1
|
Kumar M, Muthurayar T, Karthika S, Gayathri S, Varalakshmi P, Ashokkumar B. Anti-Diabetic Potentials of Lactobacillus Strains by Modulating Gut Microbiota Structure and β-Cells Regeneration in the Pancreatic Islets of Alloxan-Induced Diabetic Rats. Probiotics Antimicrob Proteins 2025; 17:1096-1116. [PMID: 38329697 DOI: 10.1007/s12602-024-10221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Diabetes mellitus, a most common endocrine disorder of glucose metabolism, has become a global epidemic and poses a serious public health threat with an increased socio-economic burden. Escalating incidence of diabetes is correlated with changes in lifestyle and food habits that cause gut microbiome dysbiosis and β-cells damage, which can be addressed with dietary interventions containing probiotics. Hence, the search for probiotics of human origin with anti-diabetic, anti-AGE, and anti-ACE potentials has gained renewed interest for the effective management of diabetes and its associated complications. The present study used an alloxan (AXN)-induced diabetic rat model to investigate the effects of potential probiotic Lacticaseibacillus casei MKU1, Lactiplantibacillus pentosus MKU3, and Lactiplantibacillus plantarum MKU7 administration individually on physiochemical parameters related to diabetic pathogenesis. Experimental animals were randomly allotted into six groups viz. NCG (control), DCG (AXN), DGM (metformin), DGP1 (MKU1), DGP2 (MKU3), and DGP3 (MKU7), and biochemical data like serum glucose, insulin, AngII, ACE, HbA1c, and TNF-α levels were measured until 90 days. Our results suggest that oral administration with MKU1, MKU3, or MKU7 significantly improved serum insulin levels, glycemic control, glucose tolerance, and body weight. Additionally, β-cell mass was increased by preserving islet integrity in Lactobacillus-treated diabetic rats, whereas TNF-α (~40%), AngII (~30%), and ACE levels (~50%) were strongly inhibited and enhanced sIgA production (5.8 folds) abundantly. Furthermore, Lactobacillus administration positively influenced the gut microbiome with a significant increase in the abundance of Lactobacillus species and the beneficial Bacteroides uniformis and Bacteroides fragilis, while decreased the pathogenic Proteus vulgaris and Parabacteroides distasonis. Among the probiotic treatment groups, L. pentosus MKU3 performed greatly in almost all parameters, indicating its potential use for alleviating diabetes-associated complications.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Tharmar Muthurayar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Sukumaran Karthika
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Santhalingam Gayathri
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Perumal Varalakshmi
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Balasubramaniem Ashokkumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India.
| |
Collapse
|
2
|
Lei S, Chen T, Zhou J, Zhu L, Zhang Z, Xie X, Zhang X, Khan I, Li Z. Distinct blood and oral microbiome profiles reveal altered microbial composition and functional pathways in myocardial infarction patients. Front Cell Infect Microbiol 2025; 15:1506382. [PMID: 40297611 PMCID: PMC12034738 DOI: 10.3389/fcimb.2025.1506382] [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: 10/05/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction The blood microbiome, increasingly recognized as a distinct microbial niche, may originate partly from oral translocation. We systematically compared circulating and oral microbiome profiles between healthy individuals and myocardial infarction (MI) patients to identify disease-associated signatures. Methods The current study recruited 20 participants, including 10 healthy controls and 10 patients with MI. Blood and Saliva samples were collected from each participant to analyze the association between blood and oral microbiome in MI patients using 16S rRNA gene sequencing. Results The blood microbiome showed significantly greater alpha diversity than the oral microbiome (p<0.05), but beta diversity did not differ significantly. The blood microbiome in MI patients had higher levels of Firmicutes, Bacteroidota, Actinobacteriota, genus Bacteroides, and lower Proteobacteria, whereas the oral microbiome was dominated by Firmicutes, Bacteroidota, Veillonella, and Prevotella_7. LEfSe analysis revealed distinct blood microbial taxa-Actinobacteria in MI patients and Enterobacterales in controls. In contrast, the oral microbiota of healthy subjects was enriched in Rothia, Micrococcaceae, and Micrococcales, while no distinct taxa were associated with MI. Both blood and oral microbiomes showed significant functional pathway differences (KEGG) between groups. Additionally, microbiome signatures significantly correlated with clinical and demographic markers. Discussion Our study demonstrates that the blood harbors a distinct microbiome characterized by specific microbial taxa and functional pathways rather than merely reflecting oral bacterial translocation. These findings suggest that the circulating microbiome may play an active role in the pathology of MI. Furthermore, we identified significant associations between these microbial signatures and clinical disease markers. This highlights the potential importance of the blood microbiome in understanding the mechanisms underlying MI and its diagnostic or therapeutic implications.
Collapse
Affiliation(s)
- Shengnan Lei
- Department of Medicine, Northwest MINZU University, Lanzhou, Gansu, China
- Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, Gansu, China
- Department of Northwest Institute of Eco-Environment and Resources, University of Chinese Academy of Sciences, Beijing, China
- Department of Medicine, Gansu Provincial Key Laboratory of Oral Diseases Research, Lanzhou, China
| | - Tuo Chen
- Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, Gansu, China
- Department of Northwest Institute of Eco-Environment and Resources, University of Chinese Academy of Sciences, Beijing, China
| | - Jianye Zhou
- Department of Medicine, Northwest MINZU University, Lanzhou, Gansu, China
- Department of Medicine, Gansu Provincial Key Laboratory of Oral Diseases Research, Lanzhou, China
| | - Linghong Zhu
- Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, Gansu, China
| | - Zilong Zhang
- Department of Oncological Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Xiaodong Xie
- Department of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xu Zhang
- Department of Cardiology, General Hospital of Xizang Military Region, Lhasa, China
| | - Ikram Khan
- Department of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Zhiqiang Li
- Department of Medicine, Northwest MINZU University, Lanzhou, Gansu, China
- Department of Medicine, Gansu Provincial Key Laboratory of Oral Diseases Research, Lanzhou, China
| |
Collapse
|
3
|
Zununi Vahed S, Hejazian SM, Ardalan M, Anagnostou F, Pavon-Djavid G, Barzegari A. The impacts of dietary antioxidants on cardiovascular events in hemodialysis patients: An update on the cellular and molecular mechanisms. Nutr Rev 2025; 83:e615-e634. [PMID: 38728008 DOI: 10.1093/nutrit/nuae039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular-related complications (CVCs) are the primary cause of death in patients undergoing hemodialysis (HD), accounting for greater than half of all deaths. Beyond traditional risk factors, chronic inflammation, extreme oxidative stress (OS), and endothelial dysfunction emerge as major contributors to accelerated CVCs in HD patients. Ample evidence shows that HD patients are constantly exposed to excessive OS, due to uremic toxins and pro-oxidant molecules that overwhelm the defense antioxidant mechanisms. The present study highlights the efficiency of natural antioxidant supplementation in managing HD-induced inflammation, OS, and consequently CVCs. Moreover, it discusses the underlying molecular mechanisms by which these antioxidants can decrease mitochondrial and endothelial dysfunction and ameliorate CVCs in HD patients. Given the complex nature of OS and its molecular pathways, the utilization of specific antioxidants as a polypharmacotherapy may be necessary for targeting each dysregulated signaling pathway and reducing the burden of CVCs.
Collapse
Affiliation(s)
| | | | | | - Fani Anagnostou
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
- Service of Odontology, Hospital Pitié-Salpêtrière AP-HP, Paris, France
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM UMR-S 1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging, 93430 Villetaneuse, France
| | - Abolfazl Barzegari
- Université Sorbonne Paris Nord, INSERM UMR-S 1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging, 93430 Villetaneuse, France
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Zhu T, Chen J, Zhang M, Tang Z, Tong J, Hao X, Li H, Xu J, Yang J. Tanshinone IIA Exerts Cardioprotective Effects Through Improving Gut-Brain Axis Post-Myocardial Infarction. Cardiovasc Toxicol 2024; 24:1317-1334. [PMID: 39377990 PMCID: PMC11564317 DOI: 10.1007/s12012-024-09928-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024]
Abstract
Myocardial infarction (MI) is a lethal cardiovascular disease worldwide. Emerging evidence has revealed the critical role of gut dysbiosis and impaired gut-brain axis in the pathological progression of MI. Tanshinone IIA (Tan IIA), a traditional Chinese medicine, has been demonstrated to exert therapeutic effects for MI. However, the effects of Tan IIA on gut-brain communication and its potential mechanisms post-MI are still unclear. In this study, we initially found that Tan IIA significantly reduced myocardial inflammation, apoptosis and fibrosis, therefore alleviating hypertrophy and improving cardiac function following MI, suggesting the cardioprotective effect of Tan IIA against MI. Additionally, we observed that Tan IIA improved the gut microbiota as evidenced by changing the α-diversity and β-diversity, and reduced histopathological impairments by decreasing inflammation and permeability in the intestinal tissues, indicating the substantial improvement of Tan IIA in gut function post-MI. Lastly, Tan IIA notably reduced lipopolysaccharides (LPS) level in serum, inflammation responses in paraventricular nucleus (PVN) and sympathetic hyperexcitability following MI, suggesting that restoration of Tan IIA on MI-induced brain alterations. Collectively, these results indicated that the cardioprotective effects of Tan IIA against MI might be associated with improvement in gut-brain axis, and LPS might be the critical factor linking gut and brain. Mechanically, Tan IIA-induced decreased intestinal damage reduced LPS release into serum, and reduced serum LPS contributes to decreased neuroinflammation with PVN and sympathetic inactivation, therefore protecting the myocardium against MI-induced injury.
Collapse
Affiliation(s)
- Tong Zhu
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China
| | - Jie Chen
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Mingxia Zhang
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China
| | - Zheng Tang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Jie Tong
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiuli Hao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Hongbao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, 710061, China
| | - Jin Xu
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China.
| | - Jinbao Yang
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China.
| |
Collapse
|
5
|
Zhong X, Yan J, Wei X, Xie T, Zhang Z, Wang K, Sun C, Chen W, Zhu J, Zhao X, Wang X. Shenxiang Suhe pill improves cardiac function through modulating gut microbiota and serum metabolites in rats after acute myocardial infarction. PHARMACEUTICAL BIOLOGY 2024; 62:1-12. [PMID: 38084911 PMCID: PMC11734889 DOI: 10.1080/13880209.2023.2289577] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 09/25/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023]
Abstract
CONTEXT Shenxiang Suhe pill (SXSH), a traditional Chinese medicine, is clinically effective against coronary heart disease, but the mechanism of cardiac-protective function is unclear. OBJECTIVE We investigated the cardiac-protective mechanism of SXSH via modulating gut microbiota and metabolite profiles. MATERIALS AND METHODS Sprague-Dawley (SD) male rats were randomly divided into 6 groups (n = 8): Sham, Model, SXSH (Low, 0.063 g/kg; Medium, 0.126 g/kg; High, 0.252 g/kg), and Ato (atorvastatin, 20 mg/kg). Besides the Sham group, rats were modelled with acute myocardial infarction (AMI) by ligating the anterior descending branch of the left coronary artery (LAD). After 3, 7, 14 days' administration, ultrasound, H&E staining, serum enzymic assay, 16S rRNA sequencing were conducted to investigate the SXSH efficacy. Afterwards, five groups of rats: Sham, Model, Model-ABX (AMI with antibiotics-feeding), SXSH (0.126 g/kg), SXSH-ABX were administrated for 14 days to evaluate the gut microbiota-dependent SXSH efficacy, and serum untargeted metabolomics test was performed. RESULTS 0.126 g/kg of SXSH intervention for 14 days increased ejection fraction (EF, 78.22%), fractional shortening (FS, 109.07%), and aortic valve flow velocities (AV, 21.62%), reduced lesion area, and decreased serum LDH (8.49%) and CK-MB (10.79%). Meanwhile, SXSH upregulated the abundance of Muribaculaceae (199.71%), Allobaculum (1744.09%), and downregulated Lactobacillus (65.51%). The cardiac-protective effect of SXSH was disrupted by antibiotics administration. SXSH altered serum metabolites levels, such as downregulation of 2-n-tetrahydrothiophenecarboxylic acid (THTC, 1.73%), and lysophosphatidylcholine (lysoPC, 4.61%). DISCUSSION AND CONCLUSION The cardiac-protective effect and suggested mechanism of SXSH could provide a theoretical basis for expanding its application in clinic.
