1
|
Xue J, Allaband C, Zuffa S, Poulsen O, Meadows J, Zhou D, Dorrestein PC, Knight R, Haddad GG. Gut microbiota and derived metabolites mediate obstructive sleep apnea induced atherosclerosis. Gut Microbes 2025; 17:2474142. [PMID: 40025767 PMCID: PMC11881840 DOI: 10.1080/19490976.2025.2474142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia/hypercapnia (IHC), affects predominantly obese individuals, and increases atherosclerosis risk. Since we and others have implicated gut microbiota and metabolites in atherogenesis, we dissected their contributions to OSA-induced atherosclerosis. Atherosclerotic lesions were compared between conventionally-reared specific pathogen free (SPF) and germ-free (GF) Apoe-/- mice following a high fat high cholesterol diet (HFHC), with and without IHC conditions. The fecal microbiota and metabolome were profiled using 16S rRNA gene amplicon sequencing and untargeted tandem mass spectrometry (LC-MS/MS) respectively. Phenotypic data showed that HFHC significantly increased atherosclerosis as compared to regular chow (RC) in both aorta and pulmonary artery (PA) of SPF mice. IHC exacerbated lesions in addition to HFHC. Differential abundance analysis of gut microbiota identified an enrichment of Akkermansiaceae and a depletion of Muribaculaceae (formerly S24-7) family members in the HFHC-IHC group. LC-MS/MS showed a dysregulation of bile acid profiles with taurocholic acid, taurodeoxycholic acid, and 12-ketodeoxycholic acid enriched in the HFHC-IHC group, long-chain N-acyl amides, and phosphatidylcholines. Interestingly, GF Apoe-/- mice markedly reduced atherosclerotic formation relative to SPF Apoe-/- mice in the aorta under HFHC/IHC conditions. In contrast, microbial colonization did not show a significant impact on the atherosclerotic progression in PA. In summary, this research demonstrated that (1) IHC acts cooperatively with HFHC to induce atherosclerosis; (2) gut microbiota modulate atherogenesis, induced by HFHC/IHC, in the aorta not in PA; (3) different analytical methods suggest that a specific imbalance between Akkermansiaceae and Muribaculaceae bacterial families mediate OSA-induced atherosclerosis; and (4) derived bile acids, such as deoxycholic acid and lithocholic acid, regulate atherosclerosis in OSA. The knowledge obtained provides novel insights into the potential therapeutic approaches to prevent and treat OSA-induced atherosclerosis.
Collapse
MESH Headings
- Animals
- Gastrointestinal Microbiome/physiology
- Atherosclerosis/etiology
- Atherosclerosis/microbiology
- Atherosclerosis/metabolism
- Sleep Apnea, Obstructive/complications
- Sleep Apnea, Obstructive/microbiology
- Sleep Apnea, Obstructive/metabolism
- Mice
- Male
- Bacteria/classification
- Bacteria/genetics
- Bacteria/metabolism
- Bacteria/isolation & purification
- Diet, High-Fat/adverse effects
- Feces/microbiology
- Mice, Inbred C57BL
- RNA, Ribosomal, 16S/genetics
- Bile Acids and Salts/metabolism
- Metabolome
- Specific Pathogen-Free Organisms
- Disease Models, Animal
- Tandem Mass Spectrometry
- Mice, Knockout, ApoE
- Apolipoproteins E/genetics
Collapse
Affiliation(s)
- Jin Xue
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Celeste Allaband
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, San Diego, CA, USA
| | - Orit Poulsen
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Jason Meadows
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Dan Zhou
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Pieter C. Dorrestein
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Gabriel G. Haddad
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
- The Division of Respiratory Medicine, Rady Children’s Hospital, San Diego, CA, USA
| |
Collapse
|
2
|
Wu H, Chen J, Guo S, Deng J, Zhou Z, Zhang X, Qi T, Yu F, Yang Q. Advances in the acting mechanism and treatment of gut microbiota in metabolic dysfunction-associated steatotic liver disease. Gut Microbes 2025; 17:2500099. [PMID: 40394806 PMCID: PMC12101596 DOI: 10.1080/19490976.2025.2500099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease(MASLD) is increasing in prevalence worldwide and has become the greatest potential risk for cirrhosis and hepatocellular liver cancer. Currently, the role of gut microbiota in the development of MASLD has become a research hotspot. The development of MASLD can affect the homeostasis of gut microbiota, and significant changes in the composition or abundance of gut microbiota and its metabolite abnormalities can influence disease progression. The regulation of gut microbiota is an important strategy and novel target for the treatment of MASLD with good prospects. In this paper, we summarize the role of gut microbiota and its metabolites in the pathogenesis of MASLD, and describe the potential preventive and therapeutic efficacy of gut microbiota as a noninvasive marker to regulate the pathogenesis of MASLD based on the "gut-hepatic axis", which will provide new therapeutic ideas for the clinic.
Collapse
Affiliation(s)
- Huaying Wu
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Jingjing Chen
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Shuyuan Guo
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jinhao Deng
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Zimeng Zhou
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Xuan Zhang
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - TianTian Qi
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- Department of Spine Surgery, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qi Yang
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
3
|
Zhou P, Xu HJ, Wang L. Cardiovascular protective effects of natural flavonoids on intestinal barrier injury. Mol Cell Biochem 2025; 480:3343-3362. [PMID: 39820766 DOI: 10.1007/s11010-025-05213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
Natural flavonoids may be utilized as an important therapy for cardiovascular diseases (CVDs) caused by intestinal barrier damage. More research is being conducted on the protective properties of natural flavonoids against intestinal barrier injury, although the underlying processes remain unknown. Thus, the purpose of this article is to present current research on natural flavonoids to reduce the incidence of CVDs by protecting intestinal barrier injury, with a particular emphasis on intestinal epithelial barrier integrity (inhibiting oxidative stress, regulating inflammatory cytokine expression, and increasing tight junction protein expression). Furthermore, the mechanisms driving intestinal barrier injury development are briefly explored, as well as natural flavonoids having CVD-protective actions on the intestinal barrier. In addition, natural flavonoids with myocardial protective effects were docked with ZO-1 targets to find natural products with higher activity. These natural flavonoids can improve intestinal mechanical barrier function through anti-oxidant or anti-inflammatory mechanism, and then prevent the occurrence and development of CVDs.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Hui-Juan Xu
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Liang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
- Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China.
| |
Collapse
|
4
|
Cui Y, Wu Y, Shi P, Ni Y, Zeng H, Zhang Z, Zhao C, Sun W, Yi Q. Mitigating microplastic-induced organ Damage: Mechanistic insights from the microplastic-macrophage axes. Redox Biol 2025; 84:103688. [PMID: 40412021 DOI: 10.1016/j.redox.2025.103688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 05/10/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025] Open
Abstract
We live in a world increasingly dominated by plastic, leading to the generation of microplastic particles that pose significant global health concerns. Microplastics can enter the body via ingestion, inhalation, and direct contact, accumulating in various tissues and potentially causing harm. Despite this, the specific cellular mechanisms and signaling pathways involved remain poorly understood. Macrophages are essential in absorbing, distributing, and eliminating microplastics, playing a key role in the body's defense mechanisms. Recent evidence highlights oxidative stress signaling as a key pathway in microplastic-induced macrophage dysfunction. The accumulation of microplastics generates reactive oxygen species (ROS), disrupting normal macrophage functions and exacerbating inflammation and organ damage. This review serves as the first comprehensive examination of the interplay between microplastics, macrophages, and oxidative stress. It discusses how oxidative stress mediates macrophage responses to microplastics and explores the interactions with gut microbiota. Additionally, it reviews the organ damage resulting from alterations in macrophage function mediated by microplastics and offers a novel perspective on the defense, assessment, and treatment of microplastic-induced harm from the viewpoint of macrophages.
Collapse
Affiliation(s)
- Yinxing Cui
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China; Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Yuqi Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Pan Shi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Yan Ni
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Huaying Zeng
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Zhao Zhang
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Chunling Zhao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China.
| | - Weichao Sun
- Department of Orthopedics, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, 518035, China.
| | - Qian Yi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China.
| |
Collapse
|
5
|
Obare LM, Stephens VR, Wanjalla CN. Understanding residual risk of cardiovascular disease in people with HIV. Curr Opin HIV AIDS 2025:01222929-990000000-00164. [PMID: 40397567 DOI: 10.1097/coh.0000000000000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
PURPOSE OF REVIEW Traditional cardiovascular risk factors, combined with persistent systemic inflammation, contribute to the increased prevalence of atherosclerotic cardiovascular disease (ASCVD) in people with HIV (PWH). This review highlights key findings from the REPRIEVE trial on statin-based primary prevention of major adverse cardiovascular events in PWH. It explores HIV-specific immune mechanisms contributing to residual cardiovascular risk. RECENT FINDINGS In REPRIEVE, statin therapy used for primary prevention of major adverse cardiovascular events in PWH decreased the plasma lipoprotein-associated phospholipase A2, oxidized low-density lipoprotein, and high-sensitivity C-reactive protein (hs-CRP). However, several inflammatory markers including soluble CD14 (sCD14), sCD163, interleukin (IL)-1β, interleukin (IL)-6, IL-10, and caspase 1 did not change. The HIV reservoir, dysfunctional CD4+ T cells, immunoglobulin G N-glycans, antiapolipoprotein A1 autoantibodies, trained immunity, and clonal hematopoiesis of indeterminate potential may contribute to residual inflammation. SUMMARY Despite antiretroviral and statin therapy, residual ASCVD risk in PWH underscores the need for targeted interventions. Anti-inflammatory therapies, including IL-6 and IL-1β inhibitors, CCR5 antagonists (e.g., maraviroc, cenicriviroc mesylate), and immunomodulatory agents like methotrexate and colchicine, are being explored. Understanding HIV-driven immune dysregulation may lead to novel strategies to mitigate cardiovascular risk in this population.
Collapse
Affiliation(s)
- Laventa M Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center
| | | | - Celestine N Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center
- The Center for AIDS Health Disparities Research
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
6
|
Sule RO, Rivera GDT, Vaidya T, Gartrell E, Gomes AV. Environmental Toxins and Oxidative Stress: The Link to Cardiovascular Diseases. Antioxidants (Basel) 2025; 14:604. [PMID: 40427486 DOI: 10.3390/antiox14050604] [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: 04/01/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a leading global health concern, responsible for substantial morbidity and mortality. In recent years, as our understanding of the multifaceted nature of CVDs has increased, it has become increasingly evident that traditional risk factors alone do not account for the entirety of cardiovascular morbidity and mortality. Environmental toxins, a heterogeneous group of substances ubiquitous in our surroundings, have now entered the spotlight as offenders in the development and progression of CVDs. Environmental toxins include heavy metals, air pollutants, pesticides, and endocrine-disrupting chemicals, among others. Upon exposure, they can elicit oxidative stress, a condition characterized by an imbalance between the production of reactive oxygen species (ROS) and the body's ability to detoxify and repair the resulting damage. Oxidative stress triggers a cascade of events, including inflammation, endothelial dysfunction, lipid peroxidation, and vascular remodeling, which can contribute to the development of atherosclerosis, hypertension, and other cardiovascular pathologies. This article delves into the molecular mechanisms underpinning oxidative stress-mediated cardiovascular damage induced by environmental toxins, emphasizing the role of specific toxins in this process. Further research is necessary to understand how individual susceptibility and genotype influence the impact of environmental toxins on oxidative stress and the risk of CVD.
