1
|
Zhao Y, Zhong K, Zheng Y, Xia X, Lin X, Kowark A, Wang X, Zhang D, Duan X. Postoperative delirium risk in patients with hyperlipidemia: A prospective cohort study. J Clin Anesth 2024; 98:111573. [PMID: 39094442 DOI: 10.1016/j.jclinane.2024.111573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
STUDY OBJECTIVE Hyperlipidemia and postoperative delirium (POD) significantly affect patients' quality of life; however, the question of whether hyperlipidemia constitutes a risk factor for POD remain unclear. This study aimed to investigate whether patients with hyperlipidemia face elevated risks of developing POD and to identify potential causes for this increased risk. DESIGN A prospective cohort study. SETTING Operating room. PATIENTS Patients were adults scheduled for colorectal cancer surgery in 2023. EXPOSURES The exposure factor was hyperlipidemia, and the patients were divided into hyperlipidemia group and non-hyperlipidemia group. MEASUREMENTS POD occurrence within three days post-surgery was assessed using the 3-Minute Diagnostic Interview for Confusion Assessment Method. Over one year, these patients were monitored through telephone to evaluate their survival and cognitive function. Logistic regression analysis was performed to evaluate the risk factors for POD development in patients with hyperlipidemia and to construct a clinical prediction model. MAIN RESULTS This study included 555 patients. POD incidence was 21.6% in the hyperlipidemia group and 12.7% in the non-hyperlipidemia group. One year following surgery, patients with hyperlipidemia and POD exhibited significantly higher rates of mortality and cognitive decline than did those without POD (p < 0.001). A multifactorial logistic clinical prediction model was constructed from seven independent risk factors for POD development in patients with hyperlipidemia, including education, preoperative total cholesterol (TC), preoperative triglyceride (TG), diet, history of hypertension, Sedation-Agitation Scale, and postoperative trimethylamine N-oxide expression level, and it had the highest predictive value for POD development in patients with hyperlipidemia. CONCLUSIONS Compared with those without hyperlipidemia, patients with hyperlipidemia had higher POD incidence. Elevated serum TC and TG levels are independent risk factors for POD in patients with hyperlipidemia. The study's findings could help develop strategies for improving POD and hyperlipidemia treatment.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China
| | - Ke Zhong
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China
| | - Yujie Zheng
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China
| | - Xiaoli Xia
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China
| | - Xue Lin
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China
| | - Ana Kowark
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Xiaobin Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China.
| | - Daiying Zhang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China.
| | - Xiaoxia Duan
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; Operating room, The Affiliated Hospital, Southwest Medical University,Luzhou, Sichuan Province 646000, China; School of Nursing & Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University,Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
2
|
Lin L, Xiang S, Chen Y, Liu Y, Shen D, Yu X, Wu Z, Sun Y, Chen K, Luo J, Wei G, Wang Z, Ning Z. Gut microbiota: Implications in pathogenesis and therapy to cardiovascular disease (Review). Exp Ther Med 2024; 28:427. [PMID: 39301250 PMCID: PMC11411594 DOI: 10.3892/etm.2024.12716] [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: 06/13/2023] [Accepted: 06/03/2024] [Indexed: 09/22/2024] Open
Abstract
The gut microbiota refers to the diverse bacterial community residing in the gastrointestinal tract. Recent data indicate a strong correlation between alterations in the gut microbiota composition and the onset of various diseases, notably cardiovascular disorders. Evidence suggests the gut-cardiovascular axis signaling molecules released by the gut microbiota play a pivotal role in regulation. This review systematically delineates the association between dysbiosis of the gut microbiota and prevalent cardiovascular diseases, including atherosclerosis, hypertension, myocardial infarction and heart failure. Furthermore, it provides an overview of the putative pathogenic mechanisms by which dysbiosis in the gut microbiota contributes to the progression of cardiovascular ailments. The potential modulation of gut microbiota as a preventive strategy against cardiovascular diseases through dietary interventions, antibiotic therapies and probiotic supplementation is also explored and discussed within the present study.
Collapse
Affiliation(s)
- Li Lin
- Department of Biochemistry, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Shaowei Xiang
- Department of Neurosurgery, Enshi State Central Hospital, Enshi, Hubei 445000, P.R. China
| | - Yuan Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yan Liu
- Department of Internal Medicine, The Second Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dingwen Shen
- Department of Parasitology, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xiaoping Yu
- Department of Function, The Second Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhe Wu
- Department of Histology and Embryology, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yanling Sun
- Department of Histology and Embryology, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Kequan Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Jia Luo
- School of Sport, Xianning Vocational and Technical College, Xianning, Hubei 437100, P.R. China
| | - Guilai Wei
- School of Art and Design, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhiguo Wang
- Department of Dermatology, The First Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhifeng Ning
- Department of Human Anatomy, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
3
|
Loh YH, Lv J, Goh Y, Sun X, Zhu X, Muheyati M, Luan Y. Remodeling of T-Tubules and Associated Calcium Handling Dysfunction in Heart Failure: Mechanisms and Therapeutic Insights. Can J Cardiol 2024:S0828-282X(24)01035-3. [PMID: 39455023 DOI: 10.1016/j.cjca.2024.10.017] [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: 03/20/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
In cardiomyocytes, transverse tubules (T-tubules) are sarcolemmal invaginations that facilitate excitation-contraction coupling (ECC) and diastolic function. The clinical significance of T-tubules has become evident as their remodeling is recognized as a hallmark feature of heart failure (HF) and a key contributor to disrupted Ca2+ homeostasis, compromised cardiac function, and arrhythmogenesis. Further investigations have revealed that T-tubule remodeling is particularly pronounced in HF with reduced ejection fraction (HFrEF), but not in HF with preserved ejection fraction (HFpEF), implying that T-tubule remodeling may play a crucial pathophysiological role in HFrEF. While research on the functional importance of T-tubules is ongoing due to their complexity, T-tubule remodeling has been found to be reversible. Such finding has triggered a surge in studies aimed at identifying specific therapeutic approaches for HFrEF. This review discusses the functional importance of T-tubules and their microdomains, the pathophysiology of T-tubule remodeling, and the potential mechanisms of current HFrEF therapeutic approaches in reversing T-tubule alterations. We also highlight discrepancies regarding the roles of T-tubule proteins in the recovery process across studies to offer valuable insights for future research.
Collapse
Affiliation(s)
- Yi Hao Loh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Jingyi Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yenfang Goh
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xiangjie Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Xianfeng Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Critical Care Medicine, Hangzhou Ninth People's Hospital, China
| | - Muergen Muheyati
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; School of Medicine, Shaoxing University, China.
| |
Collapse
|
4
|
Sasidharan Pillai S, Gagnon CA, Foster C, Ashraf AP. Exploring the Gut Microbiota: Key Insights Into Its Role in Obesity, Metabolic Syndrome, and Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:2709-2719. [PMID: 39040013 PMCID: PMC11479700 DOI: 10.1210/clinem/dgae499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/22/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
The gut microbiota (GM), comprising trillions of microorganisms in the gastrointestinal tract, is a key player in the development of obesity and related metabolic disorders, such as type 2 diabetes (T2D), metabolic syndrome (MS), and cardiovascular diseases. This mini-review delves into the intricate roles and mechanisms of the GM in these conditions, offering insights into potential therapeutic strategies targeting the microbiota. The review elucidates the diversity and development of the human GM, highlighting its pivotal functions in host physiology, including nutrient absorption, immune regulation, and energy metabolism. Studies show that GM dysbiosis is linked to increased energy extraction, altered metabolic pathways, and inflammation, contributing to obesity, MS, and T2D. The interplay between dietary habits and GM composition is explored, underscoring the influence of diet on microbial diversity and metabolic functions. Additionally, the review addresses the impact of common medications and therapeutic interventions like fecal microbiota transplantation on GM composition. The evidence so far advocates for further research to delineate the therapeutic potential of GM modulation in mitigating obesity and metabolic diseases, emphasizing the necessity of clinical trials to establish effective and sustainable treatment protocols.
Collapse
Affiliation(s)
- Sabitha Sasidharan Pillai
- Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Charles A Gagnon
- University of Alabama at Birmingham Marnix E. Heersink School of Medicine, Birmingham, AL 35294, USA
| | - Christy Foster
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ambika P Ashraf
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
5
|
Islam MM, Mahbub NU, Hong ST, Chung HJ. Gut bacteria: an etiological agent in human pathological conditions. Front Cell Infect Microbiol 2024; 14:1291148. [PMID: 39439902 PMCID: PMC11493637 DOI: 10.3389/fcimb.2024.1291148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 08/12/2024] [Indexed: 10/25/2024] Open
Abstract
Through complex interactions with the host's immune and physiological systems, gut bacteria play a critical role as etiological agents in a variety of human diseases, having an impact that extends beyond their mere presence and affects the onset, progression, and severity of the disease. Gaining a comprehensive understanding of these microbial interactions is crucial to improving our understanding of disease pathogenesis and creating tailored treatment methods. Correcting microbial imbalances may open new avenues for disease prevention and treatment approaches, according to preliminary data. The gut microbiota exerts an integral part in the pathogenesis of numerous health conditions, including metabolic, neurological, renal, cardiovascular, and gastrointestinal problems as well as COVID-19, according to recent studies. The crucial significance of the microbiome in disease pathogenesis is highlighted by this role, which is comparable to that of hereditary variables. This review investigates the etiological contributions of the gut microbiome to human diseases, its interactions with the host, and the development of prospective therapeutic approaches. To fully harness the benefits of gut microbiome dynamics for improving human health, future research should address existing methodological challenges and deepen our knowledge of microbial interactions.