Collapse
Affiliation(s)
- Xinqin Zhong
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junyuan Yan
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xing Wei
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tian Xie
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaojian Zhang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kaiyue Wang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Congying Sun
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wei Chen
- Hangzhou Hu Qing Yu Tang Pharmaceutical Co., Ltd, Hangzhou, China
| | - Jiaming Zhu
- Hangzhou Hu Qing Yu Tang Pharmaceutical Co., Ltd, Hangzhou, China
| | - Xin Zhao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoying Wang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
6
|
Zhang Y, Wang H, Sang Y, Liu M, Wang Q, Yang H, Li X. Gut microbiota in health and disease: advances and future prospects. MedComm (Beijing) 2024; 5:e70012. [PMID: 39568773 PMCID: PMC11577303 DOI: 10.1002/mco2.70012] [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: 06/28/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
The gut microbiota plays a critical role in maintaining human health, influencing a wide range of physiological processes, including immune regulation, metabolism, and neurological function. Recent studies have shown that imbalances in gut microbiota composition can contribute to the onset and progression of various diseases, such as metabolic disorders (e.g., obesity and diabetes) and neurodegenerative conditions (e.g., Alzheimer's and Parkinson's). These conditions are often accompanied by chronic inflammation and dysregulated immune responses, which are closely linked to specific forms of cell death, including pyroptosis and ferroptosis. Pathogenic bacteria in the gut can trigger these cell death pathways through toxin release, while probiotics have been found to mitigate these effects by modulating immune responses. Despite these insights, the precise mechanisms through which the gut microbiota influences these diseases remain insufficiently understood. This review consolidates recent findings on the impact of gut microbiota in these immune-mediated and inflammation-associated conditions. It also identifies gaps in current research and explores the potential of advanced technologies, such as organ-on-chip models and the microbiome-gut-organ axis, for deepening our understanding. Emerging tools, including single-bacterium omics and spatial metabolomics, are discussed for their promise in elucidating the microbiota's role in disease development.
Collapse
Affiliation(s)
- Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Hong Wang
- School of Traditional Chinese Medicine Southern Medical University Guangzhou China
| | - Yiwei Sang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Mei Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Qing Wang
- School of Life Sciences Beijing University of Chinese Medicine Beijing China
| | - Hongjun Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs China Academy of Chinese Medical Sciences Beijing China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| |
Collapse
|
7
|
Kolobaric A, Andreescu C, Jašarević E, Hong CH, Roh HW, Cheong JY, Kim YK, Shin TS, Kang CS, Kwon CO, Yoon SY, Hong SW, Aizenstein HJ, Karim HT, Son SJ. Gut microbiome predicts cognitive function and depressive symptoms in late life. Mol Psychiatry 2024; 29:3064-3075. [PMID: 38664490 PMCID: PMC11449789 DOI: 10.1038/s41380-024-02551-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 10/05/2024]
Abstract
Depression in older adults with cognitive impairment increases progression to dementia. Microbiota is associated with current mood and cognition, but the extent to which it predicts future symptoms is unknown. In this work, we identified microbial features that reflect current and predict future cognitive and depressive symptoms. Clinical assessments and stool samples were collected from 268 participants with varying cognitive and depressive symptoms. Seventy participants underwent 2-year follow-up. Microbial community diversity, structure, and composition were assessed using high-resolution 16 S rRNA marker gene sequencing. We implemented linear regression to characterize the relationship between microbiome composition, current cognitive impairment, and depressive symptoms. We leveraged elastic net regression to discover features that reflect current or future cognitive function and depressive symptoms. Greater microbial community diversity associated with lower current cognition in the whole sample, and greater depression in participants not on antidepressants. Poor current cognitive function associated with lower relative abundance of Bifidobacterium, while greater GABA degradation associated with greater current depression severity. Future cognitive decline associated with lower cognitive function, lower relative abundance of Intestinibacter, lower glutamate degradation, and higher baseline histamine synthesis. Future increase in depressive symptoms associated with higher baseline depression and anxiety, lower cognitive function, diabetes, lower relative abundance of Bacteroidota, and lower glutamate degradation. Our results suggest cognitive dysfunction and depression are unique states with an overall biological effect detectable through gut microbiota. The microbiome may present a noninvasive readout and prognostic tool for cognitive and psychiatric states.
Collapse
Affiliation(s)
- A Kolobaric
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, USA
| | - C Andreescu
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - E Jašarević
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Magee-Womens Research Institute, Pittsburgh, USA
| | - C H Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - H W Roh
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - J Y Cheong
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Y K Kim
- Institute of MD Healthcare Inc, Seoul, Republic of Korea
| | - T S Shin
- Institute of MD Healthcare Inc, Seoul, Republic of Korea
| | - C S Kang
- Institute of MD Healthcare Inc, Seoul, Republic of Korea
| | - C O Kwon
- Institute of MD Healthcare Inc, Seoul, Republic of Korea
| | - S Y Yoon
- Institute of MD Healthcare Inc, Seoul, Republic of Korea
| | - S W Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - H J Aizenstein
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - H T Karim
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA.
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, USA.
| | - S J Son
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA.
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
8
|
Jiang W, Yan Z, Zheng X, Huang S, Hu Y, Xiong F, He B, Wu Y, Fu Q, Li Z, Zhou B. Targeting the Ferroptosis and Endoplasmic Reticulum Stress Signaling Pathways by CBX7 in Myocardial Ischemia/reperfusion Injury. Cell Biochem Biophys 2024; 82:2171-2181. [PMID: 38809351 DOI: 10.1007/s12013-024-01324-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 05/30/2024]
Abstract
Ferroptosis and endoplasmic reticulum stress (ERS) are common events in the process of myocardial ischemia/reperfusion injury (IRI). The suppression of chromobox7 (CBX7) has been reported to protect against ischemia/reperfusion injury, This research is purposed to expose the impacts and mechanism of CBX7 in myocardial IRI. CBX7 expression was detected using RT-qPCR and western blotting analysis. CCK-8 assay detected cell viability. Inflammatory response and oxidative stress were detected by ELISA, DCFH-DA probe and related assay kits. Flow cytometry analysis and caspase3 activity assay were used to detect cell apoptosis. C11-BODIPY 581/591 staining and ferro-orange staining were used to detect lipid reactive oxygen species (ROS) and Fe2+ level, respectively. Western blotting was used to detect the expression of proteins associated with apoptosis, ferroptosis and ERS. In the hypoxia/reoxygenation (H/R) model of rat cardiomyocytes H9c2, CBX7 was highly expressed. CBX7 interference significantly protected against inflammatory response, oxidative stress, apoptosis, ferroptosis and ERS induced by H/R in H9c2 cells. Moreover, after the pretreatment with ferroptosis activator erastin or ERS agonist Tunicamycin (TM), the protective effects of CBX7 knockdown on the inflammation, oxidative stress and apoptosis in H/R-induced H9c2 cells was partially abolished. To summarize, CBX7 down-regulation may exert anti-ferroptosis and anti-ERS activities to alleviate H/R-stimulated myocardial injury.
Collapse
Affiliation(s)
- Weipeng Jiang
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Zeyu Yan
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Xueou Zheng
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Shiyi Huang
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Yue Hu
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Fengjuan Xiong
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Bufan He
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Yingzhi Wu
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Qiang Fu
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Zhiliang Li
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China
| | - Baihua Zhou
- Department of Cardiology, South China Hospital of Shenzhen University, Longgang District, Shenzhen City, 518116, Guangdong, China.
| |
Collapse
|
9
|
Ramos-Regalado L, Alcover S, Badimon L, Vilahur G. The Influence of Metabolic Risk Factors on the Inflammatory Response Triggered by Myocardial Infarction: Bridging Pathophysiology to Treatment. Cells 2024; 13:1125. [PMID: 38994977 PMCID: PMC11240659 DOI: 10.3390/cells13131125] [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: 05/13/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
Myocardial infarction (MI) sets off a complex inflammatory cascade that is crucial for effective cardiac healing and scar formation. Yet, if this response becomes excessive or uncontrolled, it can lead to cardiovascular complications. This review aims to provide a comprehensive overview of the tightly regulated local inflammatory response triggered in the early post-MI phase involving cardiomyocytes, (myo)fibroblasts, endothelial cells, and infiltrating immune cells. Next, we explore how the bone marrow and extramedullary hematopoiesis (such as in the spleen) contribute to sustaining immune cell supply at a cardiac level. Lastly, we discuss recent findings on how metabolic cardiovascular risk factors, including hypercholesterolemia, hypertriglyceridemia, diabetes, and hypertension, disrupt this immunological response and explore the potential modulatory effects of lifestyle habits and pharmacological interventions. Understanding how different metabolic risk factors influence the inflammatory response triggered by MI and unraveling the underlying molecular and cellular mechanisms may pave the way for developing personalized therapeutic approaches based on the patient's metabolic profile. Similarly, delving deeper into the impact of lifestyle modifications on the inflammatory response post-MI is crucial. These insights may enable the adoption of more effective strategies to manage post-MI inflammation and improve cardiovascular health outcomes in a holistic manner.
Collapse
Affiliation(s)
- Lisaidy Ramos-Regalado
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Sebastià Alcover
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Lina Badimon
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Ciber CV, Institute Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Chair, Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08025 Barcelona, Spain (S.A.)
- Ciber CV, Institute Carlos III, 28029 Madrid, Spain
| |
Collapse
|
10
|
Wang J, Zhang H, Yuan H, Chen S, Yu Y, Zhang X, Gao Z, Du H, Li W, Wang Y, Xia P, Wang J, Song M. Prophylactic Supplementation with Lactobacillus Reuteri or Its Metabolite GABA Protects Against Acute Ischemic Cardiac Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307233. [PMID: 38487926 PMCID: PMC11095141 DOI: 10.1002/advs.202307233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/06/2023] [Indexed: 05/16/2024]
Abstract
The gut microbiome has emerged as a potential target for the treatment of cardiovascular disease. Ischemia/reperfusion (I/R) after myocardial infarction is a serious complication and whether certain gut bacteria can serve as a treatment option remains unclear. Lactobacillus reuteri (L. reuteri) is a well-studied probiotic that can colonize mammals including humans with known cholesterol-lowering properties and anti-inflammatory effects. Here, the prophylactic cardioprotective effects of L. reuteri or its metabolite γ-aminobutyric acid (GABA) against acute ischemic cardiac injury caused by I/R surgery are demonstrated. The prophylactic gavage of L. reuteri or GABA confers cardioprotection mainly by suppressing cardiac inflammation upon I/R. Mechanistically, GABA gavage results in a decreased number of proinflammatory macrophages in I/R hearts and GABA gavage no longer confers any cardioprotection in I/R hearts upon the clearance of macrophages. In vitro studies with LPS-stimulated bone marrow-derived macrophages (BMDM) further reveal that GABA inhibits the polarization of macrophages toward the proinflammatory M1 phenotype by inhibiting lysosomal leakage and NLRP3 inflammasome activation. Together, this study demonstrates that the prophylactic oral administration of L. reuteri or its metabolite GABA attenuates macrophage-mediated cardiac inflammation and therefore alleviates cardiac dysfunction after I/R, thus providing a new prophylactic strategy to mitigate acute ischemic cardiac injury.
Collapse
Affiliation(s)
- Jiawan Wang
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- Beijing Chao‐Yang HospitalDepartment of AnesthesiologyBeijing100020China
| | - Hao Zhang
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hailong Yuan
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifeng475004China
| | - Siqi Chen
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Ying Yu
- University of Chinese Academy of SciencesBeijing100049China
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyChinese Academy of SciencesBeijing100101China
| | - Xuan Zhang
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyChinese Academy of SciencesBeijing100101China
| | - Zeyu Gao
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
| | - Heng Du
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Weitao Li
- Department of ImmunologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifeng475004China
| | - Pengyan Xia
- Department of ImmunologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Jun Wang
- University of Chinese Academy of SciencesBeijing100049China
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyChinese Academy of SciencesBeijing100101China
| | - Moshi Song
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
11
|
Yi B, Zhao Y, Yan H, Li Z, Zhang P, Fang Z, Zhao Y, Yang H, Guo N. Targeted arginine metabolomics combined with metagenomics revealed the potential mechanism of Pueraria lobata extract in treating myocardial infarction. J Chromatogr A 2024; 1719:464732. [PMID: 38387153 DOI: 10.1016/j.chroma.2024.464732] [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: 12/07/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
The extraction methods for traditional Chinese medicine (TCM) may have varying therapeutic effects on diseases. Currently, Pueraria lobata (PL) is mostly extracted with ethanol, but decoction, as a TCM extraction method, is not widely adopted. In this study, we present a strategy that integrates targeted metabolomics, 16 s rDNA sequencing technology and metagenomics for exploring the potential mechanism of the water extract of PL (PLE) in treating myocardial infarction (MI). Using advanced analytical techniques like ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), we comprehensively characterized PLE's chemical composition. Further, we tested its efficacy in a rat model of MI induced by ligation of the left anterior descending branch of the coronary artery (LAD). We assessed cardiac enzyme levels and conducted echocardiograms. UPLC-MS/MS was used to compare amino acid differences in serum. Furthermore, we investigated fecal samples using 16S rDNA sequencing and metagenomic sequencing to study intestinal flora diversity and function. This study demonstrated PLE's effectiveness in reducing cardiac injury in LAD-ligated rats. Amino acid metabolomics revealed significant improvements in serum levels of arginine, citrulline, proline, ornithine, creatine, creatinine, and sarcosine in MI rats, which are key compounds in the arginine metabolism pathway. Enzyme-linked immunosorbent assay (ELISA) results showed that PLE significantly improved arginase (Arg), nitric oxide synthase (NOS), and creatine kinase (CK) contents in the liver tissue of MI rats. 16 s rDNA and metagenome sequencing revealed that PLE significantly improved intestinal flora imbalance in MI rats, particularly in taxa such as Tuzzerella, Desulfovibrio, Fournierella, Oscillibater, Harryflintia, and Holdemania. PLE also improved the arginine metabolic pathway in the intestinal microorganisms of MI rats. The findings indicate that PLE effectively modulates MI-induced arginine levels and restores intestinal flora balance. This study, the first to explore the mechanism of action of PLE in MI treatment considering amino acid metabolism and intestinal flora, expands our understanding of the potential of PL in MI treatment. It offers fresh insights into the mechanisms of PL, guiding further research and development of PL-based medicines.
