladaptive responses in both organs, leading to cardiorenal syndrome (CRS). Acute kidney injury (AKI) induced by acute heart failure is referred to as type 1 CRS, and a frequent cause of this type of CRS is acute myocardial infarction (AMI). Diabetes mellitus increases the risk of AMI and also the risk of AKI of various causes. However, there have been only a few studies in which animal models of diabetes were used to examine how diabetes modulates AMI-induced AKI. In this review, we summarize findings regarding the mechanisms of type 1 CRS and the impact of diabetes on both AMI and renal susceptibility to AKI and we discuss mechanisms by which diabetes modulates AMI-induced AKI. Hemodynamic alterations induced by AMI could be augmented by diabetes via its detrimental effect on infarct size and contractile function of the non-infarcted region in the heart. Diabetes increases susceptibility of renal cells to hypoxia and oxidative stress by modulation of signaling pathways that regulate cell survival and autophagy. Recent studies have shown that diabetes mellitus even at early stage of cardiomyopathy/nephropathy predisposes the kidney to AMI-induced AKI, in which activation of toll-like receptors and reactive oxygen species derived from NADPH oxidases are involved. Further analysis of crosstalk between diabetic cardiomyopathy and diabetic kidney disease is necessary for obtaining a more comprehensive understanding of modulation of the AMI-AKI axis by diabetes.
Collapse
Affiliation(s)
- Tetsuji Miura
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Japan.,Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
27
|
Ahmed MM, Zaki A, Alhazmi A, Alsharif KF, Bagabir HA, Haque S, Manda K, Ahmad S, Ali SM, Ishrat R. Identification and Validation of Pathogenic Genes in Sepsis and Associated Diseases by Integrated Bioinformatics Approach. Genes (Basel) 2022; 13:genes13020209. [PMID: 35205254 PMCID: PMC8872348 DOI: 10.3390/genes13020209] [Citation(s) in RCA: 4] [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: 12/12/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Sepsis is a clinical syndrome with high mortality and morbidity rates. In sepsis, the abrupt release of cytokines by the innate immune system may cause multiorgan failure, leading to septic shock and associated complications. In the presence of a number of systemic disorders, such as sepsis, infections, diabetes, and systemic lupus erythematosus (SLE), cardiorenal syndrome (CRS) type 5 is defined by concomitant cardiac and renal dysfunctions Thus, our study suggests that certain mRNAs and unexplored pathways may pave a way to unravel critical therapeutic targets in three debilitating and interrelated illnesses, namely, sepsis, SLE, and CRS. Sepsis, SLE, and CRS are closely interrelated complex diseases likely sharing an overlapping pathogenesis caused by erroneous gene network activities. We sought to identify the shared gene networks and the key genes for sepsis, SLE, and CRS by completing an integrative analysis. Initially, 868 DEGs were identified in 16 GSE datasets. Based on degree centrality, 27 hub genes were revealed. The gProfiler webtool was used to perform functional annotations and enriched molecular pathway analyses. Finally, core hub genes (EGR1, MMP9, and CD44) were validated using RT-PCR analysis. Our comprehensive multiplex network approach to hub gene discovery is effective, as evidenced by the findings. This work provides a novel research path for a new research direction in multi-omics biological data analysis.
Collapse
Affiliation(s)
- Mohd Murshad Ahmed
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Almaz Zaki
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India; (A.Z.); (S.A.)
| | - Alaa Alhazmi
- Medical Laboratory Technology Department, SMIRES for Consultation in Specialized, Jazan University, Jazan 45142, Saudi Arabia;
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia;
| | - Hala Abubaker Bagabir
- Department of Medical Physiology, Faculty of Medicine, King Abdulaziz University, Rabigh 21589, Saudi Arabia;
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
| | - Kailash Manda
- Institute of Nuclear Medicine and Applied Sciences, Defense Research Development Organization, New Delhi 110054, India;
| | - Shaniya Ahmad
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India; (A.Z.); (S.A.)
| | - Syed Mansoor Ali
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India; (A.Z.); (S.A.)