Collapse
Affiliation(s)
- Rasheed O Sule
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Gabriela Del Toro Rivera
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Tanishq Vaidya
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Emily Gartrell
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA
| |
Collapse
|
7
|
Kang H, Huang D, Zhang W, Wang J, Liu Z, Wang Z, Jiang G, Gao A. Propionic acid/FBP1 is involved in polystyrene nanoplastic-induced cardiac injury via the gut-heart axis. Part Fibre Toxicol 2025; 22:10. [PMID: 40346689 PMCID: PMC12063461 DOI: 10.1186/s12989-025-00626-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Micro-/nanoplastics (MNPLs) are widely found in the environment and have toxic effects on various organs and systems. However, the role of the gut-cardiac axis in cardiotoxicity induced by MNPLs has not yet been elucidated through research. RESULTS In this study, we examined the effects of 80 nm polystyrene nanoplastics (PS-NPs) on the heart and human cardiomyocytes (AC16) cells. Histopathological examination showed that NPs caused impaired cardiac function and increased myocardial collagen deposition. In view of the potential influence of gut microbiota and its metabolites on cardiac function, we conduct this study to investigate the specific effects they have on cardiac function. Analysis of cecal contents by 16 s ribosomal RNA (rRNA) and short chain fatty acids (SCFAs) revealed that colonic tissue damage, intestinal flora disorder, and reduction of propionic acid induced by PS-MPs were closely related to cardiac function. Further transcriptomic analysis of heart and colon tissues indicated that propionic acid may reduce cardiac function by reducing the expression of fructose-1, 6-biphosphatase 1 (FBP1). The hypothesis was further verified by in vitro intervention experiments with sodium propionate and FBP1 activator (BML-275). CONCLUSIONS In summary, our study systematically demonstrated the role of gut-heart axis in NPs-induced cardiac injury, and the specific process was that NPs exposure reduced propionate level, which in turn inhibited FBP1 expression to impair cardiac function. These findings provide new insights into NPs-induced cardiotoxicity and identifie potential therapeutic targets, providing clues for the prevention and treatment of NPs-induced cardiac injury in the future.
Collapse
Affiliation(s)
- Huiwen Kang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Danyang Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Wei Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - JingYu Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Ziyan Liu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Ziyan Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Guangyu Jiang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Ai Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, 10 Xitoutiao, You An Men, Beijing, 100069, China.
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
8
|
Yu S, Huang F, Huang Y, Yan F, Li Y, Xu S, Zhao Y, Zhang X, Chen R, Chen X, Zhang P. Deciphering the influence of gut and oral microbiomes on menopause for healthy aging. J Genet Genomics 2025; 52:601-614. [PMID: 39577767 DOI: 10.1016/j.jgg.2024.11.010] [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: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Menopause is characterized by the cessation of menstruation and a decline in reproductive function, which is an intrinsic component of the aging process. However, it has been a frequently overlooked field of women's health. The oral and gut microbiota, constituting the largest ecosystem within the human body, are important for maintaining human health and notably contribute to the healthy aging of menopausal women. Therefore, a comprehensive review elucidating the impact of the gut and oral microbiota on menopause for healthy aging is of paramount importance. This paper presents the current understanding of the microbiome during menopause, with a particular focus on alterations in the oral and gut microbiota. Our study elucidates the complex interplay between the microbiome and sex hormone levels, explores microbial crosstalk dynamics, and investigates the associations between the microbiome and diseases linked to menopause. Additionally, this review explores the potential of microbiome-targeting therapies for managing menopause-related diseases. Given that menopause can last for approximately 30 years, gaining insights into how the microbiome and menopause interact could pave the way for innovative interventions, which may result in symptomatic relief from menopause and an increase in quality of life in women.
Collapse
Affiliation(s)
- Shuting Yu
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Feiling Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing 100730, China
| | - Yixuan Huang
- Beijing ClouDNA Technology Co., Ltd., Beijing 101407, China
| | - Fangxu Yan
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yi Li
- Hunan Agriculture University, Changsha, Hunan 410128, China
| | - Shenglong Xu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yan Zhao
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xinlei Zhang
- Beijing ClouDNA Technology Co., Ltd., Beijing 101407, China
| | - Rong Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing 100730, China.
| | - Xingming Chen
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
9
|
Arenas-Montes J, Alcala-Diaz JF, Garcia-Fernandez H, Gutierrez-Mariscal FM, Lopez-Moreno A, Luque-Cordoba D, Arenas-de Larriva AP, Torres-Peña JD, Luque RM, Prodam F, Priego-Capote F, Delgado-Lista J, Lopez-Miranda J, Camargo A. A microbiota pattern associated with cardiovascular events in secondary prevention: the CORDIOPREV study. Eur Heart J 2025:ehaf181. [PMID: 40197788 DOI: 10.1093/eurheartj/ehaf181] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/21/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND AND AIMS Preventing new cardiovascular events in patients with established cardiovascular disease (CVD) is a daunting task for clinicians. Intestinal microbiota may help identify patients at risk, thus improving the strategies of secondary prevention. The aim of this study was to evaluate the baseline differences between the gut microbiota from coronary heart disease (CHD) patients suffering new major adverse cardiovascular events (MACEs) in the following 7 years, compared with CHD patients who did not undergo new MACE in this period, and to build a score associated with the risk of suffering new MACE. METHODS Within the framework of the CORDIOPREV study, a clinical trial that involved 1002 patients with CHD, intestinal microbiota was examined in patients with available faecal samples (n = 679, 132 MACE), through 16S metagenomics on the Illumina MiSeq and Quiime2 software. Lipopolysaccharide (LPS) was measured using limulus amoebocyte lysate test. RESULTS Random survival forest identified 10 bacterial taxa with a higher predictive power for MACE incidence. Receiver operating characteristic curves yielded an area under the curve of 65.2% (59.1%-71.3%) in the training set and 68.6% (59.3%-77.9%) in the validation set. The intestinal microbiota risk score was associated with a MACE incidence hazard ratio of 2.01 (95% confidence interval 1.37-3.22). Lipopolysaccharide analysis showed a greater LPS post-prandial fold change in the MACE group (P = .005). CONCLUSIONS These results reinforce the relationship between intestinal microbiota and CVD and suggest that a microbiota profile is associated with MACE in CHD patients, in addition to higher endotoxaemia.
Collapse
Affiliation(s)
- Javier Arenas-Montes
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Helena Garcia-Fernandez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Alejandro Lopez-Moreno
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Diego Luque-Cordoba
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- Department of Analytical Chemistry, Annex Marie Curie Building, Campus of Rabanales, University of Cordoba, Cordoba 14071, Spain
- Consortium for Biomedical Research in Frailty & Healthy Ageing, CIBERFES, Carlos III Institute of Health, Madrid 28029, Spain
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Raul M Luque
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba 14071, Spain
| | - Flavia Prodam
- Department of Health Sciences, Unit of Endocrinology, Università del Piemonte Orientale, Novara 28100, Italy
| | - Feliciano Priego-Capote
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- Department of Analytical Chemistry, Annex Marie Curie Building, Campus of Rabanales, University of Cordoba, Cordoba 14071, Spain
- Consortium for Biomedical Research in Frailty & Healthy Ageing, CIBERFES, Carlos III Institute of Health, Madrid 28029, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
10
|
Liu X, Ju W, Qiang E, Li D, Liang Q, Guo M, Yun W, Chen Z. Hesperidin improves cardiac fibrosis induced by β-adrenergic activation through modulation of gut microbiota. J Pharmacol Exp Ther 2025; 392:103578. [PMID: 40378637 DOI: 10.1016/j.jpet.2025.103578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/18/2025] [Accepted: 03/29/2025] [Indexed: 05/19/2025] Open
Abstract
Cardiac fibrosis is a prevalent characteristic of various cardiovascular diseases and poses a significant global health challenge. Recent research has established a robust correlation between gut microbiota and cardiovascular diseases. Hesperidin has been shown to possess cardioprotective properties to some extent. Furthermore, studies suggest that hesperidin may enhance overall health by regulating intestinal flora. However, there is a lack of reports regarding the effects of hesperidin on cardiac fibrosis. This study aimed to investigate the mechanisms by which hesperidin ameliorates cardiac fibrosis through the regulation of gut microbiota and associated metabolites. Cardiac fibrosis was induced in C57BL/6 mice via subcutaneous injection of isoproterenol (5 mg/kg per day) for a duration of 7 days. Echocardiography was used to assess cardiac function, while Masson staining, western blot analysis, and real-time polymerase chain reaction were used to evaluate fibrosis-related indicators. Changes in gut microbiota were analyzed through 16S ribosomal RNA gene sequencing. Our findings indicate that hesperidin significantly mitigates cardiac fibrosis in mice. These beneficial effects are associated with improvements in the dysbiosis of intestinal microbiota observed in fibrotic mouse models. The involvement of gut microbiota in cardiac fibrosis was further corroborated by administering hesperidin therapy to mice depleted of gut microbiota. To our knowledge, this study provides the first evidence that the modulation of gut microbiota by hesperidin contributes to improved outcomes in cardiac fibrosis. The use of traditional Chinese medicine to modulate gut microbiota presents a promising strategy for the treatment of cardiac fibrosis. SIGNIFICANCE STATEMENT: The work is extremely interesting because it acts on a frontier of science that relates the influence of the intestinal microbiota with human physiological systems and associated pathologies. This study provides the first evidence that the modulation of gut microbiota by hesperidin contributes to improved outcomes in cardiac fibrosis.
Collapse
Affiliation(s)
- Xia Liu
- The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Weiwei Ju
- The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Erjiao Qiang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dongning Li
- Dalian Municipal Women and Children's Medical Center (Group), Dalian, China
| | - Qing Liang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Meina Guo
- Department of Geriatrics and Special Services Medicine, Xinqiao Hospital, Army Military Medical University, Chongqing, China
| | - Weijing Yun
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China.
| | - Zhenzhen Chen
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
11
|
Khavani M, Mehranfar A, Mofrad MRK. Unravelling the Glycan Code: Molecular Dynamics and Quantum Chemistry Reveal How O-Glycan Functional Groups Govern OgpA Selectivity in Mucin Degradation by Akkermansia muciniphila. Microb Biotechnol 2025; 18:e70091. [PMID: 40181232 PMCID: PMC11968330 DOI: 10.1111/1751-7915.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 04/05/2025] Open
Abstract
Mucins, heavily O-glycosylated glycoproteins, are a key component of mucus, and certain gut microbiota, including Akkermansia muciniphila, can utilise mucin glycans as a carbon source. Akkermansia muciniphila produces the O-glycopeptidase enzyme OgpA, which cleaves peptide bonds at the N-terminus of serine (Ser) or threonine (Thr) residues carrying O-glycan substitutions, with selectivity influenced by the O-glycan functional groups. Using molecular dynamics (MD) simulations and quantum chemistry calculations, we explored how different O-glycan groups affect OgpA's selectivity. Our results show that peptides bind to the enzyme via hydrogen bonds, π-π interactions, van der Waals forces and electrostatic interactions, with key residues, including Tyr90, Val138, Gly176, Tyr210 and Glu91, playing important roles. The primary determinant of selectivity is the interaction between the peptide's functional group and the enzyme's binding cavity, while peptide-enzyme interface interactions are secondary. Quantum chemistry calculations reveal that OgpA prefers peptides with a lower electrophilic character. This study provides new insights into mucin degradation by gut microbiota enzymes, advancing our understanding of this critical biological process.