Collapse
Affiliation(s)
- Bojiao Yi
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yurou Zhao
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Han Yan
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zeyu Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Pin Zhang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhengyu Fang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuping Zhao
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Na Guo
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
12
|
Ahmad AF, Caparrós-Martin JA, Gray N, Lodge S, Wist J, Lee S, O'Gara F, Dwivedi G, Ward NC. Gut microbiota and metabolomics profiles in patients with chronic stable angina and acute coronary syndrome. Physiol Genomics 2024; 56:48-64. [PMID: 37811721 DOI: 10.1152/physiolgenomics.00072.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The gut microbiota and its associated metabolites may be involved in the development and progression of CVD, although the mechanisms and impact on clinical outcomes are not fully understood. This study investigated the gut microbiome profile and associated metabolites in patients with chronic stable angina (CSA) and acute coronary syndrome (ACS) compared with healthy controls. Bacterial alpha diversity in stool from patients with ACS or CSA was comparable to healthy controls at both baseline and follow-up visits. Differential abundance analysis identified operational taxonomic units (OTUs) assigned to commensal taxa differentiating patients with ACS from healthy controls at both baseline and follow-up. Patients with CSA and ACS had significantly higher levels of trimethylamine N-oxide compared with healthy controls (CSA: 0.032 ± 0.023 mmol/L, P < 0.01 vs. healthy, and ACS: 0.032 ± 0.023 mmol/L, P = 0.02 vs. healthy, respectively). Patients with ACS had reduced levels of propionate and butyrate (119 ± 4 vs. 139 ± 5.1 µM, P = 0.001, and 14 ± 4.3 vs. 23.5 ± 8.1 µM, P < 0.001, respectively), as well as elevated serum sCD14 (2245 ± 75.1 vs. 1834 ± 45.8 ng/mL, P < 0.0001) and sCD163 levels (457.3 ± 31.8 vs. 326.8 ± 20.7 ng/mL, P = 0.001), compared with healthy controls at baseline. Furthermore, a modified small molecule metabolomic and lipidomic signature was observed in patients with CSA and ACS compared with healthy controls. These findings provide evidence of a link between gut microbiome composition and gut bacterial metabolites with CVD. Future time course studies in patients to observe temporal changes and subsequent associations with gut microbiome composition are required to provide insight into how these are affected by transient changes following an acute coronary event.NEW & NOTEWORTHY The study found discriminative microorganisms differentiating patients with acute coronary syndrome (ACS) from healthy controls. In addition, reduced levels of certain bacterial metabolites and elevated sCD14 and sCD163 were observed in patients with ACS compared with healthy controls. Furthermore, modified small molecule metabolomic and lipidomic signatures were found in both patient groups. Although it is not known whether these differences in profiles are associated with disease development and/or progression, the findings provide exciting options for potential new disease-related mechanism(s) and associated therapeutic target(s).
Collapse
Affiliation(s)
- Adilah F Ahmad
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Jose A Caparrós-Martin
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Microbiology, PathWest Laboratory Medicine, Perth, Western Australia, Australia
| | - Fergal O'Gara
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Natalie C Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
13
|
Zhang P, Fang Z, Zhao M, Yi B, Huang Y, Yang H, Guo N, Zhao C. Ethanol extract of Pueraria lobata improve acute myocardial infarction in rats via regulating gut microbiota and bile acid metabolism. Phytother Res 2023; 37:5932-5946. [PMID: 37697496 DOI: 10.1002/ptr.8005] [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: 04/13/2023] [Revised: 06/15/2023] [Accepted: 08/20/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND AND AIM Acute myocardial infarction (AMI) is a multifactorial disease with high mortality rate worldwide. Ethanol extract of Pueraria lobata (EEPL) has been widely used in treating cardiovascular diseases in China. This study aimed to explore the underlying therapeutic mechanism of EEPL in AMI rats. EXPERIMENTAL PROCEDURE We first evaluated the anti-AMI efficacy of EEPL through immunohistochemistry staining and biochemical indexes. Then, UPLC-MS/MS, 16S rDNA, and shotgun metagenomic sequencing were used to analyze the alterations in bile acid metabolism and intestinal flora. Finally, the influence of EEPL on ilem bile acid metabolism, related enzymes expression, and transporter proteins expression in rats were verified by mass spectrometry image and ELISA. KEY RESULTS The results showed that EEPL can reduce cardiac impairment in AMI rats. Besides, EEPL effectively increased bile acid levels and regulated gut microbiota disturbance in AMI rats via increasing CYP7A1 expression and restoring intestinal microbiota diversity, separately. Moreover, it can increase bile acids reabsorption and fecal excretion through inhibiting FXR-FGF15 signaling pathway and increasing OST-α expression, which associated with Lachnoclostridium. CONCLUSIONS AND IMPLICATIONS Our findings demonstrated that EEPL alleviated AMI partially by remediating intestinal dysbiosis and promoting bile acid biosynthesis, which provided new targets for AMI treatment.
Collapse
Affiliation(s)
- Pin Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhengyu Fang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Bojiao Yi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yihe Huang
- School of Public Health, Shenyang Medical College, Shenyang, China
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Na Guo
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunjie Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
14
|
Zhan C, Tang T, Wu E, Zhang Y, He M, Wu R, Bi C, Wang J, Zhang Y, Shen B. From multi-omics approaches to personalized medicine in myocardial infarction. Front Cardiovasc Med 2023; 10:1250340. [PMID: 37965091 PMCID: PMC10642346 DOI: 10.3389/fcvm.2023.1250340] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Abstract
Myocardial infarction (MI) is a prevalent cardiovascular disease characterized by myocardial necrosis resulting from coronary artery ischemia and hypoxia, which can lead to severe complications such as arrhythmia, cardiac rupture, heart failure, and sudden death. Despite being a research hotspot, the etiological mechanism of MI remains unclear. The emergence and widespread use of omics technologies, including genomics, transcriptomics, proteomics, metabolomics, and other omics, have provided new opportunities for exploring the molecular mechanism of MI and identifying a large number of disease biomarkers. However, a single-omics approach has limitations in understanding the complex biological pathways of diseases. The multi-omics approach can reveal the interaction network among molecules at various levels and overcome the limitations of the single-omics approaches. This review focuses on the omics studies of MI, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, and other omics. The exploration extended into the domain of multi-omics integrative analysis, accompanied by a compilation of diverse online resources, databases, and tools conducive to these investigations. Additionally, we discussed the role and prospects of multi-omics approaches in personalized medicine, highlighting the potential for improving diagnosis, treatment, and prognosis of MI.
Collapse
Affiliation(s)
- Chaoying Zhan
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Tong Tang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Erman Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- KeyLaboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengqiao He
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rongrong Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Bi
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- KeyLaboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jiao Wang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yingbo Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Bairong Shen
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Liu J, Zhu R, Song J, Sohaib M, Wang S, Mao J, Qi J, Xiong X, Zhou W, Guo L. Limosilactobacillus reuteri consumption significantly reduces the total cholesterol concentration without affecting other cardiovascular disease risk factors in adults: A systematic review and meta-analysis. Nutr Res 2023; 117:1-14. [PMID: 37419064 DOI: 10.1016/j.nutres.2023.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023]
Abstract
As one of the most significant probiotics, Limosilactobacillus reuteri (L. reuteri) has been exploited as a nutritional supplement. We hypothesized that L. reuteri consumption might improve the significant risk factors of cardiovascular disease, including blood pressure, blood lipid, and blood glucose. However, previous clinical studies have shown controversial results. This study aims to explore the effect of L. reuteri consumption on these risk factors. PubMed, Embase, Scopus, the Cochrane Library, and Web of Science were searched for eligible randomized controlled trials published before May 2022. A total of 6 studies with 4 different L. reuteri strains and including 512 participants were included. The results showed that L. reuteri consumption significantly reduced total cholesterol (TC) by -0.26 mmol/L compared with the control group. In contrast, it did not affect systolic blood pressure, diastolic blood pressure, fasting blood glucose, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol (LDL-C), or triglycerides. Subgroup analysis showed a significant reduction in TC when participants were <55 years old, had a body mass index between 25 and 30, or had hypercholesterolemia. In addition, TC decreased significantly when L. reuteri supplementation was >5 × 109 colony-forming unit or the length of the intervention was <12 weeks. Strain subgroup analysis showed that L. reuteri NCIMB 30242 significantly reduced TC and LDL-C. In conclusion, L. reuteri consumption has a significant TC-lowering effect, which can effectively reduce the risks of cardiovascular disease associated with hypercholesterolemia. However, the results do not support the effectiveness of L. reuteri consumption on other metabolic outcomes. Further examination of larger sample sizes is needed to confirm these findings.
Collapse
Affiliation(s)
- Jinshu Liu
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021; School of Pharmacy, Jilin University, Changchun, Jilin, China, 130021
| | - Ruiting Zhu
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Jinping Song
- Rongchang Bio-Pharmaceutical Co. Ltd., Yantai, Shandong, China, 264006
| | | | - Saikun Wang
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Jing Mao
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Jiahe Qi
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Xuance Xiong
- Medical College, Beihua University, Jilin, Jilin, China, 132013
| | - Wei Zhou
- The First Hospital of Jilin University, Changchun, Jilin, China, 130021.
| | - Lirong Guo
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021.
| |
Collapse
|
16
|
Sánchez-Quintero MJ, Delgado J, Martín Chaves L, Medina-Vera D, Murri M, Becerra-Muñoz VM, Estévez M, Crespo-Leiro MG, Paz López G, González-Jiménez A, A. G. Ranea J, Queipo-Ortuño MI, Plaza-Andrades I, Rodríguez-Capitán J, Pavón-Morón FJ, Jiménez-Navarro MF. Multi-Omics Approach Reveals Prebiotic and Potential Antioxidant Effects of Essential Oils from the Mediterranean Diet on Cardiometabolic Disorder Using Humanized Gnotobiotic Mice. Antioxidants (Basel) 2023; 12:1643. [PMID: 37627638 PMCID: PMC10451832 DOI: 10.3390/antiox12081643] [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: 07/19/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Essential oils sourced from herbs commonly used in the Mediterranean diet have demonstrated advantageous attributes as nutraceuticals and prebiotics within a model of severe cardiometabolic disorder. The primary objective of this study was to assess the influences exerted by essential oils derived from thyme (Thymus vulgaris) and oregano (Origanum vulgare) via a comprehensive multi-omics approach within a gnotobiotic murine model featuring colonic microbiota acquired from patients diagnosed with coronary artery disease (CAD) and type-2 diabetes mellitus (T2DM). Our findings demonstrated prebiotic and potential antioxidant effects elicited by these essential oils. We observed a substantial increase in the relative abundance of the Lactobacillus genus in the gut microbiota, accompanied by higher levels of short-chain fatty acids and a reduction in trimethylamine N-oxide levels and protein oxidation in the plasma. Moreover, functional enrichment analysis of the cardiac tissue proteome unveiled an over-representation of pathways related to mitochondrial function, oxidative stress, and cardiac contraction. These findings provide compelling evidence of the prebiotic and antioxidant actions of thyme- and oregano-derived essential oils, which extend to cardiac function. These results encourage further investigation into the promising utility of essential oils derived from herbs commonly used in the Mediterranean diet as potential nutraceutical interventions for mitigating chronic diseases linked to CAD and T2DM.