- Correspondence: (S.M.A.); (R.I.)
| | - Romana Ishrat
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
- Correspondence: (S.M.A.); (R.I.)
| |
Collapse
|
28
|
Clark KC, Kwitek AE. Multi-Omic Approaches to Identify Genetic Factors in Metabolic Syndrome. Compr Physiol 2021; 12:3045-3084. [PMID: 34964118 PMCID: PMC9373910 DOI: 10.1002/cphy.c210010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metabolic syndrome (MetS) is a highly heritable disease and a major public health burden worldwide. MetS diagnosis criteria are met by the simultaneous presence of any three of the following: high triglycerides, low HDL/high LDL cholesterol, insulin resistance, hypertension, and central obesity. These diseases act synergistically in people suffering from MetS and dramatically increase risk of morbidity and mortality due to stroke and cardiovascular disease, as well as certain cancers. Each of these component features is itself a complex disease, as is MetS. As a genetically complex disease, genetic risk factors for MetS are numerous, but not very powerful individually, often requiring specific environmental stressors for the disease to manifest. When taken together, all sequence variants that contribute to MetS disease risk explain only a fraction of the heritable variance, suggesting additional, novel loci have yet to be discovered. In this article, we will give a brief overview on the genetic concepts needed to interpret genome-wide association studies (GWAS) and quantitative trait locus (QTL) data, summarize the state of the field of MetS physiological genomics, and to introduce tools and resources that can be used by the physiologist to integrate genomics into their own research on MetS and any of its component features. There is a wealth of phenotypic and molecular data in animal models and humans that can be leveraged as outlined in this article. Integrating these multi-omic QTL data for complex diseases such as MetS provides a means to unravel the pathways and mechanisms leading to complex disease and promise for novel treatments. © 2022 American Physiological Society. Compr Physiol 12:1-40, 2022.
Collapse
Affiliation(s)
- Karen C Clark
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Anne E Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
29
|
Trapecar M. Multiorgan microphysiological systems as tools to interrogate interorgan crosstalk and complex diseases. FEBS Lett 2021; 596:681-695. [PMID: 34923635 DOI: 10.1002/1873-3468.14260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
Metabolic and inflammatory disorders such as autoimmune and neurodegenerative diseases are increasing at alarming rates. Many of these are not tissue-specific occurrences but complex, systemic pathologies of unknown origin for which no cure exists. Such complexity obscures causal relationships among factors regulating disease progression. Emerging technologies mimicking human physiology, such as microphysiological systems (MPSs), offer new possibilities to provide clarity in systemic metabolic and inflammatory diseases. Controlled interaction of multiple MPSs and the scalability of biological complexity in MPSs, supported by continuous multiomic monitoring, might hold the key to identifying novel relationships between interorgan crosstalk, metabolism, and immunity. In this perspective, I aim to discuss the current state of modeling multiorgan physiology and evaluate current opportunities and challenges.
Collapse
Affiliation(s)
- Martin Trapecar
- Department of Medicine, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
30
|
Ishizuka S, Yamamoto M, Hirouchi H, Yotsuya M, Ohkubo M, Sato M, Abe S. Muscle-Bone Relationship in Temporomandibular Joint Disorders after Partial Discectomy. J Oral Biosci 2021; 63:436-443. [PMID: 34555528 DOI: 10.1016/j.job.2021.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Temporomandibular joint osteoarthritis (TMJ-OA) causes degenerative changes in TMJ tissues. The inter-tissue crosstalk that exacerbates illness and organic changes in bone secondary to TMJ-OA potentially affects the muscles; therefore, patients with a muscular disease might also suffer from bone disease. However, knowledge gaps exist concerning muscle pathology at the onset of TMJ-OA. In this study, we documented the pathogeneses of the bone and muscle at the onset of TMJ-OA using a mouse model. METHODS We performed a partial resection of the TMJ disk to establish a mouse model of TMJ-OA. After the onset of TMJ-OA, we performed various measurements at 8, 12, and 16 weeks post-surgery in the defined groups. RESULTS The volume of the mandibular head in the TMJ-OA group was significantly greater than that in the control group. The temporal muscles in the TMJ-OA group were significantly deformed compared with those in the control group; however, between-group comparisons did not reveal significant differences in the mandibular head or temporal muscles after surgery. Therefore, we hypothesized that the degree of mandibular head hypertrophy would alter the temporal muscles. A subsequent analysis of the correlation between the bone and muscle confirmed that the deformity of the temporal muscle increased with increasing hypertrophy of the mandibular head. Temporal and masseter muscle contact was observed in 25% of surgical groups. CONCLUSIONS This study demonstrates that TMJ-OA progressed when organic changes occurred in bones and muscles, supporting the symbiotic relationship between bones and muscles.