Collapse
Affiliation(s)
- Mohammad Khavani
- Molecular Cell Biomechanics Laboratory, Department of Bioengineering and Mechanical EngineeringUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Aliyeh Mehranfar
- Molecular Cell Biomechanics Laboratory, Department of Bioengineering and Mechanical EngineeringUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Mohammad R. K. Mofrad
- Molecular Cell Biomechanics Laboratory, Department of Bioengineering and Mechanical EngineeringUniversity of California BerkeleyBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrative Bioimaging DivisionLawrence Berkeley National LabBerkeley, CAUSA
| |
Collapse
|
12
|
Tan X, Wu J, Zhang H, Li Y, Huang Y, Zheng P, Xie P. Biogeography of intestinal mucus-associated microbiome: Depletion of genus Pseudomonas is associated with depressive-like behaviors in female cynomolgus macaques. J Adv Res 2025; 70:393-404. [PMID: 38735389 PMCID: PMC11976423 DOI: 10.1016/j.jare.2024.05.013] [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: 10/25/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION Depression is a debilitating and poorly understood mental disorder. There is an urgency to explore new potential biological mechanisms of depression and the gut microbiota is a promising research area. OBJECTIVES Our study was aim to understand regional heterogeneity and potential molecular mechanisms underlying depression induced by dysbiosis of mucus-associated microbiota. METHODS Here, we only selected female macaques because they are more likely to form a natural social hierarchy in a harem-like environment. Because high-ranking macaques rarely displayed depressive-like behaviors, we selected seven monkeys from high-ranking individuals as control group (HC) and the same number of low-ranking ones as depressive-like group (DL), which displayed significant depressive-like behaviors. Then, we collected mucus from the duodenum, jejunum, ileum, cecum and colon of DL and HC monkeys for shotgun metagenomic sequencing, to profile the biogeography of mucus-associated microbiota along duodenum to colon. RESULTS Compared with HC, DL macaques displayed noticeable depressive-like behaviors such as longer duration of huddle and sit alone behaviors (negative emotion behaviors), and fewer duration of locomotion, amicable and ingestion activities (positive emotion behaviors). Moreover, the alpha diversity index (Chao) could predict aforementioned depressive-like behaviors along duodenum to colon. Further, we identified that genus Pseudomonas was consistently decreased in DL group throughout the entire intestinal tract except for the jejunum. Specifically, there were 10, 18 and 28 decreased Pseudomonas spp. identified in ileum, cecum and colon, respectively. Moreover, a bacterial module mainly composed of Pseudomonas spp. was positively associated with three positive emotion behaviors. Functionally, Pseudomonaswas mainly involved in microbiota derived lipid metabolisms such as PPAR signaling pathway, cholesterol metabolism, and fat digestion and absorption. CONCLUSION Different regions of intestinal mucus-associated microbiota revealed that depletion of genus Pseudomonas is associated with depressive-like behaviors in female macaques, which might induce depressive phenotypes through regulating lipid metabolism.
Collapse
Affiliation(s)
- Xunmin Tan
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Jing Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Hanping Zhang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Yifan Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Yu Huang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China.
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China.
| |
Collapse
|
13
|
Liu Y, Wang R, Zhou J, Lyu Q, Zhao X, Yang X, Chen K, Gao Z, Li X. Myricetin alleviates high-fat diet-induced atherosclerosis in ApoE -/- mice by regulating bile acid metabolism involved in gut microbiota remodeling. Food Funct 2025; 16:2737-2749. [PMID: 40059779 DOI: 10.1039/d5fo00374a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Atherosclerosis poses a significant threat to global health. This study aimed to investigate the effects of myricetin (MYR) on high-fat diet (HFD)-induced atherosclerosis in ApoE-/- mice. Our findings demonstrated that MYR treatment significantly reduced the formation of atherosclerotic plaques, particularly at a high dose of 100 mg kg-1 day-1. Additionally, MYR markedly attenuated lipid metabolism disorders in ApoE-/- mice by decreasing body weight, improving serum lipid profiles, and reducing lipid deposition. Analysis of 16S rRNA sequencing revealed that MYR treatment enhanced the abundance of probiotic g_Lachnospiraceae_NK4A136, while it reduced that of obesity-associated genera, including Rikenellaceae_RC9_gut_group and Alistipes. Metabolomic analysis and RT-qPCR tests indicated that MYR upregulated hepatic bile acid biosynthesis, evidenced by increased total bile acid levels and enhanced expression of key enzymes CYP7A1 and CYP8B1, particularly through the classical biosynthetic pathway. Spearman's correlation analysis revealed strong associations between the regulated bile acids and these aforementioned bacteria. Therefore, our results demonstrated that MYR exerts an anti-atherosclerotic effect by modulating the gut-liver axis.
Collapse
Affiliation(s)
- Yilong Liu
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Ruoqi Wang
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Jinren Zhou
- Department of Vascular Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Qiang Lyu
- School of Pharmacy, Zhejiang Chinese Medical University, 548, Binwen Road, Hangzhou 310053, China
| | - Xiaoyong Zhao
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| | - Kunsong Chen
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
| | - Zhiwei Gao
- Department of Vascular Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Xian Li
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, Zhejiang University, Hangzhou 310058, China.
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| |
Collapse
|
14
|
Wei C, Yu X, Chen Y, Jiang J, Cao M, Chen X. Fecal occult blood is associated with an increased risk of cerebral small vessel disease in elderly patients. Med Clin (Barc) 2025; 164:287-291. [PMID: 39613713 DOI: 10.1016/j.medcli.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND AND PURPOSE Studies have shown that fecal occult blood is associated with an increased risk of ischemic stroke, but the relationship between fecal occult blood and cerebral small vessel disease (CSVD) remains largely unknown. This study aimed to identify predictors for CSVD, with fecal occult blood and its proxies as potential influencing factors in elderly patients. METHOD Patients aged 65 years or older with various chronic diseases were enrolled. The presence of CSVD was evaluated by brain MRI results. Fecal occult blood was measured by fecal immunochemical test. Logistic regression analysis was used for the association between the presence of fecal occult blood and the risk of CSVD. RESULTS Logistic regression analysis indicated that a prevalence of positive fecal occult blood was related to CSVD (Model 1, adjusted OR=1.63, 95% CI: 1.15-2.29, P=0.006). We subsequently grouped all subjects as positive fecal occult blood (259, 16.48%) and negative fecal occult blood (1313, 83.52%), and logistic regression analysis indicated that a prevalence of CSVD was related to positive fecal occult blood (Model 2, adjusted OR=1.50, 95% CI: 1.08-2.08, P=0.015). In addition, the ratios of lacunes (67.18% vs. 53.85%, P<0.001) and enlarged perivascular spaces (43.63% vs. 34.42%, P=0.005) were higher in patients with positive fecal occult blood than in controls. CONCLUSIONS The presence of fecal occult blood is probably related to the risk of CSVD in elderly patients and could be used as a screening tool for CSVD in elderly populations.
Collapse
Affiliation(s)
- Cunsheng Wei
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Xiaorong Yu
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Yuan Chen
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Junying Jiang
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Meng Cao
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Xuemei Chen
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| |
Collapse
|
15
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Jinjin Wang
- Department of Gastroenterology, The First People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Liu Y, Zhao T, Wang Z, Zhang Y, Shen J, Lu B. The microbiome- and metabolome-modulating activity of dietary cholesterol: insights from the small and large intestines. Food Funct 2025; 16:1872-1887. [PMID: 39931947 DOI: 10.1039/d4fo03049d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Cholesterol is an important lipid molecule that affects the gut microbiome upon ingestion. We systematically investigated the effects of cholesterol on the microbiota of the large and small intestines using ex vivo and in vivo models, combining flow cytometry, metabolomics, and metagenomics. The results showed that cholesterol directly causes a loss of bacterial membrane polarity and integrity, as well as a reduction in microbial metabolic activity. Cholesterol directly affected the global metabolism of the large and small intestinal microbiota, including amino acid, carbohydrate, and nucleotide metabolism. Ex vivo and in vivo studies shared similar results, showing that cholesterol increased the abundance of the primary bile acid-metabolizing bacteria Clostridium and Dorea in the large intestinal microbiota, confirming the enrichment effect of cholesterol on these bacteria. In the in vivo model, increased conjugated bile acids in the small intestine and decreased abundance of BSH-containing Bifidobacterium were observed due to cholesterol. Only in vivo models have demonstrated that cholesterol increases phosphatidylcholine levels in both the small and large intestines, which may be related to the effects of cholesterol on host metabolism. The pro-inflammatory capacity of the intestinal microbiota was enhanced by cholesterol, as evidenced by the increased levels of IL-1β and TNF-α in THP-1 cells upon stimulation with cholesterol-treated microbiota. This study comprehensively elucidates the effects of cholesterol on the composition and metabolic functions of the microbiota in both the large and small intestines. It offers a novel perspective on the ways in which cholesterol affects host metabolism via the gut microbiome.
Collapse
Affiliation(s)
- Yan Liu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, 310058, China.
| | - Tian Zhao
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, 310058, China.
| | - Zhangtie Wang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, 310058, China.
| | - Yansong Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, 310058, China.
| | - Jianfu Shen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, 310058, China.
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
17
|
Li Y, Lyu L, Ding H. The potential roles of gut microbiome in porto-sinusoidal vascular disease: an under-researched crossroad. Front Microbiol 2025; 16:1556667. [PMID: 40099185 PMCID: PMC11911366 DOI: 10.3389/fmicb.2025.1556667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Accumulating evidence indicates that patients with liver diseases exhibit distinct microbiological profiles, which can be attributed to the bidirectional relationship of the gut-liver axis. Porto-sinusoidal vascular disease (PSVD) has recently been introduced to describe a group of vascular diseases of the liver, involving the portal venules and sinusoids. Although the pathophysiology of PSVD is not yet fully understood, several predisposing conditions, including immunodeficiency, inflammatory bowel disease, abdominal bacterial infections are associated with the increasing in intestinal permeability and microbial translocation, supporting the role of altered gut microbiota and gut-derived endotoxins in PSVD etiopathogenesis. Recent studies have proposed that the gut microbiome may play a crucial role in the pathophysiology of intrahepatic vascular lesions, potentially influencing the onset and progression of PSVD in this context. This review aims to summarize the current understanding of the gut microbiome's potential role in the pathogenesis of hepatic microvascular abnormalities and thrombosis, and to briefly describe their interactions with PSVD. The insights into gut microbiota and their potential influence on the onset and progression of PSVD may pave the way for new diagnostic, prognostic, and therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing Youan Hospital Affiliated with Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Li P, Jiang W. A New Insight on Atherosclerosis Mechanism and Lipid-Lowering Drugs. Rev Cardiovasc Med 2025; 26:25321. [PMID: 40160588 PMCID: PMC11951287 DOI: 10.31083/rcm25321] [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: 06/21/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 04/02/2025] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease primarily affecting large and medium-sized arteries, involving complex pathological mechanisms such as inflammatory responses, lipid metabolism disorders and vascular plaque formation. In recent years, several emerging research hotspots have appeared in the field of atherosclerosis, including gut microbiota, pyroptosis, ferroptosis, autophagy, cuproptosis, exosomes and non-coding RNA. Traditional lipid-lowering drugs play a crucial role in the treatment of AS but are not able to significantly reverse the pathological changes. This article aims to summarize the latest research progress in the pathogenesis of AS and the diagnosis and treatment of the disease by comprehensively analyzing relevant literature mainly from the past five years. Additionally, the mechanisms of action and research advances of statins, cholesterol absorption inhibitors, fibrates and novel lipid-lowering drugs are reviewed to provide new insights into the diagnosis and treatment of AS.