Collapse
Grants
- PI-0170-2018, PI-0131/2020, and PI-0245-2021 Consejería de Salud y Familias-Junta de Andalucía and European Regional Development Funds/European Social Fund
- UMA20-FEDERJA-074 Universidad de Málaga, Consejería de Economía, Conocimiento, Empresas y Universidad-Junta de Andalucía and ERDF/ESF
- ProyExcel_01009 Consejería de Transformación Económica, Industria, Conocimiento y Universidades-Junta de Andalucía and ERDF/ESF
- SEC/FEC-INV-BAS 23 Sociedad Española de Cardiología and Fundación Andaluza de Cardiología
- PT20/00101 Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación-Gobierno de España
- CB16/11/00360 CIBERCV-Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación-Gobierno de España and ERDF/ESF
- Q-2918001-E Cátedra de Terapias Avanzadas en Patología Cardiovascular, Universidad de Málaga
Collapse
Affiliation(s)
- María José Sánchez-Quintero
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Josué Delgado
- Higiene y Salud Alimentaria, Faculty of Veterinary, University of Extremadura, 10003 Cáceres, Spain;
- Instituto Universitario de Investigación de Carne y Productos Cárnicos (IPROCAR), University of Extremadura, 10003 Cáceres, Spain;
| | - Laura Martín Chaves
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
| | - Dina Medina-Vera
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Department of Dermatology and Medicine, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
- Clinical Management Unit of Mental Health, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Mora Murri
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Clinical Management Unit of Endocrinology and Nutrition, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Biomedical Research Network Center for the Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Víctor M. Becerra-Muñoz
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Mario Estévez
- Instituto Universitario de Investigación de Carne y Productos Cárnicos (IPROCAR), University of Extremadura, 10003 Cáceres, Spain;
| | - María G. Crespo-Leiro
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Service of Cardiology, Complexo Hospitalario Universitario A Coruña (CHUAC), University of A Coruña, Instituto Investigación Biomédica A Coruña (INIBIC), 15006 A Coruña, Spain
| | - Guillermo Paz López
- Bioinformatics, Common Support Structures (ECAI), IBIMA Plataforma BIONAND, 29590 Málaga, Spain; (G.P.L.); (A.G.-J.); (J.A.G.R.)
| | - Andrés González-Jiménez
- Bioinformatics, Common Support Structures (ECAI), IBIMA Plataforma BIONAND, 29590 Málaga, Spain; (G.P.L.); (A.G.-J.); (J.A.G.R.)
| | - Juan A. G. Ranea
- Bioinformatics, Common Support Structures (ECAI), IBIMA Plataforma BIONAND, 29590 Málaga, Spain; (G.P.L.); (A.G.-J.); (J.A.G.R.)
- Department of Molecular Biology and Biochemistry, Faculty of Science, University of Málaga, 29010 Málaga, Spain
- CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Isabel Queipo-Ortuño
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Intercenter Clinical Management Unit of Medical Oncology, Hospitales Universitarios Regional y Virgen de la Victoria y Centro de Investigaciones Médico Sanitarias (CIMES), 29010 Málaga, Spain
- Department of Surgical Specialties, Biochemistry, and Immunology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
| | - Isaac Plaza-Andrades
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Intercenter Clinical Management Unit of Medical Oncology, Hospitales Universitarios Regional y Virgen de la Victoria y Centro de Investigaciones Médico Sanitarias (CIMES), 29010 Málaga, Spain
| | - Jorge Rodríguez-Capitán
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Francisco Javier Pavón-Morón
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Manuel F. Jiménez-Navarro
- Biomedical Research Institute of Malaga and Nanomedicine Platform (IBIMA Plataforma BIONAND), 29590 Málaga, Spain; (M.J.S.-Q.); (L.M.C.); (D.M.-V.); (M.M.); (V.M.B.-M.); (M.I.Q.-O.); (I.P.-A.); (M.F.J.-N.)
- Heart Area, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| |
Collapse
|
17
|
Chen Y, Chen R, Wang X, Zhou Y, Hong L, Xiong N, Zhu J, Ye S, Tan X. Effect of probiotic supplementation on in-hospital mortality in patients with acute myocardial infarction: a study protocol for an open-label, randomized, controlled, superiority clinical trial. Trials 2023; 24:429. [PMID: 37355630 DOI: 10.1186/s13063-023-07443-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/08/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Recent studies have demonstrated a correlation between intestinal flora and the severity of myocardial infarction as well as post-myocardial infarction repair. However, few studies have investigated whether probiotics reduce mortality and improve cardiovascular outcomes in patients with acute myocardial infarction. In this study, we will conduct a randomized controlled trial (RCT) to evaluate the effect of probiotics on in-hospital mortality and the incidence of major adverse cardiovascular events (MACE) in patients with acute myocardial infarction (AMI). METHODS This is an open-label, randomized, controlled, superiority clinical trial involving 2594 adult patients who were diagnosed with acute myocardial infarction. Patients will be randomized to (1) receive bifidobacteria triple viable capsule (Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis) 840 mg, twice a day, plus standard treatment strategy during the hospital stay, for a maximum of 30 days, or (2) receive the standard treatment strategy and will not take the bifidobacterium triple live capsule. The primary outcome was in-hospital all-cause mortality. DISCUSSION The purpose of this clinical trial is to determine whether probiotics can reduce in-hospital mortality and improve prognosis in patients with AMI, and the results will provide evidence for probiotics as a complementary treatment for AMI. TRIAL REGISTRATION Chinese Clinical Trials Registry ChiCTR2000038797. Registered on 2 October 2020.
Collapse
Affiliation(s)
- Yequn Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), Shantou, China
| | - Rongbing Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Shantou University Medical College, Shantou, China
| | - Xin Wang
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yan Zhou
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Shantou University Medical College, Shantou, China
| | - Liekai Hong
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Chaonan Minsheng Hospital, Shantou, China
| | - Nianling Xiong
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Shantou University Medical College, Shantou, China
| | - Jinxiu Zhu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Shantou University Medical College, Shantou, China
| | - Shu Ye
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Shantou University Medical College, Shantou, China
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Xuerui Tan
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China.
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College (SUMC), Shantou, China.
- Shantou University Medical College, Shantou, China.
| |
Collapse
|
18
|
Liao J, Zhang Y, Ma C, Wu G, Zhang W. Microbiome-metabolome reveals that the Suxiao Jiuxin pill attenuates acute myocardial infarction associated with fatty acid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116529. [PMID: 37086873 DOI: 10.1016/j.jep.2023.116529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Suxiao Jiuxin pill (SJP) is a Chinese medical patent drug on the national essential drug list of China, with well-established cardiovascular protective effects in the clinic. However, the mechanisms underlying the protective effects of SJP on cardiovascular disease have not yet been elucidated clearly, especially its relationship with the gut microbiota. AIM OF THE STUDY This study aimed to investigate the cardioprotective effect of SJP against isoproterenol-induced acute myocardial infarction (AMI) by integrating the gut microbiome and metabolome. METHODS A rat model of AMI was generated using isoproterenol. Firstly, the effect of antibiotic (ABX) treatment on the blood absorption and excretion of the main components of SJP were studied. Secondly, 16S rRNA sequencing and untargeted metabolomics were used to discover the improvement of SJP treatment on gut microbiota and host metabolism in AMI rats. Finally, targeted metabolomics was used to verify the effects of SJP treatment on host metabolism in AMI rats. RESULT The results showed that ABX treatment could affect the blood absorption and fecal excretion of the main active components of SJP. At the same time, SJP can restore the richness and diversity of gut microbiota, and multiple gut microbiota (including Jeotgalicoccus, Lachnospiraceae, and Blautia) are significantly associated with fatty acids. Untargeted metabolomics also found that SJP could restore the levels of various fatty acid metabolites in serum and cecal contents (p < 0.01, FC > 1.5 and VIP >1). Targeted metabolomics further confirmed that 41, 21, and 39 fatty acids were significantly altered in serum, cecal contents, and heart samples, respectively. Interestingly, these fatty acids belong to the class of eicosanoids, and SJP can significantly downregulate these eicosanoids in AMI rats. CONCLUSION The results of this study suggest that SJP may exert its cardioprotective effects by remodeling the gut microbiota and host fatty acid metabolism.
Collapse
Affiliation(s)
- Jingyu Liao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangdong, 510006, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuhao Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chi Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Gaosong Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Weidong Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangdong, 510006, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
19
|
Zhong X, Zhao Y, Huang L, Liu J, Wang K, Gao X, Zhao X, Wang X. Remodeling of the gut microbiome by Lactobacillus johnsonii alleviates the development of acute myocardial infarction. Front Microbiol 2023; 14:1140498. [PMID: 36970663 PMCID: PMC10030800 DOI: 10.3389/fmicb.2023.1140498] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionThe gut microbial community, which can be disturbed or repaired by changes in the internal environment, contributes to the development of acute myocardial infarction (AMI). Gut probiotics play a role in microbiome remodeling and nutritional intervention post-AMI. A newly isolated Lactobacillus johnsonii strain EU03 has shown potential as a probiotic. Here, we investigated the cardioprotective function and mechanism of L. johnsonii through gut microbiome remodeling in AMI rats.MethodsA rat model of left anterior descending coronary artery ligation (LAD)-mediated AMI was assessed with echocardiography, histology, and serum cardiac biomarkers to evaluate the beneficial effects of L. johnsonii. The immunofluorescence analysis was utilized to visualize the intestinal barrier changes. Antibiotic administration model was used for assessing the gut commensals’ function in the improvement of cardiac function post-AMI. The underlying beneficial mechanism through L. johnsonii enrichment was further investigated by metagenomics and metabolomics analysis.ResultsA 28-day treatment with L. johnsonii protected cardiac function, delayed cardiac pathology, suppressed myocardial injury cytokines, and improved gut barrier integrity. The microbiome composition was reprogrammed by enhancing the abundance of L. johnsonii. Microbiome dysbiosis by antibiotics abrogated the improvement of cardiac function post-AMI by L. johnsonii. L. johnsonii enrichment caused remodeling of gut microbiome by increasing the abundance of Muribaculaceae, Lactobacillus, and decreasing Romboutsia, Clostridia UCG-014, which were correlated with cardiac traits and serum metabolic biomarkers 16,16-dimethyl-PGA2, and Lithocholate 3-O-glucuronide.ConclusionThese findings reveal that gut microbiome remodeling by L. johnsonii ameliorates the cardiac function post-AMI and might advance microbiome-targeted nutritional intervention.
Collapse
Affiliation(s)
- Xinqin Zhong
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yucui Zhao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Huang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiarui Liu
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kaiyue Wang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xiumei Gao,
| | - Xin Zhao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Xin Zhao,
| | - Xiaoying Wang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Xiaoying Wang,
| |
Collapse
|
20
|
Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. Int J Mol Sci 2023; 24:ijms24054577. [PMID: 36902008 PMCID: PMC10003569 DOI: 10.3390/ijms24054577] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) therapy brings great hope to the treatment of myocardial injuries, while extracellular vesicles may be one of the main mechanisms of its action. iPSC-derived small extracellular vesicles (iPSCs-sEVs) can carry genetic and proteinaceous substances and mediate the interaction between iPSCs and target cells. In recent years, more and more studies have focused on the therapeutic effect of iPSCs-sEVs in myocardial injury. IPSCs-sEVs may be a new cell-free-based treatment for myocardial injury, including myocardial infarction, myocardial ischemia-reperfusion injury, coronary heart disease, and heart failure. In the current research on myocardial injury, the extraction of sEVs from mesenchymal stem cells induced by iPSCs was widely used. Isolation methods of iPSCs-sEVs for the treatment of myocardial injury include ultracentrifugation, isodensity gradient centrifugation, and size exclusion chromatography. Tail vein injection and intraductal administration are the most widely used routes of iPSCs-sEV administration. The characteristics of sEVs derived from iPSCs which were induced from different species and organs, including fibroblasts and bone marrow, were further compared. In addition, the beneficial genes of iPSC can be regulated through CRISPR/Cas9 to change the composition of sEVs and improve the abundance and expression diversity of them. This review focused on the strategies and mechanisms of iPSCs-sEVs in the treatment of myocardial injury, which provides a reference for future research and the application of iPSCs-sEVs.