Collapse
Affiliation(s)
- Satoshi Ishizuka
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan; Tokyo Dental College Research Branding Project, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan.
| | - Masahito Yamamoto
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan; Tokyo Dental College Research Branding Project, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan.
| | - Hidetomo Hirouchi
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan.
| | - Mamoru Yotsuya
- Tokyo Dental College Research Branding Project, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan; Department of Fixed Prosthodontics, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan.
| | - Mai Ohkubo
- Tokyo Dental College Research Branding Project, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan; Department of Oral Health and Clinical Science, Division of Dysphagia Rehabilitation, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan.
| | - Masaki Sato
- Tokyo Dental College Research Branding Project, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan; Laboratory of Biology, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan.
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan; Tokyo Dental College Research Branding Project, 2-9-18 Kanda-misakicho, Tokyo, 101-0061, Japan.
| |
Collapse
|
31
|
Heidarpour M, Bashiri S, Vakhshoori M, Heshmat-Ghahdarijani K, Khanizadeh F, Ferdowsian S, Shafie D. The association between platelet-to-lymphocyte ratio with mortality among patients suffering from acute decompensated heart failure. BMC Cardiovasc Disord 2021; 21:454. [PMID: 34537010 PMCID: PMC8449504 DOI: 10.1186/s12872-021-02260-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 09/13/2021] [Indexed: 02/08/2023] Open
Abstract
Background Platelet-to-lymphocyte ratio (PLR) is an inflammation index suggested to have the prognostic capability in heart failure (HF). We sought to investigate the association of PLR with cardiovascular disease (CVD) mortality and creatinine (Cr) rise among Iranian individuals suffering from acute decompensated HF (ADHF). Methods This retrospective cohort study was in the context of the Persian Registry Of cardioVascular diseasE/Heart Failure (PROVE/HF) study. 405 individuals with ADHF admitted to the emergency department were recruited from April 2019 to March 2020. PLR was calculated by division of platelet to absolute lymphocyte counts and categorized based on quartiles. We utilized the Kaplan–Meier curve to show the difference in mortality based on PLR quartiles. Cr rise was defined as the increment of at least 0.3 mg/dl from baseline. Cox proportional hazard ratio (HR) was used to investigate the association of PLR with CVDs mortality. Results Mean age of participants was 65.9 ± 13.49 years (males: 67.7%). The mean follow-up duration was 4.26 ± 2.2 months. CVDs mortality or re-hospitalization was not significantly associated with PLR status. Multivariate analysis of PLR quartiles showed a minimally reduced likelihood of CVDs death in 2nd quartile versus the first one (HR 0.40, 95% confidence interval (CI) 0.16–1.01, P = 0.054). Cr rise had no remarkable relation with PLR status in neither model. Conclusion PLR could not be used as an independent prognostic factor among ADHF patients. Several studies are required clarifying the exact utility of this index.