Collapse
Affiliation(s)
- Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
6
|
Sun Y, Lin X, Liu Z, Hu L, Sun P, Shen G, Fan F, Zhang Y, Li J. Association between plasma trimethylamine N-oxide and coronary heart disease: new insights on sex and age differences. Front Cardiovasc Med 2024; 11:1397023. [PMID: 39434851 PMCID: PMC11491342 DOI: 10.3389/fcvm.2024.1397023] [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: 03/06/2024] [Accepted: 09/18/2024] [Indexed: 10/23/2024] Open
Abstract
Aim Elevated plasma trimethylamine N-oxide (TMAO) is related to atherosclerosis. Whether the relationship of TMAO and coronary heart disease (CHD) is influenced by sex or age is uncertain. We aim to explore the sex and age differences in the relationship between plasma TMAO and CHD risk and severity. Methods A case-control study was conducted in patients undergoing elective coronary angiography. Matched by sex, age (±2 years), and operation date (±180 days), a total of 429 CHD case-control pairs were included. Plasma TMAO was quantified using liquid chromatography-tandem mass spectrometry. Logistic regression analyses were performed to evaluate the association between plasma TMAO and CHD risk and severity. Results The overall median (interquartile range) plasma TMAO level was 0.11 (0.06-0.18) μg/ml. After stratification by sex and age, and adjustment for common CHD risk factors, the association between TMAO and CHD risk was significant in the older (≥65 years) male subgroup [odds ratios (OR) = 1.57, 95% confidence interval (CI): 1.09-2.28, P = 0.016], but not in other sex-age subgroups (all P > 0.05). The relationship of plasma TMAO and CHD risk was modified by age (adjusted P interaction = 0.001) in male individuals. Plasma TMAO was also associated with a higher risk of multi-vessel disease in male patients with CHD (OR = 1.65, 95% CI: 1.18-2.32, P = 0.004), but not in females. Conclusions Plasma TMAO is significantly positively associated with the risk and severity of CHD in Chinese men. Age has an interactive effect on the relationship between plasma TMAO and CHD risk in men. Our findings warrant further investigation.
Collapse
Affiliation(s)
- Yangyang Sun
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Xipeng Lin
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Zhihao Liu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Lihua Hu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Pengfei Sun
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Geng Shen
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Fangfang Fan
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University, Beijing, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University, Beijing, China
| |
Collapse
|
7
|
Zheng H, Sechi LA, Navarese EP, Casu G, Vidili G. Metabolic dysfunction-associated steatotic liver disease and cardiovascular risk: a comprehensive review. Cardiovasc Diabetol 2024; 23:346. [PMID: 39342178 PMCID: PMC11439309 DOI: 10.1186/s12933-024-02434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed nonalcoholic fatty liver disease (NAFLD), poses a significant global health challenge due to its increasing prevalence and strong association with cardiovascular disease (CVD). This comprehensive review summarizes the current knowledge on the MASLD-CVD relationship, compares analysis of how different terminologies for fatty liver disease affect cardiovascular (CV) risk assessment using different diagnostic criteria, explores the pathophysiological mechanisms connecting MASLD to CVD, the influence of MASLD on traditional CV risk factors, the role of noninvasive imaging techniques and biomarkers in the assessment of CV risk in patients with MASLD, and the implications for clinical management and prevention strategies. By incorporating current research and clinical guidelines, this review provides a comprehensive overview of the complex interplay between MASLD and cardiovascular health.
Collapse
Affiliation(s)
- Haixiang Zheng
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, 515041, Shantou, China
| | - Leonardo Antonio Sechi
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Complex Structure of Microbiology and Virology, AOU Sassari, 07100, Sassari, Italy
| | - Eliano Pio Navarese
- Clinical and Experimental Cardiology, Clinical and Interventional Cardiology, University of Sassari, Sassari, Italy
| | - Gavino Casu
- Clinical and Experimental Cardiology, Clinical and Interventional Cardiology, University of Sassari, Sassari, Italy
| | - Gianpaolo Vidili
- Department of Medicine, Surgery, and Pharmacy, University of Sassari, Azienda Ospedaliero, 07100, Sassari, Italy.
| |
Collapse
|
8
|
Cuervo L, McAlpine PL, Olano C, Fernández J, Lombó F. Low-Molecular-Weight Compounds Produced by the Intestinal Microbiota and Cardiovascular Disease. Int J Mol Sci 2024; 25:10397. [PMID: 39408727 PMCID: PMC11477366 DOI: 10.3390/ijms251910397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is the main cause of mortality in industrialized countries, with over 500 million people affected worldwide. In this work, the roles of low-molecular-weight metabolites originating from the gut microbiome, such as short-chain fatty acids, hydrogen sulfide, trimethylamine, phenylacetic acid, secondary bile acids, indoles, different gases, neurotransmitters, vitamins, and complex lipids, are discussed in relation to their CVD-promoting or preventing activities. Molecules of mixed microbial and human hepatic origin, such as trimethylamine N-oxide and phenylacetylglutamine, are also presented. Finally, dietary agents with cardioprotective effects, such as probiotics, prebiotics, mono- and poly-unsaturated fatty acids, carotenoids, and polyphenols, are also discussed. A special emphasis is given to their gut microbiota-modulating properties.
Collapse
Affiliation(s)
- Lorena Cuervo
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Patrick L. McAlpine
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos Olano
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Javier Fernández
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Felipe Lombó
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
9
|
Bano N, Khan S, Ahamad S, Kanshana JS, Dar NJ, Khan S, Nazir A, Bhat SA. Microglia and gut microbiota: A double-edged sword in Alzheimer's disease. Ageing Res Rev 2024; 101:102515. [PMID: 39321881 DOI: 10.1016/j.arr.2024.102515] [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: 04/23/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
The strong association between gut microbiota (GM) and brain functions such as mood, behaviour, and cognition has been well documented. Gut-brain axis is a unique bidirectional communication system between the gut and brain, in which gut microbes play essential role in maintaining various molecular and cellular processes. GM interacts with the brain through various pathways and processes including, metabolites, vagus nerve, HPA axis, endocrine system, and immune system to maintain brain homeostasis. GM dysbiosis, or an imbalance in GM, is associated with several neurological disorders, including anxiety, depression, and Alzheimer's disease (AD). Conversely, AD is sustained by microglia-mediated neuroinflammation and neurodegeneration. Further, GM and their products also affect microglia-mediated neuroinflammation and neurodegeneration. Despite the evidence connecting GM dysbiosis and AD progression, the involvement of GM in modulating microglia-mediated neuroinflammation in AD remains elusive. Importantly, deciphering the mechanism/s by which GM regulates microglia-dependent neuroinflammation may be helpful in devising potential therapeutic strategies to mitigate AD. Herein, we review the current evidence regarding the involvement of GM dysbiosis in microglia activation and neuroinflammation in AD. We also discuss the possible mechanisms through which GM influences the functioning of microglia and its implications for therapeutic intervention. Further, we explore the potential of microbiota-targeted interventions, such as prebiotics, probiotics, faecal microbiota transplantation, etc., as a novel therapeutic strategy to mitigate neuroinflammation and AD progression. By understanding and exploring the gut-brain axis, we aspire to revolutionize the treatment of neurodegenerative disorders, many of which share a common theme of microglia-mediated neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Jitendra Singh Kanshana
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburg, PA, USA.
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA.
| | - Sumbul Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
10
|
Lee YH, Lee JH, Jeon SM, Park IK, Jang HB, Kim SA, Park SD, Shim JJ, Hong SS, Lee JH. The Effect of Organic Vegetable Mixed Juice on Blood Circulation and Intestine Flora: Randomized, Double-Blinded, Placebo-Controlled Clinical Trial. Diseases 2024; 12:223. [PMID: 39329892 PMCID: PMC11431145 DOI: 10.3390/diseases12090223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Epidemiological evidence suggests that fruit and vegetable intake significantly positively affects cardiovascular health. Since vegetable juice is more accessible than raw vegetables, it attracts attention as a health functional food for circulatory diseases. Therefore, this study measured blood lipids, antioxidants, blood circulation indicators, and changes in the microbiome to confirm the effect of organic vegetable mixed juice (OVJ) on improving blood circulation. This 4-week, randomized, double-blinded, placebo-controlled study involved adult men and women with borderline total cholesterol (TC) and low-density lipoprotein (LDL) levels. As a result, blood lipid profile indicators, such as TC, triglycerides, LDL cholesterol, and apolipoprotein B, decreased (p < 0.05) in the OVJ group compared with those in the placebo group. Additionally, the antioxidant biomarker superoxide dismutase increased (p < 0.05). In contrast, systolic and diastolic blood viscosities, as blood circulation-related biomarkers, decreased (p < 0.05) in the OVJ group compared with those in the placebo group. After the intervention, a fecal microbiome analysis confirmed differences due to changes in the intestinal microbiome composition between the OVJ and placebo groups. In conclusion, our research results confirmed that consuming OVJ improves blood circulation by affecting the blood lipid profile, antioxidant enzymes, and microbiome changes.
Collapse
Affiliation(s)
- Yun-Ha Lee
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Jae-Ho Lee
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Soo-Min Jeon
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Il-Kyu Park
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Hyun-Bin Jang
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Soo-A Kim
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Soo-Dong Park
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Jae-Jung Shim
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| | - Seong-Soo Hong
- Department of Gastroenterology, Vievis Namuh Hospital, 627, Nonhyeon-ro, Gangnam-gu, Seoul 06117, Republic of Korea
| | - Jae-Hwan Lee
- R&BD Center, Hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (Y.-H.L.); (J.-H.L.); (S.-M.J.); (I.-K.P.); (H.-B.J.); (S.-A.K.); (S.-D.P.); (J.-J.S.)
| |
Collapse
|
11
|
Murugan G, Kothandan G, Padmanaban R. Anticipatory in silico vaccine designing based on specific antigenic epitopes from Streptococcus mutans against diabetic pathogenesis. In Silico Pharmacol 2024; 12:86. [PMID: 39310673 PMCID: PMC11411028 DOI: 10.1007/s40203-024-00260-x] [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: 02/20/2024] [Accepted: 09/08/2024] [Indexed: 09/25/2024] Open
Abstract
The metabolic disorder Type 2 Diabetes Mellitus (T2DM) is characterized by hyperglycaemia, causing increased mortality and healthcare burden globally. Recent studies emphasize the impact of metabolites in the gut microbiome on T2DM pathogenesis. One such microbial metabolite, imidazole propionate (Imp) derived from histidine metabolism, is shown to interfere with insulin signalling and other key metabolic processes. The key enzyme urocanate reductase (UrdA) is involved in ImP production. Hence, we propose to develop a novel therapeutic vaccine against the gut microbe producing Imp based on UrdA as a target for treating T2DM using immunoinformatics approach. Antigenic, non-allergic, non-toxic, and immunogenic B cell and T cell potential epitopes were predicted using immunoinformatics servers and tools. These epitopes were adjoined using linker sequences, and to increase immunogenicity, adjuvants were added at the N-terminal end of the final vaccine construct. Further, to confirm the vaccine's safety, antigenic and non-allergic characteristics of the developed vaccine construct were assessed. The tertiary structure of the UrdA vaccine sequence was predicted using molecular modelling tools. A molecular docking study was utilized to understand the vaccine construct interaction with immune receptors, followed by molecular dynamics simulation and binding free energy calculations to assess stability of the complex. In silico cloning techniques were employed to evaluate the expression and translation effectiveness of the developed vaccine in pET vector. In conclusion, this study developed an in silico epitope-based vaccine construct as a novel adjunct therapeutic for T2DM. Graphical Abstract
Collapse
Affiliation(s)
- Gopinath Murugan
- Immunodynamics and Interface Laboratory, Centre for Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu 600025 India
| | - Gugan Kothandan
- Biopolymer Modeling Laboratory, Centre for Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, India
| | - Rajashree Padmanaban
- Immunodynamics and Interface Laboratory, Centre for Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu 600025 India
| |
Collapse
|
12
|
Du J, Zhao X, Ding X, Han Q, Duan Y, Ren Q, Wang H, Song C, Wang X, Zhang D, Zhu H. The Role of the Gut Microbiota in Complications among Hemodialysis Patients. Microorganisms 2024; 12:1878. [PMID: 39338552 PMCID: PMC11434415 DOI: 10.3390/microorganisms12091878] [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: 07/31/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
The composition of the gut microbiota varies among end-stage renal disease (ESRD) patients on the basis of their mode of renal replacement therapy (RRT), with notably more pronounced dysbiosis occurring in those undergoing hemodialysis (HD). Interventions such as dialysis catheters, unstable hemodynamics, strict dietary restrictions, and pharmacotherapy significantly alter the intestinal microenvironment, thus disrupting the gut microbiota composition in HD patients. The gut microbiota may influence HD-related complications, including cardiovascular disease (CVD), infections, anemia, and malnutrition, through mechanisms such as bacterial translocation, immune regulation, and the production of gut microbial metabolites, thereby affecting both the quality of life and the prognosis of patients. This review focuses on alterations in the gut microbiota and its metabolites in HD patients. Additionally, understanding the impact of the gut microbiota on the complications of HD could provide insights into the development of novel treatment strategies to prevent or alleviate complications in HD patients.