Collapse
|
21
|
Salazar J, Morillo V, Suárez MK, Castro A, Ramírez P, Rojas M, Añez R, D’Marco L, Chacín-González M, Bermudez V. Role of Gut Microbiome in Atherosclerosis: Molecular and Therapeutic Aspects. Curr Cardiol Rev 2023; 19:e020223213408. [PMID: 36733248 PMCID: PMC10494273 DOI: 10.2174/1573403x19666230202164524] [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: 06/26/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is one of the most relevant and prevalent cardiovascular diseases of our time. It is one of the pathological entities that increases the morbidity and mortality index in the adult population. Pathophysiological connections have been observed between atherosclerosis and the gut microbiome (GM), represented by a group of microorganisms that are present in the gut. These microorganisms are vital for metabolic homeostasis in humans. Recently, direct and indirect mechanisms through which GM can affect the development of atherosclerosis have been studied. This has led to research into the possible modulation of GM and metabolites as a new target in the prevention and treatment of atherosclerosis. The goal of this review is to analyze the physiopathological mechanisms linking GM and atherosclerosis that have been described so far. We also aim to summarize the recent studies that propose GM as a potential target in atherosclerosis management.
Collapse
Affiliation(s)
- Juan Salazar
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Valery Morillo
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - María K Suárez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Ana Castro
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Paola Ramírez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Milagros Rojas
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición. Hospital General Universitario Gregorio Marañón, Madrid, España
| | - Luis D’Marco
- Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, 46115, Spain
| | | | - Valmore Bermudez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| |
Collapse
|
22
|
Ma LT, Lian JX, Bai Y, Shang MJ, Zhang ZZ, Wu FF, Chen J, Meng XB, Zheng J, Li T, Li YQ, Wang JJ. Adeno-associated virus vector intraperitoneal injection induces colonic mucosa and submucosa transduction and alters the diversity and composition of the faecal microbiota in rats. Front Cell Infect Microbiol 2022; 12:1028380. [PMID: 36619753 PMCID: PMC9813966 DOI: 10.3389/fcimb.2022.1028380] [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: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Viral vector technology, especially recombinant adeno-associated virus vector (rAAV) technology, has shown great promise in preclinical research for clinical applications. Several studies have confirmed that rAAV can successfully transduce the enteric nervous system (ENS), and rAAV gene therapy has been approved by the Food and Drug Administration (FDA) for the treatment of the early childhood blindness disease Leber congenital amaurosis and spinal muscular atrophy (SMA). However, until now, it has not been possible to determine the effect of AAV9 on intestinal microbiota. METHODS We examined the efficiency of AAV9-mediated ascending colon, transverse colon and descending colon transduction through intraperitoneal (IP) injection, performed 16S rRNA gene amplicon sequencing and analysed specific faecal microbial signatures following AAV9 IP injection via bioinformatics methods in Sprague-Dawley (SD) rats. RESULTS Our results showed (1) efficient transduction of the mucosa and submucosa of the ascending, transverse, and descending colon following AAV9 IP injection; (2) a decreased alpha diversity and an altered overall microbial composition following AAV9 IP injection; (3) significant enrichments in a total of 5 phyla, 10 classes, 13 orders, 15 families, 29 genera, and 230 OTUs following AAV9 IP injection; and (4) AAV9 can significantly upregulate the relative abundance of anaerobic microbiota which is one of the seven high-level phenotypes that BugBase could predict. CONCLUSION In summary, these data show that IP injection of AAV9 can successfully induce the transduction of the colonic mucosa and submucosa and alter the diversity and composition of the faecal microbiota in rats.
Collapse
Affiliation(s)
- Li-Tian Ma
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jing-Xuan Lian
- Department of Endocrinology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Meng-Juan Shang
- Department of Radiation Biology, Faculty of Preventive Medicine, Fourth Military Medical University, Xi'an, ShaanXi, China
| | - Zhe-Zhe Zhang
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Fei-Fei Wu
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University, Xi’an, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Xian-Bo Meng
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jin Zheng
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
- Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Haikou, China
- Department of Anatomy, College of Basic Medicine, Dali University, Dali, China
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
23
|
Khan I, Khan I, Usman M, Xiao Wei Z, Ping X, Khan S, Khan F, Jianye Z, Zhiqiang L, Lizhe A. Circulating microbiota and metabolites: Insights into cardiovascular diseases. J Clin Lab Anal 2022; 36:e24779. [DOI: 10.1002/jcla.24779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/10/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Affiliation(s)
- Ikram Khan
- Department of Microbiology, School of Life Sciences Lanzhou University Lanzhou Gansu China
- School of Stomatology Northwest Minzu University Lanzhou Gansu China
| | - Imran Khan
- Department of Microbiology Khyber Medical University Peshawar Peshawar Khyber Pakhtunkhwa Pakistan
| | - Muhammad Usman
- State Key Laboratory of Grassland Agro‐ecosystem, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Ruler Affairs, Collegeof Pastoral Agriculture Sciences and Technology Lanzhou University Lanzhou Gansu China
| | - Zhang Xiao Wei
- Department of Cardiology Lanzhou University Second Hospital Lanzhou Gansu China
| | - Xie Ping
- Department of Cardiology Gansu Provincial Hospital Lanzhou China
| | - Sarmir Khan
- Department of Reproductive Medicine, Academy of Medical Sciences The First Affiliated Hospital of Zheng University Zhengzhou Henan China
| | - Feroz Khan
- Department of Zoology, Wildlife, and Fisheries PirMehr Ali Shah Arid Agriculture University Rawalpindi Pakistan
| | - Zhou Jianye
- School of Stomatology Northwest Minzu University Lanzhou Gansu China
| | - Li Zhiqiang
- School of Stomatology Northwest Minzu University Lanzhou Gansu China
| | - An Lizhe
- Department of Microbiology, School of Life Sciences Lanzhou University Lanzhou Gansu China
| |
Collapse
|
24
|
Ma J, Lei P, Chen H, Wang L, Fang Y, Yan X, Yang Q, Peng B, Jin L, Sun D. Advances in lncRNAs from stem cell-derived exosome for the treatment of cardiovascular diseases. Front Pharmacol 2022; 13:986683. [PMID: 36147326 PMCID: PMC9486024 DOI: 10.3389/fphar.2022.986683] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality globally. Benefiting from the advantages of early diagnosis and precision medicine, stem cell-based therapies have emerged as promising treatment options for CVDs. However, autologous or allogeneic stem cell transplantation imposes a potential risk of immunological rejection, infusion toxicity, and oncogenesis. Fortunately, exosome can override these limitations. Increasing evidence has demonstrated that long non-coding RNAs (lncRNAs) in exosome from stem cell paracrine factors play critical roles in stem cell therapy and participate in numerous regulatory processes, including transcriptional silencing, transcriptional activation, chromosome modification, and intranuclear transport. Accordingly, lncRNAs can treat CVDs by directly acting on specific signaling pathways. This mini review systematically summarizes the key regulatory actions of lncRNAs from different stem cells on myocardial aging and apoptosis, ischemia-reperfusion injury, retinopathy, atherosclerosis, and hypertension. In addition, the current challenges and future prospects of lncRNAs treatment for CVDs are discussed.
Collapse
Affiliation(s)
- Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Haojie Chen
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Lei Wang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
| | - Xiaoqing Yan
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Bo Peng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
- *Correspondence: Da Sun, ; Libo Jin,
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, China
- *Correspondence: Da Sun, ; Libo Jin,
| |
Collapse
|
25
|
Khan I, Khan I, Kakakhel MA, Xiaowei Z, Ting M, Ali I, Fei Y, Jianye Z, Zhiqiang L, Lizhe A. Comparison of Microbial Populations in the Blood of Patients With Myocardial Infarction and Healthy Individuals. Front Microbiol 2022; 13:845038. [PMID: 35694288 PMCID: PMC9176212 DOI: 10.3389/fmicb.2022.845038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/31/2022] [Indexed: 01/03/2023] Open
Abstract
Increased bacterial translocation in the gut and bloodstream infections are both major comorbidities of heart failure and myocardial infarction (MI). However, the alterations in the microbiome of the blood of patients with MI remain unclear. To test this hypothesis, we conducted this case-control study to explore the microbiota compositions in the blood of Chinese patients with MI. Using high-throughput Illumina HiSeq sequencing targeting the V3–V4 region of the 16S ribosomal RNA (rRNA) gene, the microbiota communities in the blood of 29 patients with MI and 29 healthy controls were examined. In addition, the relationship between the blood microbiome and clinical features of MI was investigated. This study revealed a significant reduction in alpha diversity (Shannon index) in the MI group compared with the healthy controls. Also, a significant difference was detected in the structure and richness between the patients with MI and healthy controls. The members of the phylum Actinobacteria, class Actinobacteria, order Bifdobacteriales, family Bifidobacteriaceae, and genus Bifidobacterium were significantly abundant in the MI group, while the members of the phylum Bacteroidetes, class Bacteroidia, and order Bacteroidales were significantly enriched in the healthy controls (p < 0.05). Moreover, the functional analysis revealed a significant variation between both groups. For instance, the enrichment of genes involved in the metabolism pathways of three amino acids decreased, that is, nucleotide transport and metabolism, coenzyme transport and metabolism, and lipid transport and metabolism, among others. Our study will contribute to a better knowledge of the microbiota of blood, which will further lead to improved MI diagnosis and therapy. Further study is needed to determine the role of the blood microbiota in human health and disease.
Collapse
Affiliation(s)
- Ikram Khan
- School of Life Sciences, Lanzhou University, Lanzhou, China
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Imran Khan
- Department of Microbiology, Khyber Medical University Peshawar, Peshawar, Pakistan
| | | | | | - Mao Ting
- Lanzhou University Second Hospital, Lanzhou, China
| | - Ikram Ali
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yu Fei
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Zhou Jianye
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Li Zhiqiang
- School of Stomatology, Northwest Minzu University, Lanzhou, China
- *Correspondence: Li Zhiqiang
| | - An Lizhe
- School of Life Sciences, Lanzhou University, Lanzhou, China
- An Lizhe
| |
Collapse
|
26
|
Khan I, Khan I, Jianye Z, Xiaohua Z, Khan M, Hilal MG, Kakakhel MA, Mehmood A, Lizhe A, Zhiqiang L. Exploring blood microbial communities and their influence on human cardiovascular disease. J Clin Lab Anal 2022; 36:e24354. [PMID: 35293034 PMCID: PMC8993628 DOI: 10.1002/jcla.24354] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 01/03/2023] Open
Abstract
Background Cardiovascular disease (CVD) is the single biggest contributor to global mortality. CVD encompasses multiple disorders, including atherosclerosis, hypertension, platelet hyperactivity, stroke, hyperlipidemia, and heart failure. In addition to traditional risk factors, the circulating microbiome or the blood microbiome has been analyzed recently in chronic inflammatory diseases, including CVD in humans. Methods For this review, all relevant original research studies were assessed by searching in electronic databases, including PubMed, Google Scholar, and Web of Science, by using relevant keywords. Results This review demonstrated that elevated markers of systemic bacterial exposure are associated with noncommunicable diseases, including CVD. Studies have shown that the bacterial DNA sequence found in healthy blood belongs mainly to the Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria phyla. In cardiac events, such as stroke, coronary heart disease, and myocardial infarction, the increased proportion of Proteobacteria and Actinobacteria phyla was found. Lipopolysaccharides are a major component of Proteobacteria, which play a key role in the onset of CVD. Moreover, recently, a study reported the lower cholesterol‐degrading bacteria, including Caulobacterales order and Caulobacteraceae family were both considerably reduced in myocardial infarction. Conclusion Proteobacteria and Actinobacteria were shown to be independent markers of the risk of CVD. This finding is evidence for the new concept of the role played by blood microbiota dysbiosis in CVD. However, the association between blood microbiota and CVD is still inconsistent. Thus, more deep investigations are required in future to fully understand the role of the bacteria community in causing and preventing CVD.