Collapse
Affiliation(s)
- Maryam Heidarpour
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sepideh Bashiri
- Cardiac Rehabilitation Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrbod Vakhshoori
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiyan Heshmat-Ghahdarijani
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Shaghayegh Ferdowsian
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
32
|
The Vitamin D, IL-6 and the eGFR Markers a Possible Way to Elucidate the Lung-Heart-Kidney Cross-Talk in COVID-19 Disease: A Foregone Conclusion. Microorganisms 2021; 9:microorganisms9091903. [PMID: 34576798 PMCID: PMC8464828 DOI: 10.3390/microorganisms9091903] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Based on recent findings, we speculated the existence of the lung, heart, and kidney axis as the main pathway for the COVID-19 disease progression. Methods: This paper reports on an observational study conducted by a team of researchers and doctors of the 118-Pre-Hospital and Emergency Department of SG Moscati of Taranto City in Italy. The study was conducted on a totality of 185 participants that were divided into three groups. The study group included COVID-19 affected patients (PP n = 80), the first control group included patients with different pathologies (non-COVID-19 NNp n = 62) of the SG Moscati Hospital, and the second control group included healthy individuals (NNh n = 43). The core of the current trial was focused on assessing the level of the vitamin D (serum 25(OH) D concentration), IL-6, and the renal glomerular filtrate (eGFR) in COVID-19 disease and non-COVID-19 patients in both groups. Results: It was observed that the majority of COVID-19-infected patients showed a progressive multi-organ involvement, especially in regard to the lung, kidney, and heart. The majority of the COVID-19 patients exhibited preexisting comorbidities which include cardiovascular, respiratory, and renal disorders accompanied by a severely low level of vitamin D, extremely high level of IL-6, and low glomerular filtration rate (eGFR). The significant overall damages exerted by the immune-mediated responses under the hyper-expression of proinflammatory cytokines and interleukins, such as IL-6, may be facilitated by either a decreased level of vitamin D or the ageing process. The reduced presence of vitamin D was often found together with a reduced functionality of renal activity, as revealed by the low eGFR, and both were seen to be concomitant with an increased mortality risk in patients with lung disorders and heart failure (HF), whether it is showed at baseline or it develops during manifestation of COVID-19. Therefore, the documentation of the modifiable risk factors related to SARS-CoV-2 and lung impairment in older patients with kidney and heart disease may help the clinician to better manage the situation. Conclusions: This paper addresses how a low level of vitamin D and older age may be indicative of systemic worsening in patients with COVID-19, with a goal of providing a broader context in which to view a better therapeutic approach.
Collapse
|
33
|
Paul D, Komarova NL. Multi-scale network targeting: A holistic systems-biology approach to cancer treatment. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 165:72-79. [PMID: 34428429 DOI: 10.1016/j.pbiomolbio.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/15/2022]
Abstract
The vulnerabilities of cancer at the cellular and, recently, with the introduction of immunotherapy, at the tissue level, have been exploited with variable success. Evaluating the cancer system vulnerabilities at the organismic level through analysis of network topology and network dynamics can potentially predict novel anti-cancer drug targets directed at the macroscopic cancer networks. Theoretical work analyzing the properties and the vulnerabilities of the multi-scale network of cancer needs to go hand-in-hand with experimental research that uncovers the biological nature of the relevant networks and reveals new targetable vulnerabilities. It is our hope that attacking cancer on different spatial scales, in a concerted integrated approach, may present opportunities for novel ways to prevent treatment resistance.
Collapse
Affiliation(s)
- Doru Paul
- Medical Oncology, Weill Cornell Medicine, 1305 York Avenue 12th Floor, New York, NY, 10021, USA.