Collapse
Affiliation(s)
- Junxia Du
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaolin Zhao
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiaonan Ding
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yingjie Duan
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Qinqin Ren
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Chenwen Song
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaochen Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Dong Zhang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| |
Collapse
|
13
|
Mirzababaei A, Mahmoodi M, Keshtkar A, Ebrahimi S, Pashayee-Khamene F, Abaj F, Radmehr M, Khalili P, Mehri Hajmir M, Mirzaei K. The interaction between dietary nitrates/nitrites intake and gut microbial metabolites on metabolic syndrome: a cross-sectional study. Front Public Health 2024; 12:1398460. [PMID: 39328991 PMCID: PMC11425044 DOI: 10.3389/fpubh.2024.1398460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 08/12/2024] [Indexed: 09/28/2024] Open
Abstract
Background Metabolic syndrome (MetS) prevalence has increased globally.The evidence shows thatdiet and gut microbial metabolites includingtrimethylamine N-oxide (TMAO) and kynurenine (KYN) play an important role in developing MetS. However, there is a lack of evidence on associations between between diet and these metabolites. This study aimed to investigate the interaction between dietary nitrate/nitrite and gut microbial metabolites (TMAO, KYN) on MetS and its components. Methods This cross-sectional study included 250 adults aged 20-50 years. Dietary intake was assessed using food frequency questionnaires (FFQ), and serum TMAO and KYN levels were measured. MetS was defined usingthe National Cholesterol Education Program Adult Treatment Panel (NCEP ATP III) criteria. Result The ATPIII index revealed an 11% prevalence of metabolic syndrome among the study participants. After adjusting for confounders, significant positive interactions were found: High animal-source nitrate intake and high TMAO levels with elevated triglycerides (TG) (p interaction = 0.07) and abdominal obesity (p interaction = 0.08). High animal-source nitrate intake and high KYN levels with increased TG (p interaction = 0.01) and decreased high-density lipoprotein cholesterol (HDL) (p interaction = 0.01).Individuals with high animal-source nitrite intake and high TMAO levels showed increased risk of hypertriglyceridemia (OR: 1.57, 95%CI: 0.35-2.87, p = 0.05), hypertension (OR: 1.53, 95%CI: 0.33-2.58, p = 0.06), and lower HDL (OR: 1.96, 95%CI: 0.42-2.03, p = 0.04). Similarly, high animal-source nitrite intake with high KYN levels showed lower HDL (OR: 2.44, 95%CI: 1.92-3.89, p = 0.07) and increased risk of hypertension (OR: 2.17,95%CI: 1.69-3.40, p = 0.05). Conversely, Negative interactions were found between high plant-source nitrate/nitrite intake with high KYN and TMAO levels on MetS and some components. Conclusion There is an interaction between dietary nitrate/nitrite source (animal vs. plant) and gut microbial metabolites (TMAO and KYN) on the risk of of MetS and its components. These findings highlight the importance of considering diet, gut microbiome metabolites, and their interactions in MetS risk assessment.
Collapse
Affiliation(s)
- Atieh Mirzababaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mahmoodi
- Department of Cellular and Molecular Nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbasali Keshtkar
- Department of Disaster and Emergency Health, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Ebrahimi
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | | | - Faezeh Abaj
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, VIC, Australia
| | - Mina Radmehr
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Pardis Khalili
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahya Mehri Hajmir
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Dora D, Szőcs E, Soós Á, Halasy V, Somodi C, Mihucz A, Rostás M, Mógor F, Lohinai Z, Nagy N. From bench to bedside: an interdisciplinary journey through the gut-lung axis with insights into lung cancer and immunotherapy. Front Immunol 2024; 15:1434804. [PMID: 39301033 PMCID: PMC11410641 DOI: 10.3389/fimmu.2024.1434804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
This comprehensive review undertakes a multidisciplinary exploration of the gut-lung axis, from the foundational aspects of anatomy, embryology, and histology, through the functional dynamics of pathophysiology, to implications for clinical science. The gut-lung axis, a bidirectional communication pathway, is central to understanding the interconnectedness of the gastrointestinal- and respiratory systems, both of which share embryological origins and engage in a continuous immunological crosstalk to maintain homeostasis and defend against external noxa. An essential component of this axis is the mucosa-associated lymphoid tissue system (MALT), which orchestrates immune responses across these distant sites. The review delves into the role of the gut microbiome in modulating these interactions, highlighting how microbial dysbiosis and increased gut permeability ("leaky gut") can precipitate systemic inflammation and exacerbate respiratory conditions. Moreover, we thoroughly present the implication of the axis in oncological practice, particularly in lung cancer development and response to cancer immunotherapies. Our work seeks not only to synthesize current knowledge across the spectrum of science related to the gut-lung axis but also to inspire future interdisciplinary research that bridges gaps between basic science and clinical application. Our ultimate goal was to underscore the importance of a holistic understanding of the gut-lung axis, advocating for an integrated approach to unravel its complexities in human health and disease.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Viktória Halasy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Csenge Somodi
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Melinda Rostás
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Mógor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
15
|
Elantary R, Othman S. Role of L-carnitine in Cardiovascular Health: Literature Review. Cureus 2024; 16:e70279. [PMID: 39329040 PMCID: PMC11427024 DOI: 10.7759/cureus.70279] [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] [Accepted: 09/26/2024] [Indexed: 09/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide. Secondary preventive measures, like anti-platelet medications, B-blockers, and angiotensin-converting enzyme (ACE) inhibitors, have been found to dramatically lower the risk of cardiovascular disease. However, prolonged usage of these drugs has been linked to multiple adverse impacts. Hence, finding more efficient treatments, especially dietary strategies for long-term use in daily life, is advantageous for primary prevention and treatment. L-carnitine, a naturally occurring amino acid derivative normally synthesized in the liver and kidney, is believed to have a considerable influence on cardiovascular health. L-carnitine can enhance both contractile performance and structural integrity of the cardiac muscle by maintaining efficient energy production and reducing oxidative stress. This literature review aims to address several pressing questions regarding the role of L-carnitine in cardiovascular health: what are the physiological functions of L-carnitine, particularly in relation to cardiovascular health; how effective and safe is L-carnitine supplementation in the management of various cardiovascular diseases, primarily ischemic heart disease, heart failure, and peripheral vascular disease; what are the underlying mechanisms through which L-carnitine exerts its cardioprotective effects; what controversies exist in the current research; and finally, what should be the future directions? Through this comprehensive analysis, the review aims to enrich our understanding of L-carnitine's role in cardiovascular health, providing a robust foundation for future academic and clinical endeavors. PubMed, Embase, and Google Scholar have been used to search the following keywords: L-carnitine, cardiovascular health, mitochondrial function, and L-carnitine side effects. Then, using the existing search engine formats, some keyword combinations were used to find the related articles included and every possibility, including using every first keyword combination with another keyword, using every keyword in every place at each given box, etc. Around 308 articles were reviewed using this process, including systemic reviews, meta-analysis studies, randomized controlled trials, and literature review articles. In the end, after leaving the pure articles related to the topic as 35 articles, which are attached below with direct citation, the majority of them were very fresh articles, as recent as 2010, and back words, except just one paper related to the impact of L-carnitine post-myocardial infarction, as its data provided us with a positive and promising impact of L-carnitine in this field. L‑carnitine seems to have a pivotal role in cardiovascular health due to its energy metabolism, anti-oxidative stress, and endothelial role. The safety and effectiveness of L-carnitine administration remain an issue for scientific investigation. One of the major concerns is that the intestinal metabolism of L-carnitine generates trimethylamine-N-oxide (TMAO), a compound that has been linked with faster atherosclerosis progression.
Collapse
Affiliation(s)
- Ramy Elantary
- Department of Acute Medicine, Royal Liverpool University Hospital, Liverpool, GBR
| | - Samar Othman
- Department of Diabetes and Endocrinology, Countess of Chester Hospital, Chester, GBR
| |
Collapse
|
16
|
Wang M, Zheng L, Meng Y, Ma S, Zhao D, Xu Y. Broadening horizons: intestinal microbiota as a novel biomarker and potential treatment for hypertensive disorders of pregnancy. Front Cell Infect Microbiol 2024; 14:1446580. [PMID: 39239636 PMCID: PMC11374776 DOI: 10.3389/fcimb.2024.1446580] [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: 06/10/2024] [Accepted: 07/24/2024] [Indexed: 09/07/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are severe complications of pregnancy with high morbidity and are a major cause of increased maternal and infant morbidity and mortality. Currently, there is a lack of effective early diagnostic indicators and safe and effective preventive strategies for HDP in clinical practice, except for monitoring maternal blood pressure levels, the degree of proteinuria, organ involvement and fetal conditions. The intestinal microbiota consists of the gut flora and intestinal environment, which is the largest microecosystem of the human body and participates in material and energy metabolism, gene expression regulation, immunity regulation, and other functions. During pregnancy, due to changes in hormone levels and altered immune function, the intestinal microecological balance is affected, triggering HDP. A dysregulated intestinal microenvironment influences the composition and distribution of the gut flora and changes the intestinal barrier, driving beneficial or harmful bacterial metabolites and inflammatory responses to participate in the development of HDP and promote its malignant development. When the gut flora is dysbiotic and affects blood pressure, supplementation with probiotics and dietary fiber can be used to intervene. In this review, the interaction between the intestinal microbiota and HDP was investigated to explore the feasibility of the gut flora as a novel biomarker of HDP and to provide a new strategy and basis for the prevention and treatment of clinical HDP.