Collapse
Affiliation(s)
- Ikram Khan
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Imran Khan
- Department of Microbiology, Khyber Medical University Peshawar, Peshawar, Pakistan
| | - Zhou Jianye
- Key Laboratory of Oral Diseases of Gansu Province, School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Zhang Xiaohua
- Key Laboratory of Oral Diseases of Gansu Province, School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Murad Khan
- Department of Genetics, Hebei Key Laboratory Animal, Hebei Medical University, Shijiazhuang, China
| | - Mian Gul Hilal
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | | | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - An Lizhe
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Li Zhiqiang
- Key Laboratory of Oral Diseases of Gansu Province, School of Stomatology, Northwest Minzu University, Lanzhou, China
| |
Collapse
|
27
|
An N, Zhang G, Li Y, Yuan C, Yang F, Zhang L, Gao Y, Xing Y. Promising Antioxidative Effect of Berberine in Cardiovascular Diseases. Front Pharmacol 2022; 13:865353. [PMID: 35321323 PMCID: PMC8936808 DOI: 10.3389/fphar.2022.865353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
Berberine (BBR), an important quaternary benzylisoquinoline alkaloid, has been used in Chinese traditional medicine for over 3,000 years. BBR has been shown in both traditional and modern medicine to have a wide range of pharmacological actions, including hypoglycemic, hypolipidemic, anti-obesity, hepatoprotective, anti-inflammatory, and antioxidant activities. The unregulated reaction chain induced by oxidative stress as a crucial mechanism result in myocardial damage, which is involved in the pathogenesis and progression of many cardiovascular diseases (CVDs). Numerous researches have established that BBR protects myocardium and may be beneficial in the treatment of CVDs. Given that the pivotal role of oxidative stress in CVDs, the pharmacological effects of BBR in the treatment and/or management of CVDs have strongly attracted the attention of scholars. Therefore, this review sums up the prevention and treatment mechanisms of BBR in CVDs from in vitro, in vivo, and finally to the clinical field trials timely. We summarized the antioxidant stress of BBR in the management of coronary atherosclerosis and myocardial ischemia/reperfusion; it also analyzes the pathogenesis of oxidative stress in arrhythmia and heart failure and the therapeutic effects of BBR. In short, BBR is a hopeful drug candidate for the treatment of CVDs, which can intervene in the process of CVDs from multiple angles and different aspects. Therefore, if we want to apply it to the clinic on a large scale, more comprehensive, intensive, and detailed researches are needed to be carried out to clarify the molecular mechanism and targets of BBR.
Collapse
Affiliation(s)
- Na An
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guoxia Zhang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yingjian Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chao Yuan
- Dezhou Second People’s Hospital, Dezhou, China
| | - Fan Yang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lijing Zhang
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
28
|
Yang L, Wang T, Zhang X, Zhang H, Yan N, Zhang G, Yan R, Li Y, Yu J, He J, Jia S, Wang H. Exosomes derived from human placental mesenchymal stem cells ameliorate myocardial infarction via anti-inflammation and restoring gut dysbiosis. BMC Cardiovasc Disord 2022; 22:61. [PMID: 35172728 PMCID: PMC8851843 DOI: 10.1186/s12872-022-02508-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Myocardial infarction (MI) represents a severe cardiovascular disease with limited therapeutic agents. This study was aimed to elucidate the role of the exosomes derived from human placental mesenchymal stem cells (PMSCs-Exos) in MI. METHODS PMSCs were isolated and cultured in vitro, with identification by both transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). To further investigate the effects of PMSC-Exos on MI, C57BL/6 mice were randomly divided into Sham group, MI group, and PMSC-Exos group. After 4 weeks of the intervention, cardiac function was assessed by cardiac echocardiography, electrocardiogram and masson trichrome staining; lipid indicators were determined by automatic biochemical instrument; inflammatory cytokines were measured by cytometric bead array (CBA); gut microbiota, microbial metabolites short chain fatty acids (SCFAs) as well as lipopolysaccharide (LPS) were separately investigated by 16S rRNA high throughput sequencing, gas chromatography mass spectrometry (GC-MS) and tachypleus amebocyte lysate kit; transcriptome analysis was used to test the transcriptional components (mRNA\miRNA\cirRNA\lncRNA) of PMSC-Exos. RESULTS We found that human PMSC-Exos were obtained and identified with high purity and uniformity. MI model was successfully established. Compared to MI group, PMSC-Exos treatment ameliorated myocardial fibrosis and left ventricular (LV) remodeling (P < 0.05). Moreover, PMSC-Exos treatment obviously decreased MI molecular markers (AST/BNP/MYO/Tn-I/TC), pro-inflammatory indicators (IL-1β, IL-6, TNF-α, MCP-1), as well as increased HDL in comparison with MI group (all P < 0.05). Intriguingly, PMSC-Exos intervention notably modulated gut microbial community via increasing the relative abundances of Bacteroidetes, Proteobacteria, Verrucomicrobia, Actinobacteria, Akkermansia, Bacteroides, Bifidobacterium, Thauera and Ruminiclostridium, as well as decreasing Firmicutes (all P < 0.05), compared with MI group. Furthermore, PMSC-Exos supplementation increased gut microbiota metabolites SCFAs (butyric acid, isobutyric acid and valeric acid) and decreased LPS in comparison with MI group (all P < 0.05). Correlation analysis indicated close correlations among gut microbiota, microbial SCFAs and inflammation in MI. CONCLUSIONS Our study highlighted that PMSC-Exos intervention alleviated MI via modulating gut microbiota and suppressing inflammation.
Collapse
Affiliation(s)
- Libo Yang
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Ting Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Hua Zhang
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Ning Yan
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Guoshan Zhang
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Ru Yan
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Yiwei Li
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| | - Jingjing Yu
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jun He
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Shaobin Jia
- Clinical Medical College, Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Heart Centre and Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, 750004 Ningxia China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004 China
| | - Hao Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004 Ningxia China
| |
Collapse
|
29
|
Moldovan DC, Ismaiel A, Fagoonee S, Pellicano R, Abenavoli L, Dumitrascu DL. Gut microbiota and cardiovascular diseases axis. Minerva Med 2022; 113:189-199. [PMID: 33969961 DOI: 10.23736/s0026-4806.21.07527-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gut microbiota, a term that includes microorganisms present in the gastrointestinal tract, has become very attractive lately due to its propensity to act as a virtual organ with endocrine functions, generating various bio-active metabolites, while playing an important role in human health and diseases, including cardiovascular diseases (CVDs). Focusing on the latter field, gastrointestinal dysbiosis that is the imbalance in the gut microbiota composition has been linked to various pathologies such as hypertension, atherosclerosis, myocardial infarction and heart failure. Several pathways were demonstrated to play a role in the complex and intertwined association between the gut microbiota and host, including metabolic endotoxemia, alteration of pattern recognition receptors and short-chain fatty acids, uremic toxins, bile acids and trimethylamine-N-oxide levels, leading to CVDs. Understanding these pathways can allow the identification of metabolites that could be useful predictors for detecting incipient CVDs stages and potential therapeutic targets. In this review, we summarize the pathways associating the gut microbiota with CVDs.
Collapse
Affiliation(s)
- Dora C Moldovan
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine, Regional Institute of Gastroenterology and Hepatology Prof. Dr. "O. Fodor", Cluj-Napoca, Romania
| | - Abdulrahman Ismaiel
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania -
- Second Department of Internal Medicine, Cluj-Napoca, Romania
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging, National Research Council, Molecular Biotechnology Center, Turin, Italy
| | - Rinaldo Pellicano
- Unit of Gastroenterology, Molinette Hospital, Città della Salute e della Scienza, Turin, Italy
| | - Ludovico Abenavoli
- Department of Health Sciences, The Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Dan L Dumitrascu
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Second Department of Internal Medicine, Cluj-Napoca, Romania
| |
Collapse
|
30
|
Ten-year changes in plasma L-carnitine levels and risk of coronary heart disease. Eur J Nutr 2021; 61:1353-1362. [PMID: 34799774 DOI: 10.1007/s00394-021-02713-x] [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: 03/11/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE L-Carnitine is abundant in animal source foods, particularly red meat, and circulating L-carnitine may be related to the incidence of coronary heart disease (CHD). We investigated whether long-term changes in plasma L-carnitine over 10 years were associated with the CHD incidence and also examined joint associations of carnitine-rich red meat consumption and L-carnitine changes on the subsequent risk of CHD. METHODS This prospective nested case-control study included 772 healthy women at baseline (386 incident CHD cases and 386 healthy controls). Plasma L-carnitine levels were measured both at the first (1989-90) and second blood collections (2000-02). Incident cases of CHD were prospectively followed from the date of the second blood collection through 2016. RESULTS Overall, a greater increase in L-carnitine from the first to the second time point was related to a higher risk of CHD, regardless of the initial L-carnitine levels (relative risk: 1.36 (95% CI 0.999, 1.84) per 1-SD increase). The 10-year changes in L-carnitine were positively associated with red meat consumption over time, and women with higher red meat intake (≥ 36 g/day) and with greater increases in L-carnitine had a 1.86 (95% CI 1.13, 3.09) times increased risk of CHD, as compared to those with lower red meat intake and lesser increases in L-carnitine. CONCLUSION Long-term increases in L-carnitine levels were associated with the subsequent incidence of CHD, especially among women with higher intake of red meat. Our results suggest the importance of atherogenic L-carnitine changes and dietary intakes over time in the prevention of CHD.
Collapse
|
31
|
Bai H, Gu RJ, Chen LY, Qian Y, Yu ML, Xu SL, Xia XF, Liu YC, Zhang HR, Gu YH, Lu SF. Electroacupuncture interventions alleviates myocardial ischemia reperfusion injury through regulating gut microbiota in rats. Microvasc Res 2021; 138:104235. [PMID: 34453991 DOI: 10.1016/j.mvr.2021.104235] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/10/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022]
Abstract
Electroacupuncture (EA) intervention has a remarkable cardioprotection against myocardial ischemia reperfusion injury (MIRI). Recently, it has been suggested that the gut microbiota plays an important role in regulating the progression and prognosis of MIRI. The purpose of this study was to illustrate the relationship between gut microbiota and cardioprotection of EA on MIRI. We conducted a MIRI model by ligating the left anterior descending coronary artery for 30 min followed by reperfusion in male Sprague Dawley rats, which then received 7 days of EA intervention. Echocardiography was employed to evaluate left ventricular function. Fecal samples were collected for microbial analysis by 16S rDNA high-throughput sequencing. Blood samples and myocardium were collected for inflammatory cytokine detection by enzyme linked immunosorbent assay (ELISA) and Western blot. Hematoxylin & eosin (HE) staining and immunofluorescence of ileum tissue were performed for intestinal damage evaluation. After 7 days of EA intervention, the left ventricular function was improved with significantly increased ejection fraction and fractional shortening. Furthermore, we found that EA intervention reversed the changed gut microbiota induced by MIRI, including Clostridiales, RF39, S24-7, Desulfovibrio, and Allobaculum, improved the impaired gut barrier, reduced the production and circulation of lipopolysaccharide (LPS), inhibited the level of interleukin 6 (IL-6) and interleukin 12 (IL-12) in periphery and decreased the expression of Toll like receptor 4 (TLR4) and IL-6 in myocardium. EA intervention could improve the impaired gut mucosal barrier and reduce the production and circulation of LPS after MIRI through regulating gut microbiota, thus inhibiting the circulation and myocardium inflammation and finally exerted the cardioprotective effect.
Collapse
Affiliation(s)
- Hua Bai
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ren-Jun Gu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li-Yao Chen
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi Qian
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mei-Ling Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sen-Lei Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xue-Feng Xia
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu-Chen Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hong-Ru Zhang
- School of Basic Medical Sciences, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi-Huang Gu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sheng-Feng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
32
|
Yang Y, Li X, Chen S, Xiao M, Liu Z, Li J, Cheng Y. Mechanism and therapeutic strategies of depression after myocardial infarction. Psychopharmacology (Berl) 2021; 238:1401-1415. [PMID: 33594503 DOI: 10.1007/s00213-021-05784-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 02/04/2021] [Indexed: 01/08/2023]
Abstract
Depression resulted as an important factor associated with the myocardial infarction (MI) prognosis. Patients with MI also have a higher risk for developing depression. Although the issue of depression after MI has become a matter of clinical concern, the molecular mechanism underlying depression after MI remains unclear, whereby several strategies suggested have not got ideal effects, such as selective serotonin reuptake inhibitors. In this review, we summarized and discussed the occurrence mechanism of depression after MI, such as 5-hydroxytryptamine (5-HT) dysfunction, altered hypothalamus-pituitary-adrenal (HPA) axis function, gut microbiota imbalance, exosomal signal transduction, and inflammation. In addition, we offered a succinct overview of treatment, as well as some promising molecules especially from natural products for the treatment of depression after MI.