| | - Natalia L Komarova
- Department of Mathematics, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
34
|
Bo CRD, de Paula VP, Strazzi APWB, Wolosker N, Aloia TPA, Mazzeo A, Kaufmann OG. Effect of unilateral renal ischemia on the contralateral kidney assessed by Caspase 3 expression. J Vasc Bras 2021; 20:e20210040. [PMID: 34349794 PMCID: PMC8294808 DOI: 10.1590/1677-5449.210040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/14/2021] [Indexed: 11/22/2022] Open
Abstract
Background Studies have demonstrated with histological analysis and Doppler flow measurement analysis that unilateral renal ischemia, which is performed in some surgeries, interfered with the contralateral kidney, identifying the phenomenon of kidney-kidney crosstalk. Objectives To identify the effects on the ischemic and contralateral kidney of renal ischemia induced by two types of clamping technique by analyzing the volume of kidney cells positive for Caspase 3. Methods Sixteen pigs were divided into 2 groups, as follows: A (n = 8) – clamping of left renal artery only and AV (n = 8) – clamping of left renal artery and vein. Immunohistochemical analyses (anti Caspase 3) were conducted with biopsy specimens collected from the ischemic and contralateral kidney at 0, 30, 60, and 90 minutes of ischemia and morphometric analysis was performed, taking the mean to represent the volume of the Caspase 3 positive area (%). Results Morphometric analysis of specimens collected at 30, 60, and 90 minutes of ischemia showed that the mean area marked for Caspase 3 was statistically larger in the contralateral kidney than the ischemic kidney in both groups: clamped renal artery (A) and clamped renal artery and vein (AV). Comparing the ischemic and contralateral kidney, there was no statistically significant difference in the area marked for Caspase 3 between the two types of clamping. Conclusions In the experimental model of unilateral renal ischemia, the non-ischemic kidney exhibited cell damage, demonstrated by Caspase 3 expression. The type of hilum clamping does not appear to influence the area marked for Caspase 3.
Collapse
Affiliation(s)
- Carolina Rodrigues Dal Bo
- Faculdade Israelita de Ciências da Saúde Albert Einstein - FICSAE, São Paulo, SP, Brasil.,Hospital Israelita Albert Einstein - HIAE, São Paulo, SP, Brasil
| | - Vitória Penido de Paula
- Faculdade Israelita de Ciências da Saúde Albert Einstein - FICSAE, São Paulo, SP, Brasil.,Hospital Israelita Albert Einstein - HIAE, São Paulo, SP, Brasil
| | | | - Nelson Wolosker
- Faculdade Israelita de Ciências da Saúde Albert Einstein - FICSAE, São Paulo, SP, Brasil.,Hospital Israelita Albert Einstein - HIAE, São Paulo, SP, Brasil.,Faculdade de Medicina da Universidade de São Paulo - USP, São Paulo, SP, Brasil
| | - Thiago Pinheiro Arrais Aloia
- Instituto de Ensino e Pesquisa - IIEP, Hospital Albert Einstein, São Paulo, SP, Brasil.,Faculdade de Medicina da Universidade de São Paulo - USP, São Paulo, SP, Brasil
| | - Angela Mazzeo
- Instituto de Ensino e Pesquisa - IIEP, Hospital Albert Einstein, São Paulo, SP, Brasil
| | - Oskar Grau Kaufmann
- Instituto de Ensino e Pesquisa - IIEP, Hospital Albert Einstein, São Paulo, SP, Brasil.,Faculdade de Medicina da Universidade de São Paulo - USP, São Paulo, SP, Brasil
| |
Collapse
|
35
|
Fujiu K, Manabe I. Nerve-macrophage interactions in cardiovascular disease. Int Immunol 2021; 34:81-95. [PMID: 34173833 DOI: 10.1093/intimm/dxab036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/25/2021] [Indexed: 01/09/2023] Open
Abstract
The heart is highly innervated by autonomic neurons, and dynamic autonomic regulation of the heart and blood vessels is essential for animals to carry out the normal activities of life. Cardiovascular diseases, including heart failure and myocardial infarction, are often characterized in part by an imbalance in autonomic nervous system activation, with excess sympathetic and diminished parasympathetic activation. Notably, however, this is often accompanied by chronic inflammation within the cardiovascular tissues, which suggests there are interactions between autonomic dysregulation and inflammation. Recent studies have been unraveling the mechanistic links between autonomic nerves and immune cells within cardiovascular disease. The autonomic nervous system and immune system also act in concert to coordinate the actions of multiple organs that not only maintain homeostasis but also likely play key roles in disease-disease interactions, such as cardiorenal syndrome and multimorbidity. In this review, we summarize the physiological and pathological interactions between autonomic nerves and macrophages in the context of cardiovascular disease.