Collapse
Affiliation(s)
- Min Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yang Meng
- Jilin Province Product Quality Supervision and Inspection Institute, Changchun, China
| | - Shuai Ma
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Donghai Zhao
- Department of Pathology, Jilin Medical College, Jilin, China
| | - Ying Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Hamamah S, Lobiuc A, Covasa M. Antioxidant Role of Probiotics in Inflammation-Induced Colorectal Cancer. Int J Mol Sci 2024; 25:9026. [PMID: 39201713 PMCID: PMC11354872 DOI: 10.3390/ijms25169026] [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: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Colorectal cancer (CRC) continues to be a significant contributor to global morbidity and mortality. Emerging evidence indicates that disturbances in gut microbial composition, the formation of reactive oxygen species (ROS), and the resulting inflammation can lead to DNA damage, driving the pathogenesis and progression of CRC. Notably, bacterial metabolites can either protect against or contribute to oxidative stress by modulating the activity of antioxidant enzymes and influencing signaling pathways that govern ROS-induced inflammation. Additionally, microbiota byproducts, when supplemented through probiotics, can affect tumor microenvironments to enhance treatment efficacy and selectively mediate the ROS-induced destruction of CRC cells. This review aims to discuss the mechanisms by which taxonomical shifts in gut microbiota and related metabolites such as short-chain fatty acids, secondary bile acids, and trimethylamine-N-oxide influence ROS concentrations to safeguard or promote the onset of inflammation-mediated CRC. Additionally, we focus on the role of probiotic species in modulating ROS-mediated signaling pathways that influence both oxidative status and inflammation, such as Nrf2-Keap1, NF-κB, and NLRP3 to mitigate carcinogenesis. Overall, a deeper understanding of the role of gut microbiota on oxidative stress may aid in delaying or preventing the onset of CRC and offer new avenues for adjunct, CRC-specific therapeutic interventions such as cancer immunotherapy.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Andrei Lobiuc
- Department of Medicine and Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania;
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Medicine and Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania;
| |
Collapse
|
18
|
Livzan MA, Bikbavova GR, Lisyutenko NS, Romanyuk AE, Drapkina OM. Cardiovascular Risk in Patients with Inflammatory Bowel Diseases-The Role of Endothelial Dysfunction. Diagnostics (Basel) 2024; 14:1722. [PMID: 39202210 PMCID: PMC11353271 DOI: 10.3390/diagnostics14161722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Inflammatory bowel disease (IBD) is associated with an increased risk of cardiovascular disease (CVD). Cardiovascular pathology in people with IBD has not been well studied to date, and a direct link between cardiovascular events and IBD has not been established. The mechanisms underlying this association include the parallel and dynamic interaction of inflammation, modulation of the composition of the gut microbiota, endothelial dysfunction, thrombogenicity, and increased endothelial and epithelial permeability. Endothelial dysfunction is a common aspect of the pathogenesis of IBD and atherosclerotic CVD and can be considered one of the most important factors leading to the development and progression of cardiovascular pathology in patients with IBD. The purpose of this literature review is to describe the mechanisms underlying the development of endothelial dysfunction and disorders of the structure and function of the gut-vascular barrier in the pathogenesis of the cardiovascular manifestation of IBD.
Collapse
Affiliation(s)
- Maria A. Livzan
- Department of Faculty Therapy, Omsk State Medical University, 644099 Omsk, Russia;
| | - Galiya R. Bikbavova
- Department of Internal Medicine and Endocrinology, Omsk State Medical University, 644099 Omsk, Russia;
| | - Natalya S. Lisyutenko
- Department of Internal Medicine and Endocrinology, Omsk State Medical University, 644099 Omsk, Russia;
| | - Alisa E. Romanyuk
- Faculty of Medicine, Omsk State Medical University, 644099 Omsk, Russia;
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia;
| |
Collapse
|
19
|
Zhu Y, Lai Y, Hu Y, Fu Y, Zhang Z, Lin N, Huang W, Zheng L. The mechanisms underlying acute myocardial infarction in chronic kidney disease patients undergoing hemodialysis. Biomed Pharmacother 2024; 177:117050. [PMID: 38968794 DOI: 10.1016/j.biopha.2024.117050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death in chronic kidney disease (CKD). Hemodialysis is one of the main treatments for patients with end-stage kidney disease. Epidemiological data has shown that acute myocardial infarction (AMI) accounts for the main reason for death in patients with CKD under hemodialysis therapy. Immune dysfunction and changes in metabolism (including a high level of inflammatory cytokines, a disorder of lipid and mineral ion homeostasis, accumulation of uremic toxins et al.) during CKD can deteriorate stability of atherosclerotic plaque and promote vascular calcification, which are exactly the pathophysiological mechanisms underlying the occurrence of AMI. Meanwhile, the hemodialysis itself also has adverse effects on lipoprotein, the immune system and hemodynamics, which contribute to the high incidence of AMI in these patients. This review aims to summarize the mechanisms and further promising methods of prevention and treatment of AMI in CKD patients undergoing hemodialysis, which can provide an excellent paradigm for exploring the crosstalk between the kidney and cardiovascular system.
Collapse
Affiliation(s)
- Yujie Zhu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Yuchen Lai
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yuxuan Hu
- Hubei University of Science and Technology, Xianning 437100, China
| | - Yiwen Fu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Zheng Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Nan Lin
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou 350013, China
| | - Wei Huang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Institute of Brain Disorders, The Capital Medical University, Beijing 100050, China.
| |
Collapse
|
20
|
Klepinowski T, Skonieczna-Żydecka K, Łoniewski I, Pettersson SD, Wierzbicka-Woś A, Kaczmarczyk M, Palma J, Sawicki M, Taterra D, Poncyljusz W, Alshafai NS, Stachowska E, Ogilvy CS, Sagan L. A prospective pilot study of gut microbiome in cerebral vasospasm and delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Sci Rep 2024; 14:17617. [PMID: 39080476 PMCID: PMC11289281 DOI: 10.1038/s41598-024-68722-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
A recent systematic review indicated that gut-microbiota-brain axis contributes to growth and rupture of intracranial aneurysms. However, gaps were detected in the role of intestinal microbiome in cerebral vasospasm (CVS) after aneurysmal subarachnoid hemorrhage (aSAH). This is the first pilot study aiming to test study feasibility and identify differences in gut microbiota between subjects with and without CVS following aSAH. A prospective nested case-control pilot study with 1:1 matching was conducted recruiting subjects with aSAH: cases with CVS; and controls without CVS based on the clinical picture and structured bedside transcranial Doppler (TCD). Fecal samples for microbiota analyses by means of 16S rRNA gene amplicon sequencing were collected within the first 96 h after ictus. Operational taxonomic unit tables were constructed, diversity metrics calculated, phylogenetic trees built, and differential abundance analysis (DAA) performed. At baseline, the groups did not differ significantly in basic demographic and aneurysm-related characteristics (p > 0.05). Alpha-diversity (richness and Shannon Index) was significantly reduced in cases of middle cerebral artery (MCA) vasospasm (p < 0.05). In DAA, relative abundance of genus Acidaminococcus was associated with MCA vasospasm (p = 0.00013). Two butyrate-producing genera, Intestinimonas and Butyricimonas, as well as [Clostridium] innocuum group had the strongest negative correlation with the mean blood flow velocity in anterior cerebral arteries (p < 0.01; rho = - 0.63; - 0.57, and - 0.57, respectively). In total, 16 gut microbial genera were identified to correlate with TCD parameters, and two intestinal genera correlated with outcome upon discharge. In this pilot study, we prove study feasibility and present the first preliminary evidence of gut microbiome signature associating with CVS as a significant cause of stroke in subjects with aSAH.
Collapse
Affiliation(s)
- Tomasz Klepinowski
- Department of Neurosurgery, Pomeranian Medical University Hospital no. 1, Unii Lubelskiej 1, 71-252, Szczecin, Poland.
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland.
| | - Igor Łoniewski
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland
| | - Samuel D Pettersson
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Anna Wierzbicka-Woś
- Research and Development Centre, Sanprobi sp. z o.o. sp. K, Kurza Stopka 5/c, 70-535, Szczecin, Poland
| | - Mariusz Kaczmarczyk
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland
- Research and Development Centre, Sanprobi sp. z o.o. sp. K, Kurza Stopka 5/c, 70-535, Szczecin, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460, Szczecin, Poland
| | - Marcin Sawicki
- Department of Diagnostic Imaging and Interventional Radiology, Pomeranian Medical University in Szczecin Hospital no. 1, Szczecin, Poland
| | - Dominik Taterra
- Department of Orthopedics and Rehabilitation, Jagiellonian University Medical College, Zakopane, Poland
| | - Wojciech Poncyljusz
- Department of Diagnostic Imaging and Interventional Radiology, Pomeranian Medical University in Szczecin Hospital no. 1, Szczecin, Poland
| | | | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Broniewskiego 24, 70-204, Szczecin, Poland
| | - Christopher S Ogilvy
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Leszek Sagan
- Department of Neurosurgery, Pomeranian Medical University Hospital no. 1, Unii Lubelskiej 1, 71-252, Szczecin, Poland
| |
Collapse
|
21
|
Camargo LL, Rios FJ, Montezano AC, Touyz RM. Reactive oxygen species in hypertension. Nat Rev Cardiol 2024:10.1038/s41569-024-01062-6. [PMID: 39048744 DOI: 10.1038/s41569-024-01062-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Hypertension is a leading risk factor for stroke, heart disease and chronic kidney disease. Multiple interacting factors and organ systems increase blood pressure and cause target-organ damage. Among the many molecular elements involved in the development of hypertension are reactive oxygen species (ROS), which influence cellular processes in systems that contribute to blood pressure elevation (such as the cardiovascular, renal, immune and central nervous systems, or the renin-angiotensin-aldosterone system). Dysregulated ROS production (oxidative stress) is a hallmark of hypertension in humans and experimental models. Of the many ROS-generating enzymes, NADPH oxidases are the most important in the development of hypertension. At the cellular level, ROS influence signalling pathways that define cell fate and function. Oxidative stress promotes aberrant redox signalling and cell injury, causing endothelial dysfunction, vascular damage, cardiovascular remodelling, inflammation and renal injury, which are all important in both the causes and consequences of hypertension. ROS scavengers reduce blood pressure in almost all experimental models of hypertension; however, clinical trials of antioxidants have yielded mixed results. In this Review, we highlight the latest advances in the understanding of the role and the clinical implications of ROS in hypertension. We focus on cellular sources of ROS, molecular mechanisms of oxidative stress and alterations in redox signalling in organ systems, and their contributions to hypertension.