Collapse
Affiliation(s)
- Penghui Li
- Binhai New Area Hospital of TCM, 300000 Tianjin, China
| | - Wei Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300000 Tianjin, China
| |
Collapse
|
19
|
Huchzermeier R, van der Vorst EPC. Aryl hydrocarbon receptor (AHR) and nuclear factor erythroid-derived 2-like 2 (NRF2): An important crosstalk in the gut-liver axis. Biochem Pharmacol 2025; 233:116785. [PMID: 39890034 DOI: 10.1016/j.bcp.2025.116785] [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: 10/28/2024] [Revised: 12/18/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, mainly involved in detoxification. However, in the intestine, metabolites derived from the diet, which are converted by a wide range of bacteria can also activate the AHR. This intestinal AHR activation plays a key role in maintaining the gut barrier by, for example, upregulating antimicrobial peptides and anti-inflammatory cytokines. Since the gut barrier influences the gut-liver axis by regulating the leaking of metabolites, bacteria, and endotoxins into circulation and particularly into the liver, the AHR is a key factor in the gut-liver axis. Vice versa, certain liver pathologies also influence the gut microbiome, thereby altering bacteria-derived activation of the AHR. Additionally, bile acids can impact the gut via the liver and thereby also affect the AHR. The aryl hydrocarbon receptor (AHR) interacts with several molecular factors, one of which is the nuclear factor erythroid-derived 2-like 2 (NRF2), a transcription factor primarily associated with regulating antioxidant stress responses. The interplay between AHR and NRF2 has been investigated in the context of various diseases; this review highlights the significance of this interaction within the framework of the gut-liver axis.
Collapse
Affiliation(s)
- Rosanna Huchzermeier
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany; Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany; Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany.
| |
Collapse
|
20
|
Zhang L, Yin Y, Jin S. Gut microbial metabolites: The bridge connecting diet and atherosclerosis, and next-generation targets for dietary interventions. Microbiol Res 2025; 292:128037. [PMID: 39752807 DOI: 10.1016/j.micres.2024.128037] [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: 10/08/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Mounting evidence indicates that gut microbial metabolites are central hubs linking the gut microbiota to atherosclerosis (AS). Gut microbiota enriched with pathobiont bacteria responsible for producing metabolites like trimethylamine N-oxide and phenylacetylglutamine are related to an increased risk of cardiovascular events. Furthermore, gut microbiota enriched with bacteria responsible for producing short-chain fatty acids, indole, and its derivatives, such as indole-3-propionic acid, have demonstrated AS-protective effects. This study described AS-related gut microbial composition and how microbial metabolites affect AS. Summary findings revealed gut microbiota and their metabolites-targeted diets could benefit AS treatment. In conclusion, dietary interventions centered on the gut microbiota represent a promising strategy for AS treatment, and understanding diet-microbiota interactions could potentially be devoted to developing novel anti-AS therapies.
Collapse
Affiliation(s)
- Liyin Zhang
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China
| | - Yao Yin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China.
| |
Collapse
|
21
|
Zeng Y, Wu Q, Guo M, Teng F, Jiang C, Chen J, Tan X, Zeng C, Long Y, Law BYK, Xu Y. Gut microbiota-derived imidazole propionate: an emerging target for the prevention and treatment of cardiometabolic diseases. Front Endocrinol (Lausanne) 2025; 16:1409119. [PMID: 40034229 PMCID: PMC11872695 DOI: 10.3389/fendo.2025.1409119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Despite significant advancements in prevention and treatment, cardiometabolic diseases continue to pose a high burden of incidence and mortality. The chronic progression of these diseases necessitates the identification of early and complementary therapeutic targets to elucidate and mitigate residual risks in patient care. The gut microbiota acts as a sentinel between internal and external environments, transmitting modified risks associated with these factors to the host. Imidazole propionate (ImP), a histidine metabolite originating from the gut microbiota, gained attention after being found to impair glucose tolerance and insulin signaling several years ago. Epidemiological studies over the past five years have demonstrated a robust correlation between ImP and an increased risk of onset of type 2 diabetes (T2D) and obesity, exacerbation of kidney traits in chronic kidney disease (CKD), progression of atherosclerotic plaques, and elevated mortality rates in heart failure (HF). These findings suggest that ImP may serve as a pivotal target for the prevention and treatment of cardiometabolic diseases. Mechanistic insights have uncovered associations between ImP and insulin resistance, impaired glucose metabolism, chronic inflammation, and intestinal barrier damage. This review provides a comprehensive summary of the current evidence regarding the association between ImP and cardiometabolic impairment, highlighting its potential in advancing personalized approaches to disease prevention and management, and exploring the intricate interplay of diet, gut microbiota, and ImP in cardiovascular metabolic impairment. Overall, this review offers valuable insights into the multifaceted roles of ImP in cardiometabolic diseases, identifies current knowledge gaps, and discusses future research directions.
Collapse
Affiliation(s)
- Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, The Afiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Man Guo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fangyuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxia Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Long
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
22
|
Liu J, Huang S. Dietary index for gut microbiota is associated with stroke among US adults. Food Funct 2025; 16:1458-1468. [PMID: 39898733 DOI: 10.1039/d4fo04649h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Aims: Emerging evidence underscores the diet-microbiota-gut-brain axis as vital to brain health. The dietary index for gut microbiota (DI-GM), quantifying diet quality linked to gut microbiota diversity, reflects healthier gut microbiota with higher scores. Therefore, this study was designed to explore the unclear association between DI-GM and stroke. Methods: A cross-sectional analysis was conducted using data from 48 677 participants aged ≥20 years in the National Health and Nutrition Examination Survey (NHANES). Demographic and dietary data were collected, and multivariable weighted logistic regression analysis was performed to evaluate the association between the DI-GM and stroke. Additionally, restricted cubic spline (RCS), subgroup analyses, and receiver operating characteristic (ROC) curve were conducted. Results: In participants aged ≥20 years, the odds ratio (OR) was 0.96 (95% CI: 0.92-1.00, P = 0.075) in the crude model, but after adjustment, the OR was 0.93 (95% CI: 0.89-0.98, P = 0.003), while higher beneficial to gut microbiota scores were consistently associated with lower stroke prevalence with ORs of 0.87 (95% CI: 0.83-0.90, P < 0.001) in the crude model and 0.88 (95% CI: 0.83-0.93, P < 0.001) after adjustment. Among participants aged 20-29 years, no significant association was observed. For those aged ≥30 years, higher DI-GM and beneficial to gut microbiota scores were associated with lower stroke prevalence, with DI-GM showing ORs of 0.93 (95% CI: 0.89-0.97, P < 0.001) in the crude model and 0.93 (95% CI: 0.89-0.98, P = 0.003) after adjustment, and beneficial to gut microbiota scores showing ORs of 0.82 (95% CI: 0.79-0.86, P < 0.001) in the crude model and 0.88 (95% CI: 0.83-0.93, P < 0.001) after adjustment. RCS indicated a linear relationship between DI-GM and stroke. Conclusion: The DI-GM was inversely and linearly associated with stroke prevalence, particularly in adults aged 30 years and above.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China.
| | - Shaoqiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China.
| |
Collapse
|
23
|
Wang X, Cui J, Gu Z, Guo L, Liu R, Guo Y, Qin N, Yang Y. Aged garlic oligosaccharides modulate host metabolism and gut microbiota to alleviate high-fat and high-cholesterol diet-induced atherosclerosis in ApoE -/- mice. Food Chem 2025; 463:141409. [PMID: 39326312 DOI: 10.1016/j.foodchem.2024.141409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Atherosclerosis (AS) is a cardiovascular disease caused by excessive accumulation of lipids in arterial walls. In this study, we developed an AS model in ApoE-/- mice using a high-fat, high-cholesterol diet and investigated the anti-AS mechanism of aged garlic oligosaccharides (AGOs) by focusing on the gut microbiota. Results revealed that AGOs exhibited significant anti-AS effects, reduced trimethylamine N-oxide levels from 349.9 to 189.2 ng/mL, and reduced aortic lipid deposition from 31.7 % to 9.5 %. AGOs significantly increased the levels of short-chain fatty acids in feces, in which acetic, propionic, and butyric acids were increased from 1.580, 0.364, and 0.469 mg/g to 2.233, 0.774, and 0.881 mg/g, respectively. An analysis of the gut microbiota indicated that AGOs restored alpha and beta diversity, decreased the Firmicutes/Bacteroidetes ratio, and promoted the dominance of the genus Akkermansia. A metagenomic analysis revealed that AGOs alleviated AS through the ABC transporter pathway and the lipopolysaccharide biosynthesis pathway.
Collapse
Affiliation(s)
- Xiaomin Wang
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Jianglu Cui
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Ziyao Gu
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Lili Guo
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Rui Liu
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China
| | - Yu Guo
- Shanxi Agricultural Products Quality and Safety Center, Taiyuan 030006, China
| | - Nan Qin
- Institute of Pharmaceutical and Food Engineering, Shanxi University of Chinese Medicine, Yuci 030619, China.
| | - Yukun Yang
- School of Life Science, Xinghuacun College (Shanxi Institute of Brewing Technology and Industry), Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
24
|
Revol-Cavalier J, Quaranta A, Newman JW, Brash AR, Hamberg M, Wheelock CE. The Octadecanoids: Synthesis and Bioactivity of 18-Carbon Oxygenated Fatty Acids in Mammals, Bacteria, and Fungi. Chem Rev 2025; 125:1-90. [PMID: 39680864 PMCID: PMC11719350 DOI: 10.1021/acs.chemrev.3c00520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The octadecanoids are a broad class of lipids consisting of the oxygenated products of 18-carbon fatty acids. Originally referring to production of the phytohormone jasmonic acid, the octadecanoid pathway has been expanded to include products of all 18-carbon fatty acids. Octadecanoids are formed biosynthetically in mammals via cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) activity, as well as nonenzymatically by photo- and autoxidation mechanisms. While octadecanoids are well-known mediators in plants, their role in the regulation of mammalian biological processes has been generally neglected. However, there have been significant advancements in recognizing the importance of these compounds in mammals and their involvement in the mediation of inflammation, nociception, and cell proliferation, as well as in immuno- and tissue modulation, coagulation processes, hormone regulation, and skin barrier formation. More recently, the gut microbiome has been shown to be a significant source of octadecanoid biosynthesis, providing additional biosynthetic routes including hydratase activity (e.g., CLA-HY, FA-HY1, FA-HY2). In this review, we summarize the current field of octadecanoids, propose standardized nomenclature, provide details of octadecanoid preparation and measurement, summarize the phase-I metabolic pathway of octadecanoid formation in mammals, bacteria, and fungi, and describe their biological activity in relation to mammalian pathophysiology as well as their potential use as biomarkers of health and disease.