Collapse
Affiliation(s)
- Ying Yang
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Xuping Li
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Sixuan Chen
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Mingzhu Xiao
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Zhongqiu Liu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jingyan Li
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| | - Yuanyuan Cheng
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
33
|
Moludi J, Saiedi S, Ebrahimi B, Alizadeh M, Khajebishak Y, Ghadimi SS. Probiotics Supplementation on Cardiac Remodeling Following Myocardial Infarction: a Single-Center Double-Blind Clinical Study. J Cardiovasc Transl Res 2021; 14:299-307. [PMID: 32681453 DOI: 10.1007/s12265-020-10052-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/14/2020] [Indexed: 12/16/2022]
Abstract
Adverse cardiac remodeling after myocardial infarction (MI) can lead to the syndrome of heart failure (HF). Recently, changes in gut microbiota composition (dysbiosis) have appeared as a novel candidate that may be linked to the development of CR and HF. The aim of this trial was to evaluate the effects of probiotics administration on attenuating CR in patients with MI. A single-center double-blind placebo-controlled stratified randomized clinical study was conducted in 44 subjects with MI who underwent percutaneous coronary intervention (PCI). Patients were randomly assigned to take, with lunch, either a probiotic capsule containing 1.6 × 109 colony-forming unit (CFU) of bacteria (treatment group) or capsules contained inulin (control group) over 3 months. CR biomarkers (including serum procollagen III, transforming growth factor beta (TGF-β), trimethylamine N-oxide (TMAO), and matrix metallopeptidase 9 (MMP-9)) were assessed. Echocardiography results were measured at baseline and after the intervention. Significant decreases were seen in serum TGF-β concentrations (- 8.0 ± 2.1 vs. - 4.01 ± 1.8 pg/mL, p = 0.001) and TMAO levels (- 17.43 ± 10.20 vs. - 4.54 ± 8.7 mmol/L, p = 0.043), and there were no differences were seen in MMP-9 (- 4.1 ± 0.12 vs. - 4.01 + 0.15 nmol/mL, p = 0.443) and procollagen III levels (- 1.35 ± 0.70 vs. 0.01 + 0.3 mg/L, p = 0.392) subsequent to probiotics supplementation compared with the placebo group. Improvements in echocardiographic indices were also greater in the probiotics group as compared with that in the control group, but not at a significant level. Regression analysis revealed that baseline left ventricular ejection fraction (LVEF), and changes of procollagen III, predicted 62% of the final LVEF levels. Probiotics administration may have a beneficial effect on the cardiac remodeling process in patients with myocardial infarction. Iranian Registry of Clinical Trials (IRCT): IRCT20121028011288N15.
Collapse
Affiliation(s)
- Jalal Moludi
- School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Somaieh Saiedi
- Nutrition Research Center, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Alizadeh
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Sevda Saleh Ghadimi
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Choi J, Jung HT, Ferrell A, Woo S, Haddad L. Machine Learning-Based Nicotine Addiction Prediction Models for Youth E-Cigarette and Waterpipe (Hookah) Users. J Clin Med 2021; 10:jcm10050972. [PMID: 33801175 PMCID: PMC7957622 DOI: 10.3390/jcm10050972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 01/09/2023] Open
Abstract
Despite the harmful effect on health, e-cigarette and hookah smoking in youth in the U.S. has increased. Developing tailored e-cigarette and hookah cessation programs for youth is imperative. The aim of this study was to identify predictor variables such as social, mental, and environmental determinants that cause nicotine addiction in youth e-cigarette or hookah users and build nicotine addiction prediction models using machine learning algorithms. A total of 6511 participants were identified as ever having used e-cigarettes or hookah from the National Youth Tobacco Survey (2019) datasets. Prediction models were built by Random Forest with ReliefF and Least Absolute Shrinkage and Selection Operator (LASSO). ReliefF identified important predictor variables, and the Davies–Bouldin clustering evaluation index selected the optimal number of predictors for Random Forest. A total of 193 predictor variables were included in the final analysis. Performance of prediction models was measured by Root Mean Square Error (RMSE) and Confusion Matrix. The results suggested high performance of prediction. Identified predictor variables were aligned with previous research. The noble predictors found, such as ‘witnessed e-cigarette use in their household’ and ‘perception of their tobacco use’, could be used in public awareness or targeted e-cigarette and hookah youth education and for policymakers.
Collapse
Affiliation(s)
- Jeeyae Choi
- School of Nursing, University of North Carolina, Wilmington, NC 28403, USA; (A.F.); (S.W.); (L.H.)
- Correspondence: ; Tel.: +1-910-962-2487
| | - Hee-Tae Jung
- College of Information and Computer Science, University of Massachusetts, Amherst, MA 01002, USA;
| | - Anastasiya Ferrell
- School of Nursing, University of North Carolina, Wilmington, NC 28403, USA; (A.F.); (S.W.); (L.H.)
| | - Seoyoon Woo
- School of Nursing, University of North Carolina, Wilmington, NC 28403, USA; (A.F.); (S.W.); (L.H.)
| | - Linda Haddad
- School of Nursing, University of North Carolina, Wilmington, NC 28403, USA; (A.F.); (S.W.); (L.H.)
| |
Collapse
|
35
|
Song T, Guan X, Wang X, Qu S, Zhang S, Hui W, Men L, Chen X. Dynamic modulation of gut microbiota improves post-myocardial infarct tissue repair in rats via butyric acid-mediated histone deacetylase inhibition. FASEB J 2021; 35:e21385. [PMID: 33565193 DOI: 10.1096/fj.201903129rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/03/2021] [Accepted: 01/07/2021] [Indexed: 02/02/2023]
Abstract
The complex and dynamic population of gut microbiota exerts a marked influence on the host during homeostasis and disease. Imbalance of gut microbiota metabolites may lead to cardiac dysfunction in patients with heart failure, which is related to myocardial infarction(MI) severity. However, the role of gut microbiota in the repair process after MI has rarely been reported. To explore the role of gut microbiota in MI repair and its underlying mechanism, we mixed antibiotics in drinking water to interfere with gut microbiota in rats. Hematoxylin and eosin staining, Sirius red staining, western blotting, and immunohistochemistry were used to detect tissue repair and fibrosis. We found that the expressions of alpha-smooth muscle actin, collagen, and histone deacetylase (HDAC) activities were significantly increased. We detected gut microbiota at different time points after MI using 16S ribosomal RNA sequencing and detected that Prevotellaceae, Clostridiaceae, and Lachnospiraceae were significantly altered among the butyric acid producers. We administered sodium butyrate via drinking water and discovered that sodium butyrate reduced HDAC activities and adverse repair. Therefore, we speculated that gut microbiota influences the acetylation level and tissue repair process after MI by affecting butyric acid production.
Collapse
Affiliation(s)
- Tongtong Song
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xin Guan
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xuan Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Shanshan Qu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Siwei Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wenting Hui
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lihui Men
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
36
|
Microbiota, a New Playground for the Omega-3 Polyunsaturated Fatty Acids in Cardiovascular Diseases. Mar Drugs 2021; 19:md19020054. [PMID: 33498729 PMCID: PMC7931107 DOI: 10.3390/md19020054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Several cardioprotective mechanisms attributed to Omega-3 polyunsaturated fatty acids (PUFAs) have been studied and widely documented. However, in recent years, studies have supported the concept that the intestinal microbiota can play a much larger role than we had anticipated. Microbiota could contribute to several pathologies, including cardiovascular diseases. Indeed, an imbalance in the microbiota has often been reported in patients with cardiovascular disease and produces low-level inflammation. This inflammation contributes to, more or less, long-term development of cardiovascular diseases. It can also worsen the symptoms and the consequences of these pathologies. According to some studies, omega-3 PUFAs in the diet could restore this imbalance and mitigate its harmful effects on cardiovascular diseases. Many mechanisms are involved and included: (1) a reduction of bacteria producing trimethylamine (TMA); (2) an increase in bacteria producing butyrate, which has anti-inflammatory properties; and (3) a decrease in the production of pro-inflammatory cytokines. Additionally, omega-3 PUFAs would help maintain better integrity in the intestinal barrier, thereby preventing the translocation of intestinal contents into circulation. This review will summarize the effects of omega-3 PUFAs on gut micro-biota and the potential impact on cardiac health.
Collapse
|
37
|
Moon JM, Lee HJ, Han K, Kim DH, Hong SW, Soh H, Park S, Kang EA, Lee J, Koh SJ, Im JP, Kim JS. Occult Blood in Feces Is Associated With an Increased Risk of Ischemic Stroke and Myocardial Infarction: A Nationwide Population Study. J Am Heart Assoc 2020; 10:e017783. [PMID: 33372535 PMCID: PMC7955497 DOI: 10.1161/jaha.120.017783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Although occult hemoglobin in feces is universally valued as a screening tool for colorectal cancer (CRC), only few studies investigated the clinical meaning of fecal immunochemical test (FIT) in other diseases. We evaluated the clinical utility of FIT in patients with cardiovascular diseases (namely, ischemic stroke and myocardial infarction [MI]). Methods and Results Using the National Health Insurance database, participants (aged >50 years) with CRC screening records from 2009 to 2012 were screened and followed up. Subjects with a history of cardiovascular diseases and CRC were excluded. Ischemic stroke, MI, and other comorbidities were defined by International Classification of Diseases, Tenth Revision (ICD‐10), codes. Age, sex, smoking, alcohol consumption, regular exercise, diabetes mellitus, hypertension, dyslipidemia, and body mass index were adjusted in a multivariate analysis. A total of 6 277 446 subjects were eligible for analysis. During the mean 6.79 years of follow‐up, 168 570 participants developed ischemic stroke, 105 983 developed MI, and 11 253 deaths were observed. A multivariate‐adjusted model revealed that the risk of ischemic stroke was higher in the FIT‐positive population (adjusted hazard ratio [HR], 1.09; 95% CI, 1.07–1.11). Similarly, FIT‐positive subjects were at an increased risk of MI (adjusted HR, 1.09; 95% CI, 1.06–1.12). Moreover, increased all‐cause mortality was observed in the FIT‐positive population (adjusted HR, 1.15; 95% CI, 1.07–1.23). The increased risk remained consistent in the stratified analysis on anemia and CRC status. Conclusions Positive FIT findings were associated with ischemic stroke, MI, and mortality. Occult blood in feces may offer more clinical information than its well‐known conventional role in CRC screening.
Collapse
Affiliation(s)
- Jung Min Moon
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea
| | - Hyun Jung Lee
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science Soongsil University Seoul Republic of Korea
| | - Da Hye Kim
- Department of Biostatistics College of Medicine The Catholic University of Korea Seoul Republic of Korea
| | - Seung Wook Hong
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea.,Department of Gastroenterology Asan Medical Center University of Ulsan College of Medicine Seoul Republic of Korea
| | - Hosim Soh
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea
| | - Seona Park
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea
| | - Eun Ae Kang
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea.,Department of Internal Medicine and Institute of Gastroenterology Yonsei University College of Medicine Seoul Republic of Korea
| | - Jooyoung Lee
- Department of Internal Medicine Healthcare Research InstituteSeoul National University Hospital Healthcare System Gangnam Center Seoul Republic of Korea
| | - Seong-Joon Koh
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea
| | - Jong Pil Im
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea
| | - Joo Sung Kim
- Department of Internal Medicine Liver Research InstituteSeoul National University College of Medicine Seoul Republic of Korea.,Department of Internal Medicine Healthcare Research InstituteSeoul National University Hospital Healthcare System Gangnam Center Seoul Republic of Korea
| |
Collapse
|
38
|
Yang J, Kim CJ, Go YS, Lee HY, Kim MG, Oh SW, Cho WY, Im SH, Jo SK. Intestinal microbiota control acute kidney injury severity by immune modulation. Kidney Int 2020; 98:932-946. [PMID: 32470493 DOI: 10.1016/j.kint.2020.04.048] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/13/2020] [Accepted: 04/13/2020] [Indexed: 12/15/2022]
Abstract
Intestinal microbiota impacts the host immune system and influences the outcomes of chronic diseases. However, it remains uncertain whether acute kidney injury (AKI) impacts intestinal microbiota or vice versa. To determine this, we investigated the mechanistic link between AKI, microbiota, and immune response in ischemia/reperfusion injury. Microbiota alteration and its biological consequences after ischemia/reperfusion injury were examined and the effect of dysbiotic microbiota on the outcome of AKI was also assessed by colonizing germ-free mice with post-AKI microbiota. The role of Th17, Th1, Tregs cells and macrophage polarization in mediating the renoprotective effect of antibiotic induced microbiota depletion in ischemia/reperfusion injury was also determined. Increase of Enterobacteriacea, decrease of Lactobacilli, and Ruminococacceae were found to be the hallmarks of ischemia/reperfusion injury induced dysbiosis and were associated with a decreased levels of short-chain fatty acids, intestinal inflammation and leaky gut. Colonizing germ-free mice with post-AKI microbiota worsened ischemia/reperfusion injury severity with exaggerated inflammation in recipient mice compared to colonizing with microbiota from sham operated mice. Microbiota depletion by oral antibiotics protected against ischemia/reperfusion injury. This renoprotective effect was associated with reduced Th 17, Th 1 response along with expansion of regulatory T cells, and M2 macrophages. Our study demonstrated a unique bidirectional relationship between the kidney and the intestine during AKI. Intestinal dysbiosis, inflammation and leaky gut are consequences of AKI but they also represent an important modifier determining post-AKI severity. Thus, targeting the intestinal microbiota might provide a novel therapeutic strategy in AKI.