Collapse
Affiliation(s)
- Katsuhito Fujiu
- Department of Cardiovascular Medicine, the University of Tokyo, Hongo, Bunkyo, Tokyo, Japan.,Department of Advanced Cardiology, the University of Tokyo, Hongo, Bunkyo, Tokyo, Japan
| | - Ichiro Manabe
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Inohana, Chuo, Chiba, Chiba, Japan
| |
Collapse
|
36
|
Boudoulas KD, Triposkiadis F, Boudoulas H. Aortic stenosis: a complex entity with multiple coexistent abnormalities. Eur Heart J Cardiovasc Imaging 2021; 22:983-985. [PMID: 34059904 DOI: 10.1093/ehjci/jeab079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | | | - Harisios Boudoulas
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio, USA.,Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
37
|
Kadowaki T, Yamamoto F, Taneda Y, Naito Y, Clark D, Lund SS, Okamura T, Kaku K. Effects of anti-diabetes medications on cardiovascular and kidney outcomes in Asian patients with type 2 diabetes: a rapid evidence assessment and narrative synthesis. Expert Opin Drug Saf 2021; 20:707-720. [PMID: 33706621 DOI: 10.1080/14740338.2021.1898585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The cardiovascular and kidney safety of glucose-lowering drugs is a key concern in type 2 diabetes (T2D). We evaluated cardiorenal outcomes with glucose-lowering drugs in Asian patients, who comprise over half of T2D cases globally. RESEARCH DESIGN AND METHODS A rapid evidence assessment was conducted for phase III or IV, double-blind, randomized clinical trials of glucose-lowering drugs reporting cardiovascular or kidney outcomes for Asian T2D patients (Embase, Medline, Cochrane Library databases: 1 January 2008-14 June 2020). RESULTS Fifty-four publications reported exploratory data for Asians from 18 trials of dipeptidyl peptidase-4 (DPP-4) inhibitors, sodium-glucose co-transporter-2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, and insulin analogs. SGLT2 inhibitors and several GLP-1 receptor agonists were associated with reduced cardiovascular risk in Asian T2D patients, while DPP-4 inhibitors exhibited cardiovascular safety. SGLT2 inhibitors also appeared to reduce renal risk; however, kidney outcomes were lacking for DPP-4 inhibitors other than linagliptin and GLP-1 receptor agonists in Asian patients. Insulin data were inconclusive as the only trial conducted used different types of insulin as both treatment and comparator. CONCLUSIONS Cardiorenal outcomes with glucose-lowering drugs in Asian T2D patients were similar to outcomes in the overall multinational cohorts of these trials. DPP-4 inhibitors appear to demonstrate cardiovascular safety in Asians, while SGLT2 inhibitors and some GLP-1 receptor agonists may reduce cardiorenal and cardiovascular risk, respectively.
Collapse
Affiliation(s)
| | - Fumiko Yamamoto
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd, Tokyo, Japan
| | - Yusuke Taneda
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd, Tokyo, Japan
| | - Yusuke Naito
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd, Tokyo, Japan
| | - Douglas Clark
- TA CardioMetabolism Respiratory Med, Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Søren S Lund
- TA CardioMetabolism Respiratory Med, Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Tomoo Okamura
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd, Tokyo, Japan
| | - Kohei Kaku
- General Internal Medicine, Kawasaki Medical School, Okayama, Japan.,Faculty of Health and Welfare Services Management, Kawasaki University of Medical Welfare, Okayama, Japan
| |
Collapse
|
38
|
Arocha Rodulfo JI. Approach to the cardiometabolic continuum. Narrative description. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2020; 33:158-167. [PMID: 33309070 DOI: 10.1016/j.arteri.2020.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 10/22/2022]
Abstract
Atherosclerotic cardiovascular disease (ACVD) is the major adverse outcome in the evolution of several metabolic conditions. For around several decades, the cardiovascular continuum has been used as a fantastic tool to explain the evolution of ACVD from the onset of risk factor, to clinical outcomes, and to death. Nowadays, metabolic diseases such as obesity, prediabetes, and type2 diabetes have been increasing enough to become serious public health problems and notorious contributors to the morbidity and mortality rates due to ACVD, including arterial hypertension. Other conditions seem to increase the list, such as: physical inactivity with its metabolic cluster, sarcopenia, and non-alcoholic fatty liver disease (NAFLD). Moreover, obesity in childhood has been growing at an exponential rate so the excess of adiposity in children and adolescents will translate into an excess of cardiometabolic risk in adults. Several longitudinal studies confirm the strong association of paediatric obesity with the persistence of adult obesity, as well as the future development of cardiometabolic conditions, such as prediabetes, diabetes, obesity, increased risk of arterial hypertension, and ACVD. Therefore, it is time to conceptualise the cardiometabolic continuum as a tool of an early, wider and preventive intervention in order to reduce the morbidity and mortality due to ACVD.