Collapse
Affiliation(s)
- Johanna Revol-Cavalier
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Alessandro Quaranta
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - John W. Newman
- Western
Human Nutrition Research Center, Agricultural
Research Service, USDA, Davis, California 95616, United States
- Department
of Nutrition, University of California, Davis, Davis, California 95616, United States
- West
Coast Metabolomics Center, Genome Center, University of California, Davis, Davis, California 95616, United States
| | - Alan R. Brash
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Mats Hamberg
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Craig E. Wheelock
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm SE-141-86, Sweden
| |
Collapse
|
25
|
Garcia-Fernandez H, Alcala-Diaz JF, Quintana-Navarro GM, Lopez-Moreno J, Luque-Cordoba D, Ruiz-Diaz Narvaez E, Arenas-de Larriva AP, Gutierrez-Mariscal FM, Torres-Peña JD, Rodriguez-Cano D, Luque RM, Priego-Capote F, Lopez-Miranda J, Camargo A. Trimethylamine Oxidation into the Proatherogenic Trimethylamine N-Oxide Is Higher in Coronary Heart Disease Men: From the CORDIOPREV Study. World J Mens Health 2025; 43:249-258. [PMID: 39344118 PMCID: PMC11704170 DOI: 10.5534/wjmh.230366] [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: 12/13/2023] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE Cardiovascular disease (CVD) is more prevalent in men than women, but the mechanisms responsible for this are not fully understood. We aimed to evaluate differences in trimethylamine (TMA), a microbial metabolite and its oxidized form, trimethylamine N-oxide (TMAO), which is thought to promote atherosclerosis, between men and women with coronary heart disease (CHD), using as a reference a non-CVD population. MATERIALS AND METHODS This study was carried out within the framework of the CORDIOPREV study (NCT00924937; June 19, 2009), a clinical trial which included 827 men and 175 women with CHD, with a non-CVD population of 375 individuals (270 men and 105 women) as a reference group. Plasma TMA and TMAO were measured by HPLC-MS/MS. The carotid study was ultrasonically assessed bilaterally by the quantification of intima-media thickness of both common carotid arteries (IMT-CC). RESULTS We found higher TMAO levels and TMAO/TMA ratio in CHD men than CHD women (p=0.034 and p=0.026, respectively). No TMA sex differences were found in CHD patients. The TMA and TMAO levels and TMAO/TMA ratio were lower, and no differences between sexes were found in the non-CVD population. TMAO levels in CHD patients were consistent with higher IMT-CC and more carotid plaques (p=0.032 and p=0.037, respectively) and lower cholesterol efflux in CHD men than CHD women (p<0.001). CONCLUSIONS Our results suggest that CHD men have augmented TMAO levels compared with CHD women, presumably as a consequence of higher rate of TMA to TMAO oxidation, which could be associated with CVD, as these sex differences are not observed in a non-CVD population.
Collapse
Affiliation(s)
- Helena Garcia-Fernandez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Gracia M Quintana-Navarro
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Lopez-Moreno
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Diego Luque-Cordoba
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Eugenia Ruiz-Diaz Narvaez
- Department of Clinical Nutrition and Diet Therapy, Clinics Hospital, Faculty of Medical Sciences, National University of Asuncion, San Lorenzo, Paraguay
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Raul M Luque
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Feliciano Priego-Capote
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
26
|
Escobar C, Aldeguer X, Vivas D, Manzano Fernández S, Gonzalez Caballero E, Garcia Martín A, Barrios V, Freixa-Pamias R. The gut microbiota and its role in the development of cardiovascular disease. Expert Rev Cardiovasc Ther 2025; 23:23-34. [PMID: 39915986 DOI: 10.1080/14779072.2025.2463366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION The pathophysiology of cardiovascular diseases encompasses a complex interplay of genetic and environmental risk factors. Even if traditional risk factors are treated to target, there remains a residual risk. AREAS COVERED This manuscript reviews the potential role of gut microbiota in the development of cardiovascular disease, and as potential target. A systematic search was conducted until 30 October 2024 on PubMed (MEDLINE), using the MeSH terms [Gut microbiota] + [Dysbiosis] + [Cardiovascular] + [TMAO] + [bile acids] + [short-chain fatty acids]. EXPERT OPINION The term dysbiosis implies changes in equilibrium, with modifications in the composition and functionality of microbiota and a series of additional factors: reduced diversity and uniformity of microorganisms; reduced short-chain fatty acid-producing bacteria; increased gut permeability; release of metabolites, such as trimethylamine N-oxide, betaine, phenylalanine, tryptophan-kynurenine, phenylacetylglutamine, and lipopolysaccharides; and reduced secondary bile acid excretion, leading to inflammation, oxidative stress, and endothelial dysfunction and facilitating the onset of pathological conditions, including obesity, hypertension, diabetes, atherosclerosis, and heart failure. Attempts to restore gut microbiota balance through different interventions, mainly changes in diet, have been shown to positively affect individual components and metabolites and reduce the risk of cardiovascular disease. In addition, probiotics and prebiotics are potentially useful. Fecal microbiota transplantation is a promising therapy.
Collapse
Affiliation(s)
- Carlos Escobar
- Cardiology Department, University Hospital La Paz, Madrid, Spain
| | - Xavier Aldeguer
- Gastroenterology Department, Hospital Doctor Josep Trueta i Santa Caterina, Institut d'investigació Biomèdica de Girona IDIBGI, Girona/Salt, Spain
| | - David Vivas
- Cardiovascular Institute, San Carlos University Hospital, Madrid, Spain
- Cardiology Department, Cardiovascular Institute Vithas Milagrosa and Aravaca, Madrid, Spain
| | | | | | - Ana Garcia Martín
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Vivencio Barrios
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Román Freixa-Pamias
- Cardiology Department, Complex Hospitalari Moisès Broggi, Sant Joan Despí, Barcelona, Spain
| |
Collapse
|
27
|
Dong Y, Zhang Q, Xie R, Zhao J, Han Z, Li Y, Yu H, Zhang Y. Tremella fuciformis Berk Alleviated Atherosclerosis Symptoms via Nuclear Factor-Kappa B-Mediated Inflammatory Response in ApoE -/- Mice. Nutrients 2024; 17:160. [PMID: 39796594 PMCID: PMC11722796 DOI: 10.3390/nu17010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Atherosclerosis, a persistent inflammatory disease marked by the presence of atherosclerotic plaques or fibrous plaques, is a significant contributor to the onset of the development of cardiovascular disease. Tremella fuciformis Berk contains various active ingredients that have anti-inflammatory, antioxidant, and hypolipidemic properties. Nevertheless, the potential effects of T. fuciformis on atherosclerosis have not been systematically reported. METHOD In this study, ApoE-/- mice were employed as models of atherosclerosis caused by a high-fat diet (HFD) to investigate the effect of T. fuciformis. Gut microbiota and serum metabolism analysis were performed to elucidate the potential mechanism of T. fuciformis for its anti-atherosclerosis effects. RESULTS T. fuciformis significantly decreased the aortic root wall thickness and the area of lipid droplets, regulated lipid levels, and inhibited fat accumulation to improve aortic root lesions. Furthermore, T. fuciformis significantly altered serum metabolite (including diethyl phthalate and succinate) levels, regulated the abundance of microbiota, such as Coriobacteriaceae_UCG-002 and Alistipes, and suppressed the inflammatory response to ameliorate atherosclerosis via the nuclear factor-kappa B (NF-κB)-mediated inflammatory response in HFD-induced ApoE-/- mice. CONCLUSIONS These results offer a theoretical basis and data to support T. fuciformis as a potential strategy for treating atherosclerosis.
Collapse
Affiliation(s)
- Yihao Dong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (Q.Z.); (R.X.); (J.Z.); (Z.H.); (Y.L.)
| | - Qinchun Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (Q.Z.); (R.X.); (J.Z.); (Z.H.); (Y.L.)
| | - Rui Xie
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (Q.Z.); (R.X.); (J.Z.); (Z.H.); (Y.L.)
| | - Jundi Zhao
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (Q.Z.); (R.X.); (J.Z.); (Z.H.); (Y.L.)
| | - Zhihua Han
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (Q.Z.); (R.X.); (J.Z.); (Z.H.); (Y.L.)
| | - Yu Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (Q.Z.); (R.X.); (J.Z.); (Z.H.); (Y.L.)
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin 300308, China
| | - Han Yu
- College of Agriculture, Jilin Agricultural University, Changchun 130118, China
| | - Yongfeng Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China; (Y.D.); (Q.Z.); (R.X.); (J.Z.); (Z.H.); (Y.L.)
| |
Collapse
|
28
|
Qu J, Meng F, Wang Z, Xu W. Unlocking Cardioprotective Potential of Gut Microbiome: Exploring Therapeutic Strategies. J Microbiol Biotechnol 2024; 34:2413-2424. [PMID: 39467697 PMCID: PMC11729380 DOI: 10.4014/jmb.2405.05019] [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: 05/17/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 10/30/2024]
Abstract
The microbial community inhabiting the human gut resembles a bustling metropolis, wherein beneficial bacteria play pivotal roles in regulating our bodily functions. These microorganisms adeptly break down resilient dietary fibers to fuel our energy, synthesize essential vitamins crucial for our well-being, and maintain the delicate balance of our immune system. Recent research indicates a potential correlation between alterations in the composition and activities of these gut microbes and the development of coronary artery disease (CAD). Consequently, scientists are delving into the intriguing realm of manipulating these gut inhabitants to potentially mitigate disease risks. Various promising strategies have emerged in this endeavor. Studies have evidenced that probiotics can mitigate inflammation and enhance the endothelial health of our blood vessels. Notably, strains such as Lactobacilli and Bifidobacteria have garnered substantial attention in both laboratory settings and clinical trials. Conversely, prebiotics exhibit anti-inflammatory properties and hold potential in managing conditions like hypertension and hypercholesterolemia. Synbiotics, which synergistically combine probiotics and prebiotics, show promise in regulating glucose metabolism and abnormal lipid profiles. However, uncertainties persist regarding postbiotics, while antibiotics are deemed unsuitable due to their potential adverse effects. On the other hand, TMAO blockers, such as 3,3-dimethyl-1-butanol, demonstrate encouraging outcomes in laboratory experiments owing to their anti-inflammatory and tissue-protective properties. Moreover, fecal transplantation, despite yielding mixed results, warrants further exploration and refinement. In this comprehensive review, we delve into the intricate interplay between the gut microbiota and CAD, shedding light on the multifaceted approaches researchers are employing to leverage this understanding for therapeutic advancements.
Collapse
Affiliation(s)
- Jun Qu
- Department of Internal Medicine-Cardiovascular, YanTai YuHuangDing Hospital, Yantai, Shandong, P.R. China
| | - Fantao Meng
- Department of Internal Medicine-Cardiovascular, LinYi Central Hospital, LinYi, Shandong, P.R. China
| | - Zhen Wang
- Department of Internal Medicine-Cardiovascular, YanTai YuHuangDing Hospital, Yantai, Shandong, P.R. China
| | - Wenhao Xu
- Department of Internal Medicine-Cardiovascular, YanTai YuHuangDing Hospital, Yantai, Shandong, P.R. China
| |
Collapse
|
29
|
Chen X, Kang Y, Tang C, Zhang L, Guo L. TLR4 promotes smooth muscle cell-derived foam cells formation by inducing receptor-independent macropinocytosis. Biosci Biotechnol Biochem 2024; 89:22-32. [PMID: 39455413 DOI: 10.1093/bbb/zbae153] [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: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Foam cells are primarily formed through scavenger receptors that mediate the uptake of various modified low-density lipoproteins (LDL) into cells. In addition to the receptor-dependent pathway, macropinocytosis is an essential nonreceptor endocytic pathway for vascular smooth muscle cells (VSMCs) to take up lipids. However, the molecular mechanisms underlying this process remain unclear. Primary cultured VSMCs were stimulated with 200 ng/mL lipopolysaccharide (LPS) and 200 µg/mL native LDL (nLDL). We observed a significant increase in Toll-like receptor 4 (TLR4) protein expression and a significant activation of macropinocytosis, which correlated with the highest uptake of nLDL and intracellular lipid deposition in WT VSMCs. However, macropinocytosis was inhibited and lipid accumulation decreased after treatment with macropinocytosis inhibitors and Syk inhibitors in WT VSMCs. Consistently, TLR4 knockout significantly suppressed macropinocytosis and lipid droplets accumulation in VSMCs. Taken together, our findings suggest a critical role of TLR4/Syk signaling in promoting receptor-independent macropinocytosis leading to VSMC-derived foam cells formation.