Collapse
Affiliation(s)
- Livia L Camargo
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada.
| | - Francisco J Rios
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada.
- Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada.
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
22
|
Pallozzi M, De Gaetano V, Di Tommaso N, Cerrito L, Santopaolo F, Stella L, Gasbarrini A, Ponziani FR. Role of Gut Microbial Metabolites in the Pathogenesis of Primary Liver Cancers. Nutrients 2024; 16:2372. [PMID: 39064815 PMCID: PMC11280141 DOI: 10.3390/nu16142372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatobiliary malignancies, which include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are the sixth most common cancers and the third leading cause of cancer-related death worldwide. Hepatic carcinogenesis is highly stimulated by chronic inflammation, defined as fibrosis deposition, and an aberrant imbalance between liver necrosis and nodular regeneration. In this context, the gut-liver axis and gut microbiota have demonstrated a critical role in the pathogenesis of HCC, as dysbiosis and altered intestinal permeability promote bacterial translocation, leading to chronic liver inflammation and tumorigenesis through several pathways. A few data exist on the role of the gut microbiota or bacteria resident in the biliary tract in the pathogenesis of CCA, and some microbial metabolites, such as choline and bile acids, seem to show an association. In this review, we analyze the impact of the gut microbiota and its metabolites on HCC and CCA development and the role of gut dysbiosis as a biomarker of hepatobiliary cancer risk and of response during anti-tumor therapy. We also discuss the future application of gut microbiota in hepatobiliary cancer management.
Collapse
Affiliation(s)
- Maria Pallozzi
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Valeria De Gaetano
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Natalia Di Tommaso
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Lucia Cerrito
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Francesco Santopaolo
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Leonardo Stella
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Antonio Gasbarrini
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
23
|
Fei SF, Hou C, Jia F. Effects of salidroside on atherosclerosis: potential contribution of gut microbiota. Front Pharmacol 2024; 15:1400981. [PMID: 39092226 PMCID: PMC11292615 DOI: 10.3389/fphar.2024.1400981] [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: 05/08/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Much research describes gut microbiota in atherosclerotic cardiovascular diseases (ASCVD) for that the composition of the intestinal microbiome or its metabolites can directly participate in the development of endothelial dysfunction, atherosclerosis and its adverse complications. Salidroside, a natural phenylpropane glycoside, exhibits promising biological activity against the progression of ASCVD. Recent studies suggested that the gut microbiota played a crucial role in mediating the diverse beneficial effects of salidroside on health. Here, we describe the protective effects of salidroside against the progression of atherosclerosis. Salidroside regulates the abundance of gut microbiotas and gut microbe-dependent metabolites. Moreover, salidroside improves intestinal barrier function and maintains intestinal epithelial barrier function integrity. In addition, salidroside attenuates the inflammatory responses exacerbated by gut microbiota disturbance. This review delves into how salidroside functions to ameliorate atherosclerosis by focusing on its interaction with gut microbiota, uncovering the potential roles of gut microbiota in the diverse biological impacts of salidroside.
Collapse
Affiliation(s)
| | | | - Fang Jia
- Department of Cardiovascular Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
24
|
Sandireddy R, Sakthivel S, Gupta P, Behari J, Tripathi M, Singh BK. Systemic impacts of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH) on heart, muscle, and kidney related diseases. Front Cell Dev Biol 2024; 12:1433857. [PMID: 39086662 PMCID: PMC11289778 DOI: 10.3389/fcell.2024.1433857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is the most common liver disorder worldwide, with an estimated global prevalence of more than 31%. Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as non-alcoholic steatohepatitis (NASH), is a progressive form of MASLD characterized by hepatic steatosis, inflammation, and fibrosis. This review aims to provide a comprehensive analysis of the extrahepatic manifestations of MASH, focusing on chronic diseases related to the cardiovascular, muscular, and renal systems. A systematic review of published studies and literature was conducted to summarize the findings related to the systemic impacts of MASLD and MASH. The review focused on the association of MASLD and MASH with metabolic comorbidities, cardiovascular mortality, sarcopenia, and chronic kidney disease. Mechanistic insights into the concept of lipotoxic inflammatory "spill over" from the MASH-affected liver were also explored. MASLD and MASH are highly associated (50%-80%) with other metabolic comorbidities such as impaired insulin response, type 2 diabetes, dyslipidemia, hypertriglyceridemia, and hypertension. Furthermore, more than 90% of obese patients with type 2 diabetes have MASH. Data suggest that in middle-aged individuals (especially those aged 45-54), MASLD is an independent risk factor for cardiovascular mortality, sarcopenia, and chronic kidney disease. The concept of lipotoxic inflammatory "spill over" from the MASH-affected liver plays a crucial role in mediating the systemic pathological effects observed. Understanding the multifaceted impact of MASH on the heart, muscle, and kidney is crucial for early detection and risk stratification. This knowledge is also timely for implementing comprehensive disease management strategies addressing multi-organ involvement in MASH pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
25
|
Kerpa S, Schulze VR, Holzapfel M, Cvancar L, Fischer M, Maison W. Decoration of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) with N-oxides increases the T 1 relaxivity of Gd-complexes. ChemistryOpen 2024; 13:e202300298. [PMID: 38224205 PMCID: PMC11230940 DOI: 10.1002/open.202300298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/15/2023] [Indexed: 01/16/2024] Open
Abstract
High complex stability and longitudinal relaxivity of Gd-based contrast agents are important requirements for magnetic resonance imaging (MRI) because they ensure patient safety and contribute to measurement sensitivity. Charged and zwitterionic Gd3+-complexes of the well-known chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) provide an excellent basis for the development of safe and sensitive contrast agents. In this report, we describe the synthesis of DOTA-NOx, a DOTA derivative with four N-oxide functionalities via "click" functionalization of the tetraazide DOTAZA. The resulting complexes Gd-DOTA-NOx and Eu-DOTA-NOx are stable compounds in aqueous solution. NMR-spectroscopic characterization revealed a high excess of the twisted square antiprismatic (TSAP) coordination geometry over square antiprismatic (SAP). The longitudinal relaxivity of Gd-DOTA-NOx was found to be r1=7.7 mm-1 s-1 (1.41 T, 37 °C), an unusually high value for DOTA complexes of comparable weight. We attribute this high relaxivity to the steric influence and an ordering effect on outer sphere water molecules surrounding the complex generated by the strongly hydrated N-oxide groups. Moreover, Gd-DOTA-NOx was found to be stable against transchelation with high excess of EDTA (200 eq) over a period of 36 h, and it has a similar in vitro cell toxicity as clinically used DOTA-based GBCAs.
Collapse
Affiliation(s)
- Svenja Kerpa
- Department of Chemistry, Institute of Pharmacy, Universität Hamburg, Bundesstrasse 45, 20146, Hamburg, Germany
| | - Verena R Schulze
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146, Hamburg, Germany
| | - Malte Holzapfel
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146, Hamburg, Germany
| | - Lina Cvancar
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, Grindelallee 117, 20146, Hamburg, Germany
| | - Markus Fischer
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, Grindelallee 117, 20146, Hamburg, Germany
| | - Wolfgang Maison
- Department of Chemistry, Institute of Pharmacy, Universität Hamburg, Bundesstrasse 45, 20146, Hamburg, Germany
| |
Collapse
|
26
|
Vallianou NG, Kounatidis D, Psallida S, Vythoulkas-Biotis N, Adamou A, Zachariadou T, Kargioti S, Karampela I, Dalamaga M. NAFLD/MASLD and the Gut-Liver Axis: From Pathogenesis to Treatment Options. Metabolites 2024; 14:366. [PMID: 39057689 PMCID: PMC11278747 DOI: 10.3390/metabo14070366] [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: 05/14/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) poses an emerging threat topublic health. Nonalcoholic steatohepatitis (NASH) is reported to be the most rapidly rising cause of hepatocellular carcinoma in the western world. Recently, a new term has been proposed: metabolic dysfunction-associated steatotic liver disease (MASLD). The introduction of this new terminology has sparked a debate about the interchangeability of these terms. The pathogenesis of NAFLD/MASLD is thought to be multifactorial, involving both genetic and environmental factors. Among these factors, alterations in gut microbiota and gut dysbiosis have recently garnered significant attention. In this context, this review will further discuss the gut-liver axis, which refers to the bidirectional interaction between the human gut microbiota and the liver. Additionally, the therapeutic potential of probiotics, particularly next-generation probiotics and genetically engineered bacteria, will be explored. Moreover, the role of prebiotics, synbiotics, postbiotics, and phages as well as fecal microbiota transplantation will be analyzed. Particularly for lean patients with NAFLD/MASLD, who have limited treatment options, approaches that modify the diversity and composition of the gut microbiota may hold promise. However, due to ongoing safety concerns with approaches that modulate gut microbiota, further large-scale studies are necessary to better assess their efficacy and safety in treating NAFLD/MASLD.
Collapse
Affiliation(s)
- Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Dimitris Kounatidis
- Department of Internal Medicine, Hippokration General Hospital, 114 Vassilissis Sofias Str., 11527 Athens, Greece;
| | - Sotiria Psallida
- Department of Microbiology, “KAT” General Hospital of Attica, 14561 Athens, Greece;
| | - Nikolaos Vythoulkas-Biotis
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Andreas Adamou
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Tatiana Zachariadou
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Sofia Kargioti
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Irene Karampela
- Second Department of Critical Care, Attikon University Hospital, 1 Rimini Str., 12462 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| |
Collapse
|
27
|
Ge Y, Wang J, Wu L, Wu J. Gut microbiota: a potential new regulator of hypertension. Front Cardiovasc Med 2024; 11:1333005. [PMID: 38993521 PMCID: PMC11236727 DOI: 10.3389/fcvm.2024.1333005] [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: 11/04/2023] [Accepted: 04/16/2024] [Indexed: 07/13/2024] Open
Abstract
Hypertension is a significant risk factor for cardiovascular and cerebrovascular diseases and has become a global public health concern. Although hypertension results from a combination of factors, the specific mechanism is still unclear. However, increasing evidence suggests that gut microbiota is closely associated with the development of hypertension. We provide a summary of the composition and physiological role of gut microbiota. We then delve into the mechanism of gut microbiota and its metabolites involved in the occurrence and development of hypertension. Finally, we review various regimens for better-controlling hypertension from the diet, exercise, drugs, antibiotics, probiotics, and fecal transplantation perspectives.