Collapse
Affiliation(s)
- J Ildefonzo Arocha Rodulfo
- Sociedad Venezolana de Cardiología, Fundación Venezolana de Cardiología Preventiva, Chacao, Caracas, Venezuela.
| |
Collapse
|
39
|
Atherosclerosis Imaging with 18F-Sodium Fluoride PET. Diagnostics (Basel) 2020; 10:diagnostics10100852. [PMID: 33092250 PMCID: PMC7590213 DOI: 10.3390/diagnostics10100852] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/15/2020] [Indexed: 12/21/2022] Open
Abstract
The evidence on atherosclerosis imaging with 18F-sodium-fluoride (NaF) positron emission tomography (PET) is hotly debated because of the different patient characteristics, methodology, vascular beds, etc. in reported studies. This review is a continuation of a previous review on this topic, which covered the period 2010-2018. The purpose was to examine whether some of the most important questions that the previous review had left open had been elucidated by the most recent literature. Using principles of a systematic review, we ended analyzing 25 articles dealing with the carotids, coronary arteries, aorta, femoral, intracranial, renal, and penile arteries. The knowledge thus far can be summarized as follows: by targeting active arterial microcalcification, NaF uptake is considered a marker of early stage atherosclerosis, is age-dependent, and consistently associated with cardiovascular risk. Longitudinal studies on NaF uptake, conducted in the abdominal aorta only, showed unchanged uptake in postmenopausal women for nearly four years and varying uptake in prostate cancer patients over 1.5 years, despite constant or increasing calcium volume detected by computed tomography (CT). Thus, uncertainty remains about the transition from active arterial wall calcification marked by increased NaF uptake to less active or consolidated calcification detected by CT. The question of whether early-phase atherosclerosis and calcification can be modified remains also unanswered due to lack of intervention studies.
Collapse
|
40
|
Wholegrain Consumption and Risk Factors for Cardiorenal Metabolic Diseases in Chile: A Cross-Sectional Analysis of 2016-2017 Health National Survey. Nutrients 2020; 12:nu12092815. [PMID: 32937937 PMCID: PMC7576471 DOI: 10.3390/nu12092815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022] Open
Abstract
Wholegrain (WG) consumption has been associated with reduced risk factors for cardiorenal metabolic diseases (CRMD). In Latin-America. WG intake is low and scarce studies on this subject have been found. We aimed to evaluate the association between WG consumption and risk factors for CRMD in the 2016-2017 Chilean-National Health Survey. This cross-sectional study included 3110 participants representative of a total population of 11,810,647 subjects > 18 y, not taking insulin and with complete data on CRMD risk factors. Outcomes were metabolic syndrome and its components, albuminuria, and impaired glomerular filtration rate (GFR). WG consumption was categorized as regular (≥every two days), sporadic (≥once a month), and non-consumers. Associations were analyzed by multivariable logistic regressions adjusted for confounders taking into account the complex sample design of the survey. Regular WG consumers showed a lower risk of high blood pressure (OR: 0.61, 95%CI: 0.41-0.91) compared to non-consumers in fully-adjusted models. Although inverse associations were noticed with other metabolic syndrome components and impaired GFR, none was statistically significant. The association between WG and BP remained robust in the sensitivity analysis. In conclusion regular WG consumption was associated with a 39% lower risk of high blood pressure in Chilean adults.
Collapse
|