Collapse
MESH Headings
- Toll-Like Receptor 4/metabolism
- Pinocytosis/drug effects
- Animals
- Foam Cells/metabolism
- Foam Cells/cytology
- Foam Cells/drug effects
- Syk Kinase/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Signal Transduction
- Lipopolysaccharides/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Mice
- Lipoproteins, LDL/metabolism
- Lipoproteins, LDL/pharmacology
- Cells, Cultured
- Mice, Knockout
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Xue Chen
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Yulai Kang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Chunhua Tang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lili Zhang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Lu Guo
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
30
|
Liu L, Ma Y, Xu Y, Liu B, Wang C, Feng J, Li M, Yin H, Sun L, Li P, Li ZH. Mechanisms of eco-corona effects on micro(nano)plastics in marine medaka: Insights into translocation, immunity, and energy metabolism. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136236. [PMID: 39442301 DOI: 10.1016/j.jhazmat.2024.136236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/06/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Biomolecules, prevalent in the marine environment, can readily adsorb onto the surface of micro(nano)plastics (MNPs), forming eco-corona. This study indicated that 50 nm polystyrene nanoplastics (NP50), whether wrapped with eco-corona or not, can passively enter embryos, whereas 5 µm polystyrene microplastics (MP5) cannot. Additionally, translocation of MP5 from the intestine to the liver was observed in larvae, a process facilitated by eco-corona. Notably, eco-corona prolonged the retention time of MNPs in larvae. However, NP50 was more challenging to purify than MP5, irrespective of the presence of eco-corona. Interestingly, eco-corona degraded in the intestine during the uptake of MNPs, and the hard coronae that readily formed on NP50 may restrict the degradation rate. Although NP50 significantly disrupted larval microbiota homeostasis compared with MP5, eco-corona was more likely to exacerbate MP5's damage to the intestine and liver by disrupting microbiota homeostasis. Additionally, NP50 caused more significant damage to immunity and energy metabolism compared with MP5, regardless of the presence of eco-corona. This study revealed that previously overlooked biomolecules in the marine environment can enhance the translocation of MNPs and subsequently exacerbate their toxic effects, providing theoretical support for assessing the ecological risks of MNPs in real environments.
Collapse
Affiliation(s)
- Ling Liu
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Yuqing Ma
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Yanan Xu
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Bin Liu
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Cunlong Wang
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Jianxue Feng
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Mingyang Li
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Haiyang Yin
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Le Sun
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Ping Li
- Marine College, Shandong University, Weihai, Shandong 264209, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| |
Collapse
|
31
|
Liu M, Du X, Chen H, Bai C, Lan L. Systemic investigation of di-isobutyl phthalate (DIBP) exposure in the risk of cardiovascular via influencing the gut microbiota arachidonic acid metabolism in obese mice model. Regen Ther 2024; 27:290-300. [PMID: 38638558 PMCID: PMC11024931 DOI: 10.1016/j.reth.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/17/2024] [Accepted: 03/24/2024] [Indexed: 04/20/2024] Open
Abstract
Phthalate esters (PE), a significant class of organic compounds used in industry, can contaminate humans and animals by entering water and food chains. Recent studies demonstrate the influence of PE on the development and progression of heart diseases, particularly in obese people. Di-isobutyl phthalate (DIBP) was administered orally to normal and diet-induced obese mice in this research to assess cardiovascular risk. The modifications in the microbial composition and metabolites were examined using RNA sequencing and mass spectrometry analysis. Based on the findings, lean group rodents were less susceptible to DIBP exposure than fat mice because of their cardiovascular systems. Histopathology examinations of mice fed a high-fat diet revealed lesions and plagues that suggested a cardiovascular risk. In the chronic DIBP microbial remodeling metagenomics Faecalibaculum rodentium was the predominant genera in obese mice. According to metabolomics data, arachidonic acid (AA) metabolism changes caused by DIBP were linked to unfavorable cardiovascular events. Our research offers new understandings of the cardiovascular damage caused by DIBP exposure in obese people and raises the possibility that arachidonic acid metabolism could be used as a regulator of the gut microbiota to avert related diseases.
Collapse
Affiliation(s)
- Min Liu
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Xifeng Du
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Huifang Chen
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Chenkai Bai
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Lizhen Lan
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
32
|
Gan G, Zhang R, Zeng Y, Lu B, Luo Y, Chen S, Lei H, Cai Z, Huang X. Fecal microbiota transplantation validates the importance of gut microbiota in an ApoE -/- mouse model of chronic apical periodontitis-induced atherosclerosis. BMC Oral Health 2024; 24:1455. [PMID: 39614243 DOI: 10.1186/s12903-024-05230-5] [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: 05/16/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Chronic apical periodontitis (CAP) has been linked to the development of atherosclerosis, although the underlying mechanisms remain unclear. This study aimed to investigate the role of gut microbiota disruption in CAP-induced atherosclerosis development, focusing on trimethylamine N-oxide (TMAO)-related metabolites. METHODS The study utilized fecal microbiota transplantation (FMT) to transfer gut microbiota from mice with CAP to healthy mice. Atherosclerosis development was assessed by analyzing lesions in the aortic arch and aortic root. Serum lipid and inflammatory factor levels were measured. Composition and diversity of gut microbiota were analyzed using targeted metabolomics, with a focus on the ratio of Firmicutes to Bacteroidetes. The expression of hepatic flavin-containing monooxygenase 3 (FMO3) and serum TMAO levels were also evaluated. RESULTS Mice receiving gut microbiota from CAP mice showed increased atherosclerotic lesions compared to controls, without significant differences in serum lipid or inflammatory factor levels. Alterations in gut microbiota composition were observed, characterized by an increase in the Firmicutes to Bacteroidetes ratio. Peptostreptococcaceae abundance positively correlated with atherosclerosis severity, while Odoribacteraceae showed a negative correlation. No significant differences were found in hepatic FMO3 expression or serum TMAO levels. CONCLUSIONS The study confirms the role of gut microbiota disruption in CAP-mediated atherosclerosis development, independent of serum lipid or TMAO levels. Alterations in gut microbiota composition, particularly increased Firmicutes to Bacteroidetes ratio and specific bacterial families, were associated with atherosclerosis severity. These findings highlight the intricate interplay between gut microbiota and cardiovascular health in the context of CAP.
Collapse
Affiliation(s)
- Guowu Gan
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ren Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yu Zeng
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Beibei Lu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yufang Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Huaxiang Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhiyu Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Clinical Research Center for Oral Tissue Deficiency Diseases of Fujian Province, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
33
|
Buelna-Chontal M. Coffee: Fuel for Your Day or Foe for Your Arteries. Antioxidants (Basel) 2024; 13:1455. [PMID: 39765784 PMCID: PMC11672806 DOI: 10.3390/antiox13121455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Atherosclerosis, a major cause of cardiovascular diseases, is influenced by modifiable factors such as adiposity and blood cholesterol. Diet is crucial in these areas, particularly regarding antioxidant, inflammatory, and obesity effects. Coffee, a globally popular stimulant beverage, has garnered significant attention for its potential impact on cardiovascular diseases. Recent insights reinforce the need to re-examine the relationship between coffee consumption and atherosclerosis progression. Coffee's complex composition includes polyphenols, renowned for their antioxidant and anti-inflammatory properties as well as potential weight-reducing effects. In addition, studies have demonstrated that certain coffee compounds such as chlorogenic acid, caffeic, p-coumaric, and ferulic acid can prevent atherogenesis by preventing the oxidation of low-density lipoproteins. Conversely, diterpenes, found in some coffee brews, can elevate cholesterol levels, posing a risk to coronary health. Notably, coffee intake has been shown to influence gut microbiota diversity, potentially contributing to anti-obesity effects. This review explores the insights from preclinical and clinical studies investigating the potential mechanisms through which coffee consumption may reduce the risk of atherosclerosis-highlighting the potential benefits of moderate filtered coffee consumption and the potential risks associated with excessive coffee consumption. Understanding this relationship is crucial for informing public health recommendations and guiding future research.
Collapse
Affiliation(s)
- Mabel Buelna-Chontal
- Department of Cardiovascular Biomedicine, National Institute of Cardiology, Ignacio Chavez, Mexico City 14080, Mexico
| |
Collapse
|
34
|
Protasiewicz-Timofticiuc DC, Bădescu D, Moța M, Ștefan AG, Mitrea A, Clenciu D, Efrem IC, Roșu MM, Vladu BE, Gheonea TC, Moța E, Vladu IM. Back to Roots: Dysbiosis, Obesity, Metabolic Syndrome, Type 2 Diabetes Mellitus, and Obstructive Sleep Apnea-Is There an Objective Connection? A Narrative Review. Nutrients 2024; 16:4057. [PMID: 39683451 DOI: 10.3390/nu16234057] [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: 10/27/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
In recent decades, it has become clear that the gut is more than just a digestive organ; it also functions as an immune organ with regulatory capabilities and acts as a "second brain" that influences brain function due to the presence and regulatory roles of the gut microbiota (GM). The GM is a crucial component of its host and significantly impacts human health. Dysbiosis, or microbial imbalance, has been closely linked to various diseases, including gastrointestinal, neurological, psychiatric, and metabolic disorders. The aim of this narrative review is to highlight the roles of the GM in maintaining metabolic health. Sleep is a vital biological necessity, with living organisms having evolved an internal sleep-wake rhythm that aligns with a roughly 24 h light/dark cycle, and this is known as the circadian rhythm. This cycle is essential for tissue repair, restoration, and overall optimal body functioning. Sleep irregularities have become more prevalent in modern society, with fast-paced lifestyles often disrupting normal sleep patterns. Urban living factors, such as fast food consumption, shift work, exposure to artificial light and nighttime noise, medications, and social activities, can adversely affect circadian rhythms, with dysbiosis being one of the many factors incriminated in the etiology of sleep disorders.
Collapse
Affiliation(s)
| | - Diana Bădescu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Maria Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Cristian Efrem
- Department of Medical Semiology, Faculty of Dentistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Magdalena Roșu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Beatrice Elena Vladu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Theodora Claudia Gheonea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
35
|
Xue J, Allaband C, Zuffa S, Poulsen O, Meadows J, Zhou D, Dorrestein PC, Knight R, Haddad GG. Gut Microbiota and Derived Metabolites Mediate Obstructive Sleep Apnea Induced Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624205. [PMID: 39605650 PMCID: PMC11601605 DOI: 10.1101/2024.11.18.624205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia/hypercapnia (IHC), affects predominantly obese individuals, and increases atherosclerosis risk. Since we and others have implicated gut microbiota and metabolites in atherogenesis, we dissected their contributions to OSA-induced atherosclerosis. Results Atherosclerotic lesions were compared between conventionally-reared specific pathogen free (SPF) and germ-free (GF) ApoE -/- mice following a high fat high cholesterol diet (HFHC), with and without IHC conditions. The fecal microbiota and metabolome were profiled using 16S rRNA gene amplicon sequencing and untargeted tandem mass spectrometry (LC-MS/MS) respectively. Phenotypic data showed that HFHC significantly increased atherosclerosis as compared to regular chow (RC) in both aorta and pulmonary artery (PA) of SPF mice. IHC exacerbated lesions in addition to HFHC. Differential abundance analysis of gut microbiota identified an enrichment of Akkermansiaceae and a depletion of Muribaculaceae (formerly S24-7) family members in the HFHC-IHC group. LC-MS/MS showed a dysregulation of bile acid profiles with taurocholic acid, taurodeoxycholic acid, and 12-ketodeoxycholic acid enriched in the HFHC-IHC group, long-chain N-acyl amides, and phosphatidylcholines. Interestingly, GF ApoE -/- mice markedly reduced atherosclerotic formation relative to SPF ApoE -/- mice in the aorta under HFHC/IHC conditions. In contrast, microbial colonization did not show a significant impact on the atherosclerotic progression in PA. Conclusions In summary, this research demonstrated that (1) IHC acts cooperatively with HFHC to induce atherosclerosis; (2) gut microbiota modulate atherogenesis, induced by HFHC/IHC, in the aorta not in PA; (3) different analytical methods suggest that a specific imbalance between Akkermansiaceae and Muribaculaceae bacterial families mediate OSA-induced atherosclerosis; and (4) derived bile acids, such as deoxycholic acid and lithocholic acid, regulate atherosclerosis in OSA. The knowledge obtained provides novel insights into the potential therapeutic approaches to prevent and treat OSA-induced atherosclerosis.