Collapse
Affiliation(s)
- Yanmin Ge
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiaxin Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lincong Wu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Junduo Wu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
28
|
Wang Y, Qin C, Tian S, Meng Y, Chen Y, Fu S, Xu M, Wang B, Li Y, Ouyang Q, Ling H, Liu M. Dataset on cardiac structural and functional parameters in TMAO-challenged mouse. Data Brief 2024; 54:110465. [PMID: 38711736 PMCID: PMC11070669 DOI: 10.1016/j.dib.2024.110465] [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: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
Trimethylamine-N-oxide (TMAO) is a gut-derived metabolite formed from dietary choline and l-carnitine, known to impede cholesterol metabolism and is implicated in the pathogenesis of thrombosis and atherosclerosis, contributing to the etiology of cardiovascular diseases. We present a dataset derived from an experimental study designed to elucidate the cardiotoxic effects of TMAO. This dataset encompasses echocardiographic assessments from two cohorts of mice: one subjected to a 6-week regimen of 20 mg/kg/day TMAO injections (n = 16) and a control group (n = 18). Each subject's echocardiographic dataset comprises six high-resolution TIFF images, capturing both B-type and M-mode views in standard echocardiographic planes, along with two additional M-mode images enriched with analysed cardiac functional data. Complementing these images, a CSV-formatted report details critical cardiac parameters, including heart rate, ejection fraction, and fractional shortening, among others. In a novel approach to enhance data integrity and permit tailored analyses, we provide the original output files from the echocardiography apparatus, which researchers can reprocess using dedicated analysis software. This dataset is anticipated to be instrumental in advancing our understanding of the mechanistic links between TMAO exposure and cardiac dysfunction.
Collapse
Affiliation(s)
- Yingyu Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Chenchen Qin
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Shiyi Tian
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Yuqin Meng
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Yige Chen
- School of Nursing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Sunjing Fu
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Mengting Xu
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Bing Wang
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yuan Li
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Qin Ouyang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Hao Ling
- Department of Radiology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410000, Hunan, China
| | - Mingming Liu
- Institute of Microcirculation, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
- International Center of Microvascular Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
29
|
Kandalgaonkar MR, Kumar V, Vijay‐Kumar M. Digestive dynamics: Unveiling interplay between the gut microbiota and the liver in macronutrient metabolism and hepatic metabolic health. Physiol Rep 2024; 12:e16114. [PMID: 38886098 PMCID: PMC11182692 DOI: 10.14814/phy2.16114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Although the liver is the largest metabolic organ in the body, it is not alone in functionality and is assisted by "an organ inside an organ," the gut microbiota. This review attempts to shed light on the partnership between the liver and the gut microbiota in the metabolism of macronutrients (i.e., proteins, carbohydrates, and lipids). All nutrients absorbed by the small intestines are delivered to the liver for further metabolism. Undigested food that enters the colon is metabolized further by the gut microbiota that produces secondary metabolites, which are absorbed into portal circulation and reach the liver. These microbiota-derived metabolites and co-metabolites include ammonia, hydrogen sulfide, short-chain fatty acids, secondary bile acids, and trimethylamine N-oxide. Further, the liver produces several compounds, such as bile acids that can alter the gut microbial composition, which can in turn influence liver health. This review focuses on the metabolism of these microbiota metabolites and their influence on host physiology. Furthermore, the review briefly delineates the effect of the portosystemic shunt on the gut microbiota-liver axis, and current understanding of the treatments to target the gut microbiota-liver axis.
Collapse
Affiliation(s)
- Mrunmayee R. Kandalgaonkar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| | - Virender Kumar
- College of Pharmacy and Pharmaceutical SciencesUniversity of ToledoToledoOhioUSA
| | - Matam Vijay‐Kumar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| |
Collapse
|
30
|
Kobus M, Friedrich T, Zorn E, Burmeister N, Maison W. Medicinal Chemistry of Drugs with N-Oxide Functionalities. J Med Chem 2024; 67:5168-5184. [PMID: 38549449 PMCID: PMC11017254 DOI: 10.1021/acs.jmedchem.4c00254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 04/12/2024]
Abstract
Molecules with N-oxide functionalities are omnipresent in nature and play an important role in Medicinal Chemistry. They are synthetic or biosynthetic intermediates, prodrugs, drugs, or polymers for applications in drug development and surface engineering. Typically, the N-oxide group is critical for biomedical applications of these molecules. It may provide water solubility or decrease membrane permeability or immunogenicity. In other cases, the N-oxide has a special redox reactivity which is important for drug targeting and/or cytotoxicity. Many of the underlying mechanisms have only recently been discovered, and the number of applications of N-oxides in the healthcare field is rapidly growing. This Perspective article gives a short summary of the properties of N-oxides and their synthesis. It also provides a discussion of current applications of N-oxides in the biomedical field and explains the basic molecular mechanisms responsible for their biological activity.
Collapse
Affiliation(s)
- Michelle Kobus
- Universität Hamburg, Department of Chemistry, Bundesstrasse 45, 20146 Hamburg, Germany
| | - Timo Friedrich
- Universität Hamburg, Department of Chemistry, Bundesstrasse 45, 20146 Hamburg, Germany
| | - Eilika Zorn
- Universität Hamburg, Department of Chemistry, Bundesstrasse 45, 20146 Hamburg, Germany
| | - Nils Burmeister
- Universität Hamburg, Department of Chemistry, Bundesstrasse 45, 20146 Hamburg, Germany
| | - Wolfgang Maison
- Universität Hamburg, Department of Chemistry, Bundesstrasse 45, 20146 Hamburg, Germany
| |
Collapse
|
31
|
Chen X, Gu J, Zhang X. Brain-Heart Axis and the Inflammatory Response: Connecting Stroke and Cardiac Dysfunction. Cardiology 2024; 149:369-382. [PMID: 38574466 PMCID: PMC11309082 DOI: 10.1159/000538409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND In recent years, the mechanistic interaction between the brain and heart has been explored in detail, which explains the effects of brain injuries on the heart and those of cardiac dysfunction on the brain. Brain injuries are the predominant cause of post-stroke deaths, and cardiac dysfunction is the second leading cause of mortality after stroke onset. SUMMARY Several studies have reported the association between brain injuries and cardiac dysfunction. Therefore, it is necessary to study the influence on the heart post-stroke to understand the underlying mechanisms of stroke and cardiac dysfunction. This review focuses on the mechanisms and the effects of cardiac dysfunction after the onset of stroke (ischemic or hemorrhagic stroke). KEY MESSAGES The role of the site of stroke and the underlying mechanisms of the brain-heart axis after stroke onset, including the hypothalamic-pituitary-adrenal axis, inflammatory and immune responses, brain-multi-organ axis, are discussed.
Collapse
Affiliation(s)
- Xiaosheng Chen
- Department of Neurosurgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Jiajie Gu
- Department of Neurosurgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Xiaojia Zhang
- Department of Neurosurgery, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
32
|
Xie H, Yang N, Yu C, Lu L. Uremic toxins mediate kidney diseases: the role of aryl hydrocarbon receptor. Cell Mol Biol Lett 2024; 29:38. [PMID: 38491448 PMCID: PMC10943832 DOI: 10.1186/s11658-024-00550-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/19/2024] [Indexed: 03/18/2024] Open
Abstract
Aryl hydrocarbon receptor (AhR) was originally identified as an environmental sensor that responds to pollutants. Subsequent research has revealed that AhR recognizes multiple exogenous and endogenous molecules, including uremic toxins retained in the body due to the decline in renal function. Therefore, AhR is also considered to be a uremic toxin receptor. As a ligand-activated transcriptional factor, the activation of AhR is involved in cell differentiation and senescence, lipid metabolism and fibrogenesis. The accumulation of uremic toxins in the body is hazardous to all tissues and organs. The identification of the endogenous uremic toxin receptor opens the door to investigating the precise role and molecular mechanism of tissue and organ damage induced by uremic toxins. This review focuses on summarizing recent findings on the role of AhR activation induced by uremic toxins in chronic kidney disease, diabetic nephropathy and acute kidney injury. Furthermore, potential clinical approaches to mitigate the effects of uremic toxins are explored herein, such as enhancing uremic toxin clearance through dialysis, reducing uremic toxin production through dietary interventions or microbial manipulation, and manipulating metabolic pathways induced by uremic toxins through controlling AhR signaling. This information may also shed light on the mechanism of uremic toxin-induced injury to other organs, and provide insights into clinical approaches to manipulate the accumulated uremic toxins.
Collapse
Affiliation(s)
- Hongyan Xie
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, China
| | - Ninghao Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, China.
| | - Limin Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| |
Collapse
|
33
|
Charitos IA, Aliani M, Tondo P, Venneri M, Castellana G, Scioscia G, Castellaneta F, Lacedonia D, Carone M. Biomolecular Actions by Intestinal Endotoxemia in Metabolic Syndrome. Int J Mol Sci 2024; 25:2841. [PMID: 38474087 DOI: 10.3390/ijms25052841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Metabolic syndrome (MetS) is a combination of metabolic disorders that concurrently act as factors promoting systemic pathologies such as atherosclerosis or diabetes mellitus. It is now believed to encompass six main interacting conditions: visceral fat, imbalance of lipids (dyslipidemia), hypertension, insulin resistance (with or without impairing both glucose tolerance and fasting blood sugar), and inflammation. In the last 10 years, there has been a progressive interest through scientific research investigations conducted in the field of metabolomics, confirming a trend to evaluate the role of the metabolome, particularly the intestinal one. The intestinal microbiota (IM) is crucial due to the diversity of microorganisms and their abundance. Consequently, IM dysbiosis and its derivate toxic metabolites have been correlated with MetS. By intervening in these two factors (dysbiosis and consequently the metabolome), we can potentially prevent or slow down the clinical effects of the MetS process. This, in turn, may mitigate dysregulations of intestinal microbiota axes, such as the lung axis, thereby potentially alleviating the negative impact on respiratory pathology, such as the chronic obstructive pulmonary disease. However, the biomolecular mechanisms through which the IM influences the host's metabolism via a dysbiosis metabolome in both normal and pathological conditions are still unclear. In this study, we seek to provide a description of the knowledge to date of the IM and its metabolome and the factors that influence it. Furthermore, we analyze the interactions between the functions of the IM and the pathophysiology of major metabolic diseases via local and systemic metabolome's relate endotoxemia.