Collapse
Affiliation(s)
- Jihyun Yang
- Division of Nephrology, Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Chan Johng Kim
- Division of Integrative Biosciences and Biotechnology & Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Yoon Sook Go
- Division of Nephrology, Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Hee Young Lee
- Division of Nephrology, Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Myung-Gyu Kim
- Division of Nephrology, Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Se Won Oh
- Division of Nephrology, Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Won Yong Cho
- Division of Nephrology, Department of Internal Medicine, Korea University Medical College, Seoul, Korea
| | - Sin-Hyeog Im
- Division of Integrative Biosciences and Biotechnology & Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
| | - Sang Kyung Jo
- Division of Nephrology, Department of Internal Medicine, Korea University Medical College, Seoul, Korea.
| |
Collapse
|
39
|
Groot HE, van de Vegte YJ, Verweij N, Lipsic E, Karper JC, van der Harst P. Human genetic determinants of the gut microbiome and their associations with health and disease: a phenome-wide association study. Sci Rep 2020; 10:14771. [PMID: 32901066 PMCID: PMC7479141 DOI: 10.1038/s41598-020-70724-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
Small-scale studies have suggested a link between the human gut microbiome and highly prevalent diseases. However, the extent to which the human gut microbiome can be considered a determinant of disease and healthy aging remains unknown. We aimed to determine the spectrum of diseases that are linked to the human gut microbiome through the utilization of its genetic determinants as a proxy for its composition. 180 single nucleotide polymorphisms (SNPs) known to influence the human gut microbiome were used to assess the association with health and disease outcomes in 422,417 UK Biobank participants. Potential causal estimates were obtained using a Mendelian randomization (MR) approach. From the total sample analysed (mean age was 57 ± 8 years), 194,567 (46%) subjects were male. Median exposure was 66-person years (interquartile range 59-72). Eleven SNPs were significantly associated with 28 outcomes (Bonferroni corrected P value < 4.63·10-6) including food intake, hypertension, atopy, COPD, BMI, and lipids. Multiple SNP MR pointed to a possible causal link between Ruminococcus flavefaciens and hypertension, and Clostridium and platelet count. Microbiota and their metabolites might be of importance in the interplay between overlapping pathophysiological processes, although challenges remain in establishing causal relationships.
Collapse
Affiliation(s)
- Hilde E Groot
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Yordi J van de Vegte
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Erik Lipsic
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Jacco C Karper
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands.
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
40
|
Perrino C, Ferdinandy P, Bøtker HE, Brundel BJJM, Collins P, Davidson SM, den Ruijter HM, Engel FB, Gerdts E, Girao H, Gyöngyösi M, Hausenloy DJ, Lecour S, Madonna R, Marber M, Murphy E, Pesce M, Regitz-Zagrosek V, Sluijter JPG, Steffens S, Gollmann-Tepeköylü C, Van Laake LW, Van Linthout S, Schulz R, Ytrehus K. Improving translational research in sex-specific effects of comorbidities and risk factors in ischaemic heart disease and cardioprotection: position paper and recommendations of the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2020; 117:367-385. [PMID: 32484892 DOI: 10.1093/cvr/cvaa155] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/29/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Ischaemic heart disease (IHD) is a complex disorder and a leading cause of death and morbidity in both men and women. Sex, however, affects several aspects of IHD, including pathophysiology, incidence, clinical presentation, diagnosis as well as treatment and outcome. Several diseases or risk factors frequently associated with IHD can modify cellular signalling cascades, thus affecting ischaemia/reperfusion injury as well as responses to cardioprotective interventions. Importantly, the prevalence and impact of risk factors and several comorbidities differ between males and females, and their effects on IHD development and prognosis might differ according to sex. The cellular and molecular mechanisms underlying these differences are still poorly understood, and their identification might have important translational implications in the prediction or prevention of risk of IHD in men and women. Despite this, most experimental studies on IHD are still undertaken in animal models in the absence of risk factors and comorbidities, and assessment of potential sex-specific differences are largely missing. This ESC WG Position Paper will discuss: (i) the importance of sex as a biological variable in cardiovascular research, (ii) major biological mechanisms underlying sex-related differences relevant to IHD risk factors and comorbidities, (iii) prospects and pitfalls of preclinical models to investigate these associations, and finally (iv) will provide recommendations to guide future research. Although gender differences also affect IHD risk in the clinical setting, they will not be discussed in detail here.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary.,Pharmahungary Group, Hajnoczy str. 6., H-6722 Szeged, Hungary
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 161, 8200 Aarhus, Denmark
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, 1108 HV, the Netherlands
| | - Peter Collins
- Imperial College, Faculty of Medicine, National Heart & Lung Institute, South Kensington Campus, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Hester M den Ruijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), Schwabachanlage 12, 91054 Erlangen, Germany
| | - Eva Gerdts
- Department for Clinical Science, University of Bergen, PO Box 7804, 5020 Bergen, Norway
| | - Henrique Girao
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, and Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, 169609, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, 119228, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, Chris Barnard Building, University of Cape Town, Private Bag X3 7935 Observatory, Cape Town, South Africa
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, 6410 Fannin St #1014, Houston, TX 77030, USA
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Elizabeth Murphy
- Laboratory of Cardiac Physiology, Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD 20892, USA
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS Via Parea, 4, I-20138 Milan, Italy
| | - Vera Regitz-Zagrosek
- Berlin Institute of Gender in Medicine, Center for Cardiovascular Research, DZHK, partner site Berlin, Geschäftsstelle Potsdamer Str. 58, 10785 Berlin, Germany.,University of Zürich, Rämistrasse 71, 8006 Zürich, Germany
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands.,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstr.35, A - 6020 Innsbruck, Austria
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Ludwigstraße 23, 35390 Giessen, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9037 Tromsø, Norway
| |
Collapse
|
41
|
Zhu L, Hu B, Guo Y, Yang H, Zheng J, Yao X, Hu H, Liu H. Effect of Chitosan oligosaccharides on ischemic symptom and gut microbiota disbalance in mice with hindlimb ischemia. Carbohydr Polym 2020; 240:116271. [PMID: 32475560 DOI: 10.1016/j.carbpol.2020.116271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/16/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
This study was designed to explore the effect of Chitosan oligosaccharides (COS) on mouse hindlimb ischemia by femoral artery ligation. Here, we demonstrated that COS treatment statistically promoted the blood perfusion and neovascularization in ischemic hindlimb of mice, accompanied by the suppression of inflammation and oxidative stress. By 16S rDNA gene sequencing, the disbalanced gut microbiota was observed in ischemic mice, while COS treatment, at least in part, restored the abundance changes of some intestinal bacteria at either phylum or genus levels. Based on metabolomics analysis on mouse plasma by UPLC-QTOF-MS, we screened 20 metabolites with the largest responses to ischemia, several of which were markedly reversed by COS. By Spearman's correlation analysis, the changed metabolites might act as a bridge between improved intestinal bacterial structure and alleviated hindlimb ischemia of mice treated by COS. Our studies point towards a potential role of COS in treatment of peripheral ischemia diseases.
Collapse
Affiliation(s)
- Lin Zhu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Xiaowei Yao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China.
| |
Collapse
|
42
|
Daskalopoulos EP, Hermans KCM, Debets J, Strzelecka A, Leenders P, Vervoort-Peters L, Janssen BJA, Blankesteijn WM. The Beneficial Effects of UM206 on Wound Healing After Myocardial Infarction in Mice Are Lost in Follow-Up Experiments. Front Cardiovasc Med 2019; 6:118. [PMID: 31620445 PMCID: PMC6759626 DOI: 10.3389/fcvm.2019.00118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/01/2019] [Indexed: 12/30/2022] Open
Abstract
Introduction: An inadequate wound healing following myocardial infarction (MI) is one of the main etiologies of heart failure (HF) development. Interventions aiming at improving this process may contribute to preserving cardiac function after MI. Our group, as well as others, have demonstrated the crucial role of Wnt/frizzled signaling in post-MI remodeling. In this overview, we provide the results of different studies aimed at confirming an initial study from our group, in which we observed beneficial effects of administration of a peptide fragment of Wnt5a, UM206, on infarct healing in a mouse MI model. Methods: Mice were subjected to permanent left coronary artery ligation, and treated with saline (control) or UM206, administered via osmotic minipumps. Cardiac function was assessed by echocardiography and hemodynamic measurements, while infarct size and myofibroblast content were characterized by (immuno)histochemistry. Results: In total, we performed seven follow-up studies, but we were unable to reproduce the beneficial effects of UM206 on infarct healing in most of them. Variations in dose and timing of UM206 administration, its manufacturer and the genetic background of the mice could not restore the phenotype. An in-depth analysis of the datasets revealed that the absence of effect of UM206 coincided with a lack of adverse cardiac remodeling and HF development in all experimental groups, irrespective of the treatment. Discussion: Irreproducibility of experimental observations is a major issue in biomedical sciences. It can arise from a relatively low number of experimental observations in the original study, a faulty hypothesis or a variation in the experimental model that cannot be controlled. In this case, the lack of adverse cardiac remodeling and lung weight increases in the follow-up studies point out to altered experimental conditions as the most likely explanation.
Collapse
Affiliation(s)
- Evangelos P Daskalopoulos
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| | - Jacques Debets
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| | - Agnieszka Strzelecka
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| | - Peter Leenders
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| | - Lily Vervoort-Peters
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| | - Ben J A Janssen
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University (UM), Maastricht, Netherlands
| |
Collapse
|
43
|
Querio G, Antoniotti S, Levi R, Gallo MP. Trimethylamine N-Oxide Does Not Impact Viability, ROS Production, and Mitochondrial Membrane Potential of Adult Rat Cardiomyocytes. Int J Mol Sci 2019; 20:ijms20123045. [PMID: 31234461 PMCID: PMC6627059 DOI: 10.3390/ijms20123045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/06/2019] [Accepted: 06/19/2019] [Indexed: 01/10/2023] Open
Abstract
Trimethylamine N-oxide (TMAO) is an organic compound derived from dietary choline and L-carnitine. It behaves as an osmolyte, a protein stabilizer, and an electron acceptor, showing different biological functions in different animals. Recent works point out that, in humans, high circulating levels of TMAO are related to the progression of atherosclerosis and other cardiovascular diseases. However, studies on a direct role of TMAO in cardiomyocyte parameters are still limited. The purpose of this work is to study the effects of TMAO on isolated adult rat cardiomyocytes. TMAO in both 100 µM and 10 mM concentrations, from 1 to 24 h of treatment, does not affect cell viability, sarcomere length, intracellular ROS, and mitochondrial membrane potential. Furthermore, the simultaneous treatment with TMAO and known cardiac insults, such as H2O2 or doxorubicin, does not affect the treatment’s effect. In conclusion, TMAO cannot be considered a direct cause or an exacerbating risk factor of cardiac damage at the cellular level in acute conditions.
Collapse
Affiliation(s)
- Giulia Querio
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Turin, Italy.
| | - Susanna Antoniotti
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Turin, Italy.
| | - Renzo Levi
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Turin, Italy.
| | - Maria Pia Gallo
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123 Turin, Italy.
| |
Collapse
|
44
|
|
45
|
The role of microbiota in the pathogenesis of lupus: Dose it impact lupus nephritis? Pharmacol Res 2019; 139:191-198. [DOI: 10.1016/j.phrs.2018.11.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 02/06/2023]
|
46
|
Zununi Vahed S, Moghaddas Sani H, Rahbar Saadat Y, Barzegari A, Omidi Y. Type 1 diabetes: Through the lens of human genome and metagenome interplay. Biomed Pharmacother 2018; 104:332-342. [PMID: 29775902 DOI: 10.1016/j.biopha.2018.05.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/09/2018] [Accepted: 05/09/2018] [Indexed: 02/07/2023] Open
Abstract
Diabetes is a genetic- and epigenetic-related disease from which a large population worldwide suffers. Some genetic factors along with various mutations related to the immune system for disease mechanism(s) have contrastively been determined. However, sometimes mechanisms have not been fully managed for the clarification of the initiation and/or progression of diseases to help patients. In the recent years, due to familiarity with the role of gut microbiota in the health, it has been found that the changes of the microbial balance in the industrialized societies can cause a battery of modern diseases, for which we have no specific definition of how they emerge. This work aims to explore the relationship between the human gut microbiota and the immune system along with their possible role in avoiding/emerging of type 1 diabetes (T1D) accompanied with the relation between genome and metagenome and their imbalance in causing T1D. Moreover, it provides novel view on how to balance the intestinal microbiota by lifestyle to hinder the mechanisms leading to T1D.
Collapse
Affiliation(s)
| | | | - Yalda Rahbar Saadat
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|