Collapse
|
36
|
Khalil M, Di Ciaula A, Mahdi L, Jaber N, Di Palo DM, Graziani A, Baffy G, Portincasa P. Unraveling the Role of the Human Gut Microbiome in Health and Diseases. Microorganisms 2024; 12:2333. [PMID: 39597722 PMCID: PMC11596745 DOI: 10.3390/microorganisms12112333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The human gut is a complex ecosystem that supports billions of living species, including bacteria, viruses, archaea, phages, fungi, and unicellular eukaryotes. Bacteria give genes and enzymes for microbial and host-produced compounds, establishing a symbiotic link between the external environment and the host at both the gut and systemic levels. The gut microbiome, which is primarily made up of commensal bacteria, is critical for maintaining the healthy host's immune system, aiding digestion, synthesizing essential nutrients, and protecting against pathogenic bacteria, as well as influencing endocrine, neural, humoral, and immunological functions and metabolic pathways. Qualitative, quantitative, and/or topographic shifts can alter the gut microbiome, resulting in dysbiosis and microbial dysfunction, which can contribute to a variety of noncommunicable illnesses, including hypertension, cardiovascular disease, obesity, diabetes, inflammatory bowel disease, cancer, and irritable bowel syndrome. While most evidence to date is observational and does not establish direct causation, ongoing clinical trials and advanced genomic techniques are steadily enhancing our understanding of these intricate interactions. This review will explore key aspects of the relationship between gut microbiota, eubiosis, and dysbiosis in human health and disease, highlighting emerging strategies for microbiome engineering as potential therapeutic approaches for various conditions.
Collapse
Affiliation(s)
- Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Nour Jaber
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Domenica Maria Di Palo
- Division of Hygiene, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02130, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| |
Collapse
|
37
|
Puerarin alleviates atherosclerosis via the inhibition of Prevotella copri and its trimethylamine production. Gut 2024; 73:1934-1943. [PMID: 38777572 DOI: 10.1136/gutjnl-2024-331880] [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: 01/01/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Puerarin (PU) is a natural compound that exhibits limited oral bioavailability but has shown promise in the treatment of atherosclerosis (AS). However, the precise mechanisms underlying its therapeutic effects remain incompletely understood. This study aimed to investigate the effects of PU and its mechanisms in mitigating AS in both mice and humans. DESIGN The impact of PU on AS was examined in ApoE -/- mice fed a high-fat diet (HFD) and in human patients with carotid artery plaque. To explore the causal link between PU-associated gut microbiota and AS, faecal microbiota transplantation (FMT) and mono-colonisation of mice with Prevotella copri (P. copri) were employed. RESULTS PU alleviated AS by modulating the gut microbiota, as evidenced by alterations in gut microbiota composition and the amelioration of AS following FMT from PU-treated mice into ApoE-/- mice fed HFD. Specifically, PU reduced the abundance of P. copri, which exacerbated AS by producing trimethylamine (TMA). Prolonged mono-colonisation of P. copri undermines the beneficial effects of PU on AS. In clinical, the plaque scores of AS patients were positively correlated with the abundance of P. copri and plasma trimethylamine-N-oxide (TMAO) levels. A 1-week oral intervention with PU effectively decreased P. copri levels and reduced TMAO concentrations in patients with carotid artery plaque. CONCLUSION PU may provide therapeutic benefits in combating AS by targeting P. copri and its production of TMA. TRIAL REGISTRATION NUMBER ChiCTR1900022488.
Collapse
|
38
|
Kis-György R, Körtési T, Anicka A, Nagy-Grócz G. The Connection Between the Oral Microbiota and the Kynurenine Pathway: Insights into Oral and Certain Systemic Disorders. Curr Issues Mol Biol 2024; 46:12641-12657. [PMID: 39590344 PMCID: PMC11593024 DOI: 10.3390/cimb46110750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The oral microbiome, comprising bacteria, fungi, viruses, and protozoa, is essential for maintaining both oral and systemic health. This complex ecosystem includes over 700 bacterial species, such as Streptococcus mutans, which contributes to dental caries through acid production that demineralizes tooth enamel. Fungi like Candida and pathogens such as Porphyromonas gingivalis are also significant, as they can lead to periodontal diseases through inflammation and destruction of tooth-supporting structures. Dysbiosis, or microbial imbalance, is a key factor in the development of these oral diseases. Understanding the composition and functions of the oral microbiome is vital for creating targeted therapies for these conditions. Additionally, the kynurenine pathway, which processes the amino acid tryptophan, plays a crucial role in immune regulation, neuroprotection, and inflammation. Oral bacteria can metabolize tryptophan, influencing the production of kynurenine, kynurenic acid, and quinolinic acid, thereby affecting the kynurenine system. The balance of microbial species in the oral cavity can impact tryptophan levels and its metabolites. This narrative review aims to explore the relationship between the oral microbiome, oral diseases, and the kynurenine system in relation to certain systemic diseases.
Collapse
Affiliation(s)
- Rita Kis-György
- Section of Health Behaviour and Health Promotion, Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary;
- Doctoral School of Interdisciplinary Medicine, University of Szeged, Szőkefalvi–Nagy Béla u. 4/B, H-6720 Szeged, Hungary
| | - Tamás Körtési
- Department of Theoretical Health Sciences and Health Management, Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary;
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, Danube Neuroscience Research Laboratory, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Alexandra Anicka
- Department of Obstetrics and Gynecology, Semmelweis University, Üllői Út 78/A, H-1182 Budapest, Hungary;
| | - Gábor Nagy-Grócz
- Department of Theoretical Health Sciences and Health Management, Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary;
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
39
|
Mansour H, Slika H, Nasser SA, Pintus G, Khachab M, Sahebkar A, Eid AH. Flavonoids, gut microbiota and cardiovascular disease: Dynamics and interplay. Pharmacol Res 2024; 209:107452. [PMID: 39383791 DOI: 10.1016/j.phrs.2024.107452] [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: 01/28/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Cardiovascular disease (CVD) remains the leading cause of global morbidity and mortality. Extensive efforts have been invested to explicate mechanisms implicated in the onset and progression of CVD. Besides the usual suspects as risk factors (obesity, diabetes, and others), the gut microbiome has emerged as a prominent and essential factor in the pathogenesis of CVD. With its endocrine-like effects, the microbiome modulates many physiologic processes. As such, it is not surprising that dysbiosis-by generating metabolites, inciting inflammation, and altering secondary bile acid signaling- could predispose to or aggravate CVD. Nevertheless, various natural and synthetic compounds have been shown to modulate the microbiome. Prime among these molecules are flavonoids, which are natural polyphenols mainly present in fruits and vegetables. Accumulating evidence supports the potential of flavonoids in attenuating the development of CVD. The ascribed mechanisms of these compounds appear to involve mitigation of inflammation, alteration of the microbiome composition, enhancement of barrier integrity, induction of reverse cholesterol transport, and activation of farnesoid X receptor signaling. In this review, we critically appraise the methods by which the gut microbiome, despite being essential to the human body, predisposes to CVD. Moreover, we dissect the mechanisms and pathways underlying the cardioprotective effects of flavonoids.
Collapse
Affiliation(s)
- Hadi Mansour
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
| | - Maha Khachab
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Beirut, Lebanon
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
40
|
Deshayes S, Ruello P, Simard C, Dupont PA, Bauge C, Abbas A, de Boysson H, Aouba A, Manrique A. 18F-fluorodeoxyglucose PET-MR characterization of aortic inflammation in ApoE -/- mouse models of accelerated atherosclerosis: comparison of Western diet vs. uremia. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2024; 40:2335-2344. [PMID: 39305349 DOI: 10.1007/s10554-024-03238-0] [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: 02/01/2024] [Accepted: 09/06/2024] [Indexed: 11/15/2024]
Abstract
ApoE-/- mice are a widely used preclinical model of atherosclerosis, potentially accelerated by a Western diet (WD) or uremia. We aimed to compare hybrid 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography-magnetic resonance (PET-MR) and immunostaining in ApoE-/- models of accelerated atherosclerosis. Five groups were studied: standard diet-fed ApoE-/- (n = 7), standard diet-fed and uremic ApoE-/- (n = 7), WD ApoE-/- (n = 7), WD and uremic ApoE-/- (n = 6), and control C57BL/6J mice (n = 6). Uremia was induced by electrocoagulation of the right kidney at 8 weeks old, followed 2 weeks later by a contralateral nephrectomy. 18F-FDG PET-MR imaging and histological staining (anti-CD4, -CD8, -CD11c, -CD20, -CD31, -CD68, -CD163, -interferon-γ, interleukin-1α, -1β, -6, -17 A antibodies) were performed in 18-week-old mice, i.e., 8 weeks after 5/6 nephrectomy and/or WD. 18F-FDG uptake was similar in all groups. In contrast, histological staining highlighted higher percentages of CD8-, CD68-, or CD11c-positive cells in ApoE-/- aortic samples than in wild-type aortic samples. In addition, immunostaining revealed some differences between ApoE-/- mouse groups. Only the WD seemed to contribute to these differences. Using immunostaining, WD appeared to be a stronger accelerator of atherosclerosis than uremia. However, 18F-FDG PET-MR imaging failed to demonstrate in vivo increased aortic glucose uptake in these models.
Collapse
Affiliation(s)
- Samuel Deshayes
- Department of Internal Medicine, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, 14000, France
- Normandie Univ, UNICAEN, UR4650 PSIR, CHU de Caen Normandie, Caen, 14000, France
| | - Pauline Ruello
- Normandie Univ, UNICAEN, UR4650 PSIR, CHU de Caen Normandie, Caen, 14000, France
| | - Christophe Simard
- Normandie Univ, UNICAEN, UR4650 PSIR, CHU de Caen Normandie, Caen, 14000, France
| | | | - Caroline Bauge
- Normandie Univ, UNICAEN, UR4650 PSIR, CHU de Caen Normandie, Caen, 14000, France
| | - Ahmed Abbas
- Normandie Univ, UNICAEN, UR4650 PSIR, CHU de Caen Normandie, Caen, 14000, France
| | - Hubert de Boysson
- Department of Internal Medicine, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, 14000, France
- Normandie Univ, UNICAEN, UR4650 PSIR, CHU de Caen Normandie, Caen, 14000, France
| | - Achille Aouba
- Department of Internal Medicine, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, 14000, France
| | - Alain Manrique
- Normandie Univ, UNICAEN, UR4650 PSIR, CHU de Caen Normandie, Caen, 14000, France.
- Department of Nuclear Medicine, Normandie Univ, UNICAEN, CHU de Caen Normandie, Caen, 14000, France.