Collapse
Affiliation(s)
- Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, "Istitute" of Bari, 70124 Bari, Italy
| | - Maria Aliani
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, "Istitute" of Bari, 70124 Bari, Italy
| | - Pasquale Tondo
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Maria Venneri
- Istituti Clinici Scientifici Maugeri IRCCS, Genomics and Proteomics Laboratory, "Istitute" of Bari, 70124 Bari, Italy
| | - Giorgio Castellana
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, "Istitute" of Bari, 70124 Bari, Italy
| | - Giulia Scioscia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Francesca Castellaneta
- School of Clinical Biochemistry and Pathology, University of Bari (Aldo Moro), 70124 Bari, Italy
| | - Donato Lacedonia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Mauro Carone
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, "Istitute" of Bari, 70124 Bari, Italy
| |
Collapse
|
34
|
Han JM, Guo L, Chen XH, Xie Q, Song XY, Ma YL. Relationship between trimethylamine N-oxide and the risk of hypertension in patients with cardiovascular disease: A meta-analysis and dose-response relationship analysis. Medicine (Baltimore) 2024; 103:e36784. [PMID: 38181288 PMCID: PMC10766215 DOI: 10.1097/md.0000000000036784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/26/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND The gut microbiota-dependent metabolite trimethylamine N-oxide (TMAO) has recently been recognized to be one of the risk factors for cardiovascular disease (CVD). However, there is a scarcity of data on the relationship between circulating TMAO levels and hypertension in patients with CVD. Meta analysis and a dose-response relationship were used in this study to assess the relationship between circulating trimethylamine N-oxide levels and the risk of hypertension in patients with CVD. METHODS CNKI, Wanfang Database, Pubmed, Embase, Cochrane Library, and Web of Science were searched up to June 01, 2023. Meta-analysis and dose-response analysis of relative risk data from prospective cohort studies reporting on the relationship between circulating TMAO levels and hypertension risk in patients with CVD were conducted. RESULTS Fifteen studies with a total of 15,498 patients were included in the present meta-analysis. Compared with a lower circulating TMAO level, a higher TMAO level was associated with a higher risk of hypertension in patients with CVD (RR = 1.14,95%CI (1.08, 1.20)). And the higher the TMAO level, the greater the risk of hypertension. The dose-response analysis revealed a linear dose-response relationship between circulating TMAO levels and the risk of hypertension in patients with CVD. The risk of hypertension increased by 1.014% when the circulating TMAO level increased by 1 μ mol/L. CONCLUSION In patients with CVD, the level of circulating TMAO is significantly related to the risk of hypertension. The risk of hypertension increased by 1.014% for every 1 μ mol/L increase in circulating TMAO levels.
Collapse
Affiliation(s)
- Jia-Ming Han
- Medical College of Qinghai University, Xining, China
| | - Lu Guo
- Medical College of Qinghai University, Xining, China
| | - Xian-Hui Chen
- Medical College of Qinghai University, Xining, China
| | - Qian Xie
- Medical College of Qinghai University, Xining, China
| | - Xiu-Ying Song
- Medical College of Qinghai University, Xining, China
| | - Yu-Lan Ma
- Department of Cardiology, Affiliated Hospital of Qinghai University, Xining, China
| |
Collapse
|
35
|
León-Letelier RA, Dou R, Vykoukal J, Yip-Schneider MT, Maitra A, Irajizad E, Wu R, Dennison JB, Do KA, Zhang J, Schmidt CM, Hanash S, Fahrmann JF. Contributions of the Microbiome-Derived Metabolome for Risk Assessment and Prognostication of Pancreatic Cancer. Clin Chem 2024; 70:102-115. [PMID: 38175578 DOI: 10.1093/clinchem/hvad186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Increasing evidence implicates microbiome involvement in the development and progression of pancreatic ductal adenocarcinoma (PDAC). Studies suggest that reflux of gut or oral microbiota can lead to colonization in the pancreas, resulting in dysbiosis that culminates in release of microbial toxins and metabolites that potentiate an inflammatory response and increase susceptibility to PDAC. Moreover, microbe-derived metabolites can exert direct effector functions on precursors and cancer cells, as well as other cell types, to either promote or attenuate tumor development and modulate treatment response. CONTENT The occurrence of microbial metabolites in biofluids thereby enables risk assessment and prognostication of PDAC, as well as having potential for design of interception strategies. In this review, we first highlight the relevance of the microbiome for progression of precancerous lesions in the pancreas and, using liquid chromatography-mass spectrometry, provide supporting evidence that microbe-derived metabolites manifest in pancreatic cystic fluid and are associated with malignant progression of intraductal papillary mucinous neoplasm(s). We secondly summarize the biomarker potential of microbe-derived metabolite signatures for (a) identifying individuals at high risk of developing or harboring PDAC and (b) predicting response to treatment and disease outcomes. SUMMARY The microbiome-derived metabolome holds considerable promise for risk assessment and prognostication of PDAC.
Collapse
Affiliation(s)
- Ricardo A León-Letelier
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rongzhang Dou
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michele T Yip-Schneider
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anirban Maitra
- Department of Translational Molecular Pathology and Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ranran Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer B Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kim-An Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianjun Zhang
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, United States
| | - C Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
36
|
Garcia-Gutierrez E, O’Mahony AK, Dos Santos RS, Marroquí L, Cotter PD. Gut microbial metabolic signatures in diabetes mellitus and potential preventive and therapeutic applications. Gut Microbes 2024; 16:2401654. [PMID: 39420751 PMCID: PMC11492678 DOI: 10.1080/19490976.2024.2401654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/08/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetes mellitus can be subdivided into several categories based on origin and clinical characteristics. The most common forms of diabetes are type 1 (T1D), type 2 diabetes (T2D) and gestational diabetes mellitus (GDM). T1D and T2D are chronic diseases affecting around 537 million adults worldwide and it is projected that these numbers will increase by 12% over the next two decades, while GDM affects up to 30% of women during pregnancy, depending on diagnosis methods. These forms of diabetes have varied origins: T1D is an autoimmune disease, while T2D is commonly associated with, but not limited to, certain lifestyle patterns and GDM can result of a combination of genetic predisposition and pregnancy factors. Despite some pathogenic differences among these forms of diabetes, there are some common markers associated with their development. For instance, gut barrier impairment and inflammation associated with an unbalanced gut microbiota and their metabolites may be common factors in diabetes development and progression. Here, we summarize the microbial signatures that have been linked to diabetes, how they are connected to diet and, ultimately, the impact on metabolite profiles resulting from host-gut microbiota-diet interactions. Additionally, we summarize recent advances relating to promising preventive and therapeutic interventions focusing on the targeted modulation of the gut microbiota to alleviate T1D, T2D and GDM.
Collapse
Affiliation(s)
- Enriqueta Garcia-Gutierrez
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
- Departamento de Ingeniería Agronómica, Instituto de Biotecnología Vegetal, ETSIA-Universidad Politécnica de Cartagena, Cartagena, Spain
| | - A. Kate O’Mahony
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Reinaldo Sousa Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Marroquí
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Paul D. Cotter
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
37
|
She J, Sun L, Yu Y, Fan H, Li X, Zhang X, Zhuo X, Guo M, Liu J, Liu P, Tuerhongjiang G, Du B, Li H, Yu J, Yuan Z, Wu Y. A gut feeling of statin. Gut Microbes 2024; 16:2415487. [PMID: 39470680 DOI: 10.1080/19490976.2024.2415487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/21/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024] Open
Abstract
Statins, known as HMG-CoA reductase inhibitors, are widely utilized to reduce blood cholesterol levels and possess pleiotropic effects, including the influence on inflammation and macrophage proliferation. Despite their significant impact in diminishing the incidence of cardiovascular events and mortality, individual responses to statin therapy vary considerably. Understanding this variability is essential for optimizing treatment outcomes and minimizing adverse effects. The gut microbiota, a complex ecosystem of microorganisms within the gastrointestinal tract, plays a critical role in human health and disease. Emerging evidence has linked the gut microbiota to drug metabolism and response, with the potential to modulate the efficacy of statin therapy and its side effects. This review provides a comprehensive overview of the interaction between the gut microbiota and statins. It discusses how the gut microbiota can influence the therapeutic effects and side effects of statins and examines the mechanisms by which the gut microbiota affect statin response and cardiovascular diseases.
Collapse
Affiliation(s)
- Jianqing She
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi, China
| | - Lizhe Sun
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yue Yu
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Heze Fan
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Xia Li
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Xinyu Zhang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Xiaozhen Zhuo
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi, China
| | - Manyun Guo
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Junhui Liu
- Clinical Laboratory, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Peining Liu
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Gulinigaer Tuerhongjiang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Bin Du
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Hongbing Li
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Jun Yu
- Department of Medicine and Therapeutics and Institute of Digestive Disease, The State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zuyi Yuan
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yue Wu
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi, China
| |
Collapse
|
38
|
Filipovic B, Marjanovic-Haljilji M, Mijac D, Lukic S, Kapor S, Kapor S, Starcevic A, Popovic D, Djokovic A. Molecular Aspects of MAFLD-New Insights on Pathogenesis and Treatment. Curr Issues Mol Biol 2023; 45:9132-9148. [PMID: 37998750 PMCID: PMC10669943 DOI: 10.3390/cimb45110573] [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: 10/05/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Metabolic-associated liver disease (MAFLD) affects up to 70% of overweight and more than 90% of morbidly obese people, and its pathogenesis is rather complex and multifactorial. The criteria for MAFLD include the presence of hepatic steatosis in addition to one of the following three criteria: overweight or obesity, presence of type 2 diabetes mellitus (T2DM), or evidence of metabolic dysregulation. If the specific criteria are present, the diagnosis of MAFLD can be made regardless of alcohol consumption and previous liver disease. The pathophysiological mechanisms of MAFLD, including inflammation, lipotoxicity, mitochondrial disfunction, and oxidative stress, as well as the impact of intestinal gut microbiota, are constantly being elucidated. Treatment strategies that are continually emerging are based on different key points in MAFLD pathogenesis. Yet, the ideal therapeutic option has still not been found and future research is of great importance, as MAFLD represents a multisystemic disease with numerous complications.
Collapse
Affiliation(s)
- Branka Filipovic
- Department of Gastroenterology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia; (B.F.); (D.P.)
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
| | - Marija Marjanovic-Haljilji
- Department of Gastroenterology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia; (B.F.); (D.P.)
| | - Dragana Mijac
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Clinic of Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia
| | - Snezana Lukic
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Clinic of Gastroenterology and Hepatology, Clinical Center of Serbia, Koste Todorovica 2, 11000 Belgrade, Serbia
| | - Suncica Kapor
- Department of Hematology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia;
| | - Slobodan Kapor
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Institute of Anatomy “Niko Miljanic”, Dr Subotica Starijeg 4/2, 11000 Belgrade, Serbia
| | - Ana Starcevic
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Institute of Anatomy “Niko Miljanic”, Dr Subotica Starijeg 4/2, 11000 Belgrade, Serbia
| | - Dusan Popovic
- Department of Gastroenterology, Clinical and Hospital Center “Dr Dragisa Misovic—Dedinje”, Heroja Milana Tepica 1, 11020 Belgrade, Serbia; (B.F.); (D.P.)
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
| | - Aleksandra Djokovic
- Faculty of Medicine, University of Belgrade, Dr Subotica Starijeg 8, 11000 Belgrade, Serbia; (D.M.); (S.L.); (S.K.); (A.S.); (A.D.)
- Department of Cardiology, Clinical and Hospital Center “Bezanijska Kosa”, Dr Zorza Matea s/n, 11080 Belgrade, Serbia
| |
Collapse
|
39
|
Kountouras J, Kazakos E, Polyzos SA, Papaefthymiou A, Zavos C, Tzitiridou-Chatzopoulou M, Chatzopoulos D, Vardaka E, Gatopoulou A, Kyrailidi F, Mouratidou MC, Doulberis M. Potential impact of trained innate immunity on the pathophysiology of metabolic dysfunction-associated fatty liver disease. Clin Immunol 2023; 256:109776. [PMID: 37742792 DOI: 10.1016/j.clim.2023.109776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) occurs in a low-grade inflammatory milieu dependent on highly complex networks that span well-beyond the hepatic tissue injury. Dysfunctional systemic metabolism that characterizes the disease, is further induced in response to environmental cues that modify energy and metabolic cellular demands, thereby altering the availability of specific substrates that profoundly regulate, through epigenetic mechanisms, the phenotypic heterogeneity of immune cells and influence hematopoietic stem cell differentiation fate. This immuno-metabolic signaling drives the initiation of downstream effector pathways and results in the decompensation of hepatic homeostasis that precedes pro-fibrotic events. Recent evidence suggests that innate immune cells reside in different tissues in a memory effector state, a phenomenon termed trained immunity, that may be activated by subsequent exogenous (e.g., microbial, dietary) or endogenous (e.g., metabolic, apoptotic) stmuli. This process leads to long-term modifications in the epigenetic landscape that ultimately precondition the cells towards enhanced transcription of inflammatory mediators that accelerates MAFLD development and/or progression. In this mini review we aimed to present current evidence on the potential impact of trained immunity on the pathophysiology of MAFLD, shedding light on the complex immunobiology of the disease and providing novel potential therapeutic strategies to restrain the burden of the disease.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece.
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece; Department of Midwifery, School of Healthcare Sciences, University of West Macedonia, Koila, Kozani 50100, Macedonia, Greece
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Macedonia, Greece
| | - Apostolis Papaefthymiou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece; First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Macedonia, Greece; Pancreaticobiliary Medicine Unit, University College London Hospitals (UCLH), London W1W 6DN, UK
| | - Christos Zavos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece; Department of Midwifery, School of Healthcare Sciences, University of West Macedonia, Koila, Kozani 50100, Macedonia, Greece
| | - Dimitrios Chatzopoulos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece
| | - Elisabeth Vardaka
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece; Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, Alexander Campus, 57400 Thessaloniki, Macedonia, Greece
| | - Anthia Gatopoulou
- 2nd Department of Internal Medicine, General University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Foteini Kyrailidi
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece
| | - Maria C Mouratidou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki 54642, Macedonia, Greece; First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Macedonia, Greece; Gastroklinik, Private Gastroenterological Practice, Horgen 8810, Switzerland; Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
| |
Collapse
|
40
|
Jangid G, Popoola-Samuel HAO, Goda K, Anamika FNU, Gupta V, Kanagala SG, Munjal RS. Influence of Plant-Based Diet on the Cardiovascular System: A Narrative Review. Cardiol Rev 2023:00045415-990000000-00154. [PMID: 37768098 DOI: 10.1097/crd.0000000000000613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
A plant-based diet (PBD) includes multiple dietary patterns such as vegetarianism, veganism, lacto-ovo-vegetarianism, etc. A well-balanced PBD has positive effects on body mass index, hyperlipidemia, and type 2 diabetes, and it is associated with decreased risk of cardiovascular diseases. Due to its high fiber content, which lowers levels of low-density lipoprotein cholesterol, high antioxidant content, which prevents capillary wall damage; the presence of polyunsaturated fatty acids, which have an anti-inflammatory effect, improvement in glucose homeostasis through improved insulin sensitivity, and impact on the gut microbiota, it has been proven to be beneficial for the heart. Despite their possible benefits, PBDs can be low in essential nutrients such as vitamin B12, zinc, vitamin D, iodine, and amino acids. This review aims to analyze the influence of PBDs on the cardiovascular system and associated disease progression.
Collapse
Affiliation(s)
- Gurusha Jangid
- From Internal Medicine Department, Dr. Sampurnananda Medical College, Jodhpur, Rajasthan, India
| | | | - Kausalya Goda
- Internal Medicine Department, Sri Ramachandra Institute of Higher Education and Research, Tamil Nadu, India
| | - F N U Anamika
- Internal Medicine Department, University College of Medical Sciences, New Delhi, India
| | - Vasu Gupta
- Internal Medicine Department, Cleveland Clinic, Akron General, Akron, OH
| | | | | |
Collapse
|
41
|
Abstract
Epigenetics has transformed our understanding of the molecular basis of complex diseases, including cardiovascular and metabolic disorders. This review offers a comprehensive overview of the current state of knowledge on epigenetic processes implicated in cardiovascular and metabolic diseases, highlighting the potential of DNA methylation as a precision medicine biomarker and examining the impact of social determinants of health, gut bacterial epigenomics, noncoding RNA, and epitranscriptomics on disease development and progression. We discuss challenges and barriers to advancing cardiometabolic epigenetics research, along with the opportunities for novel preventive strategies, targeted therapies, and personalized medicine approaches that may arise from a better understanding of epigenetic processes. Emerging technologies, such as single-cell sequencing and epigenetic editing, hold the potential to further enhance our ability to dissect the complex interplay between genetic, environmental, and lifestyle factors. To translate research findings into clinical practice, interdisciplinary collaborations, technical and ethical considerations, and accessibility of resources and knowledge are crucial. Ultimately, the field of epigenetics has the potential to revolutionize the way we approach cardiovascular and metabolic diseases, paving the way for precision medicine and personalized health care, and improving the lives of millions of individuals worldwide affected by these conditions.
Collapse
Affiliation(s)
- Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, New York (A.A.B.)
| | - José Ordovás
- Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, at Tufts University, Boston, MA (J.O.)
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain (J.O.)
- Consortium CIBERObn, Instituto de Salud Carlos III (ISCIII), Madrid, Spain (J.O.)
| |
Collapse
|
42
|
Zhuo X, Luo H, Lei R, Lou X, Bian J, Guo J, Luo H, Zhang X, Jiao Q, Gong W. Association between Intestinal Microecological Changes and Atherothrombosis. Microorganisms 2023; 11:1223. [PMID: 37317197 DOI: 10.3390/microorganisms11051223] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of large- and medium-sized arteries that causes ischemic heart disease, strokes, and peripheral vascular disease, collectively called cardiovascular disease (CVD), and is the leading cause of CVD resulting in a high rate of mortality in the population. AS is pathological by plaque development, which is caused by lipid infiltration in the vessel wall, endothelial dysfunction, and chronic low-grade inflammation. Recently, more and more scholars have paid attention to the importance of intestinal microecological disorders in the occurrence and development of AS. Intestinal G-bacterial cell wall lipopolysaccharide (LPS) and bacterial metabolites, such as oxidized trimethylamine (TMAO) and short-chain fatty acids (SCFAs), are involved in the development of AS by affecting the inflammatory response, lipid metabolism, and blood pressure regulation of the body. Additionally, intestinal microecology promotes the progression of AS by interfering with the normal bile acid metabolism of the body. In this review, we summarize the research on the correlation between maintaining a dynamic balance of intestinal microecology and AS, which may be potentially helpful for the treatment of AS.
Collapse
Affiliation(s)
- Xinyu Zhuo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Hui Luo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| | - Rumei Lei
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Xiaokun Lou
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Jing Bian
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Junfeng Guo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Hao Luo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Xingwei Zhang
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| | - Qibin Jiao
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Wenyan Gong
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| |
Collapse
|
43
|
Kunika, Frey N, Rangrez AY. Exploring the Involvement of Gut Microbiota in Cancer Therapy-Induced Cardiotoxicity. Int J Mol Sci 2023; 24:7261. [PMID: 37108423 PMCID: PMC10138392 DOI: 10.3390/ijms24087261] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Trillions of microbes in the human intestinal tract, including bacteria, viruses, fungi, and protozoa, are collectively referred to as the gut microbiome. Recent technological developments have led to a significant increase in our understanding of the human microbiome. It has been discovered that the microbiome affects both health and the progression of diseases, including cancer and heart disease. Several studies have indicated that the gut microbiota may serve as a potential target in cancer therapy modulation, by enhancing the effectiveness of chemotherapy and/or immunotherapy. Moreover, altered microbiome composition has been linked to the long-term effects of cancer therapy; for example, the deleterious effects of chemotherapy on microbial diversity can, in turn, lead to acute dysbiosis and serious gastrointestinal toxicity. Specifically, the relationship between the microbiome and cardiac diseases in cancer patients following therapy is poorly understood. In this article, we provide a summary of the role of the microbiome in cancer treatment, while also speculating on a potential connection between treatment-related microbial changes and cardiotoxicity. Through a brief review of the literature, we further explore which bacterial families or genera were differentially affected in cancer treatment and cardiac disease. A deeper understanding of the link between the gut microbiome and cardiotoxicity caused by cancer treatment may help lower the risk of this critical and potentially fatal side effect.
Collapse
Affiliation(s)
- Kunika
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Ashraf Y. Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|