- UR4650 PSIR, GIP Cyceron, Campus Jules Horowitz, Boulevard Henri Becquerel, BP 5229, Caen, 14074, France.
| |
Collapse
|
41
|
Lee M, Ahn KS, Kim M. Effects of Artemisia asiatica ex on Akkermansia muciniphila dominance for modulation of Alzheimer's disease in mice. PLoS One 2024; 19:e0312670. [PMID: 39466764 PMCID: PMC11516174 DOI: 10.1371/journal.pone.0312670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024] Open
Abstract
The gut microbiome influences neurological disorders through bidirectional communication between the gut and the brain, i.e., the gut-brain axis. Artemisia asiatica ex, an extract of Artemisia asiatica Nakai (Stillen®, DA-9601) has been reported to improve depression by increasing brain-derived neurotropic factor. Therefore, we hypothesized that DA-9601 can be a potential therapeutic candidate for Alzheimer's disease (AD) acting through the gut-brain axis. Four groups of Tg2576 mice were used as the animal model for AD: wild type mice (n = 6), AD mice (n = 6), and DA-9601-administered AD mice given dosages of 30mg/kg/day (DA_30mg; n = 6) or 100mg/kg/day (DA_100mg; n = 6). Microglial activation, blood‒brain barrier integrity, amyloid beta accumulation, cognitive behavior, and changes in the gut microbiome were analyzed. DA-9601 improved the cognitive behavior of mice (DA_30mg **p<0.01; DA_100mg **p<0.01) and reduced amyloid beta accumulation (DA_30mg ***p<0.001; DA_100mg **p<0.01). Increased Iba-1 and upregulation of claudin-5 (DA_30mg *p<0.05) and occludin (DA_30mg **p<0.01; DA_100mg ***p<0.001) indicated altered microglial activation and improved blood‒brain barrier integrity. Akkermansia muciniphila was dramatically increased by DA-9601 administration (DA_30mg 47%; DA_100mg 61%). DA-9601 improved AD pathology with Akkermansia muciniphila dominance in the gut microbiome in a mouse model of AD, inferring that DA-9601 can affect AD through the gut-brain axis.
Collapse
Affiliation(s)
- Mijung Lee
- Department of Neurology, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Kwang-Sung Ahn
- Functional Genome Institute, PDXen. Biosystem Co., Gyeongi-do, South Korea
| | - Manho Kim
- Department of Neurology, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Neuroscience Dementia Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Protein Metabolism Medical Research Center, College of Medicine, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
42
|
Chen Y, Ni B, Yang C, Pan J, Zhang J. Long-term Helicobacter pylori infection is associated with an increased risk of carotid plaque formation: a retrospective cohort study. Front Cardiovasc Med 2024; 11:1476435. [PMID: 39512368 PMCID: PMC11540777 DOI: 10.3389/fcvm.2024.1476435] [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: 08/10/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024] Open
Abstract
Background Cardiovascular disease significantly impacts human health. The development of carotid plaques elevates the risk of cardiovascular disease, while the influence of Helicobacter pylori (H. pylori) on carotid plaques remains a subject of debate. This study aimed to investigate the association between H. pylori infection and carotid plaque using a cohort study. Methods The study included individuals who underwent multiple physical examinations at the Health Examination Center of Taizhou Hospital. The relationship between H. pylori and carotid plaque was explored using multifactorial logistic regression analysis. Participants were categorized into groups based on their H. pylori infection status at the initial and final examinations, comprising persistent infection, persistent negative, new infection, and eradication infection, to analyze variations in carotid plaque prevalence among these groups. Results In both univariate and multifactorial regression analyses, H. pylori was identified as a risk factor for carotid plaque development. Moreover, when compared to the persistent negative group, both the new infection and persistent infection groups showed a notable increase in the risk of carotid plaque. Additionally, individuals in the persistent infection group exhibited higher blood pressure and blood glucose levels than those in the persistent negative group. Likewise, there was a discrepancy in the impact of insulin resistance on carotid plaque between the H. pylori positive and negative groups. Conclusion H. pylori is a risk factor for carotid plaque, with a long-term infection associated with an increased risk of carotid plaque formation. In addition, H. pylori promoting carotid plaque formation may be related to blood pressure, blood glucose, and insulin resistance.
Collapse
Affiliation(s)
- Yi Chen
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Bingqian Ni
- Department of Otolaryngology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Chaoyu Yang
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Jingjing Pan
- Zhejiang Provincial Centers for Disease Control and Prevention, Hangzhou, China
| | - Jinshun Zhang
- Home Ward, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
- Health Management Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| |
Collapse
|
43
|
Song Y, Chen C, Li W. Ginsenoside Rb 1 in cardiovascular and cerebrovascular diseases: A review of therapeutic potentials and molecular mechanisms. CHINESE HERBAL MEDICINES 2024; 16:489-504. [PMID: 39606264 PMCID: PMC11589305 DOI: 10.1016/j.chmed.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 11/29/2024] Open
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs), which are circulatory system diseases caused by heart defects and vascular diseases, are the major noncommunicable diseases affecting global public health. With the improvement of economic level and the change of human lifestyle, the prevalence of CCVDs continues to increase. Ginseng (Panax ginseng C. A. Mey.) was widely used in traditional diseases due to its supposed tonic properties. Ginsenoside Rb1 (G-Rb1) is the most abundant active ingredient with multiple pharmacological effects extracted from ginseng, which has been shown to have potential benefits on the cardiovascular system through a variety of mechanisms, including anti-oxidation, anti-inflammatory, regulation of vasodilation, reduction of platelet adhesion, influence of calcium ion channels, improvement of lipid distribution, involving in glucose metabolism and controlling blood sugar. This review reviewed the protective effects of G-Rb1 on CCVDs and its potential mechanisms, such as atherosclerosis (AS), hypertension, coronary heart disease (CHD), ischemic stroke (IS) and periocular microvascular retinopathy. Finally, we reviewed and reported the results of in vivo and in vitro experiments using G-Rb1 to improve CCVDs, highlighted its efficacy, safety, and limitations.
Collapse
Affiliation(s)
- Yueqin Song
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
- Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Changchun 130118, China
| |
Collapse
|
44
|
Nikiforov NG. Editorial to the Special Issue "Molecular and Cellular Mechanisms of CVD: Focus on Atherosclerosis". Biomedicines 2024; 12:2148. [PMID: 39335661 PMCID: PMC11430762 DOI: 10.3390/biomedicines12092148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
The current Special Issue, "Molecular and Cellular Mechanisms of CVD: Focus on Atherosclerosis", is dedicated to exploring the various mechanisms involved in atherogenesis [...].
Collapse
Affiliation(s)
- Nikita G Nikiforov
- Laboratory of Angiopathology, The Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
45
|
Cruz Neto JPR, de Luna Freire MO, de Albuquerque Lemos DE, Ribeiro Alves RMF, de Farias Cardoso EF, de Moura Balarini C, Duman H, Karav S, de Souza EL, de Brito Alves JL. Targeting Gut Microbiota with Probiotics and Phenolic Compounds in the Treatment of Atherosclerosis: A Comprehensive Review. Foods 2024; 13:2886. [PMID: 39335815 PMCID: PMC11431284 DOI: 10.3390/foods13182886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease. Dysregulated lipid metabolism, oxidative stress, and inflammation are the major mechanisms implicated in the development of AS. In addition, evidence suggests that gut dysbiosis plays an important role in atherogenesis, and modulation of the gut microbiota with probiotics and phenolic compounds has emerged as a promising strategy for preventing and treating AS. It has been shown that probiotics and phenolic compounds can improve atherosclerosis-related parameters by improving lipid profile, oxidative stress, and inflammation. In addition, these compounds may modulate the diversity and composition of the gut microbiota and improve atherosclerosis. The studies evaluated in the present review showed that probiotics and phenolic compounds, when consumed individually, improved atherosclerosis by modulating the gut microbiota in various ways, such as decreasing gut permeability, decreasing TMAO and LPS levels, altering alpha and beta diversity, and increasing fecal bile acid loss. However, no study was found that evaluated the combined use of probiotics and phenolic compounds to improve atherosclerosis. The available literature highlights the synergistic potential between phenolic compounds and probiotics to improve their health-promoting properties and functionalities. This review aims to summarize the available evidence on the individual effects of probiotics and phenolic compounds on AS, while providing insights into the potential benefits of nutraceutical approaches using probiotic strains, quercetin, and resveratrol as potential adjuvant therapies for AS treatment through modulation of the gut microbiota.
Collapse
Affiliation(s)
- José Patrocínio Ribeiro Cruz Neto
- Department of Nutrition, Health Sciences Center, Campus I—Jd. Cidade Universitária, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (J.P.R.C.N.); (M.O.d.L.F.); (D.E.d.A.L.); (E.L.d.S.)
| | - Micaelle Oliveira de Luna Freire
- Department of Nutrition, Health Sciences Center, Campus I—Jd. Cidade Universitária, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (J.P.R.C.N.); (M.O.d.L.F.); (D.E.d.A.L.); (E.L.d.S.)
| | - Deborah Emanuelle de Albuquerque Lemos
- Department of Nutrition, Health Sciences Center, Campus I—Jd. Cidade Universitária, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (J.P.R.C.N.); (M.O.d.L.F.); (D.E.d.A.L.); (E.L.d.S.)
| | - Rayanne Maira Felix Ribeiro Alves
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba, João Pessoa 58037-760, PB, Brazil; (R.M.F.R.A.); (E.F.d.F.C.); (C.d.M.B.)
| | - Emmily Ferreira de Farias Cardoso
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba, João Pessoa 58037-760, PB, Brazil; (R.M.F.R.A.); (E.F.d.F.C.); (C.d.M.B.)
| | - Camille de Moura Balarini
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba, João Pessoa 58037-760, PB, Brazil; (R.M.F.R.A.); (E.F.d.F.C.); (C.d.M.B.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (H.D.); (S.K.)
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (H.D.); (S.K.)
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Campus I—Jd. Cidade Universitária, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (J.P.R.C.N.); (M.O.d.L.F.); (D.E.d.A.L.); (E.L.d.S.)
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Campus I—Jd. Cidade Universitária, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (J.P.R.C.N.); (M.O.d.L.F.); (D.E.d.A.L.); (E.L.d.S.)
| |
Collapse
|
46
|
Etlin S, Rose J, Bielski L, Walter C, Kleinman AS, Mason CE. The human microbiome in space: parallels between Earth-based dysbiosis, implications for long-duration spaceflight, and possible mitigation strategies. Clin Microbiol Rev 2024; 37:e0016322. [PMID: 39136453 PMCID: PMC11391694 DOI: 10.1128/cmr.00163-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
SUMMARYThe human microbiota encompasses the diverse communities of microorganisms that reside in, on, and around various parts of the human body, such as the skin, nasal passages, and gastrointestinal tract. Although research is ongoing, it is well established that the microbiota exert a substantial influence on the body through the production and modification of metabolites and small molecules. Disruptions in the composition of the microbiota-dysbiosis-have also been linked to various negative health outcomes. As humans embark upon longer-duration space missions, it is important to understand how the conditions of space travel impact the microbiota and, consequently, astronaut health. This article will first characterize the main taxa of the human gut microbiota and their associated metabolites, before discussing potential dysbiosis and negative health consequences. It will also detail the microbial changes observed in astronauts during spaceflight, focusing on gut microbiota composition and pathogenic virulence and survival. Analysis will then turn to how astronaut health may be protected from adverse microbial changes via diet, exercise, and antibiotics before concluding with a discussion of the microbiota of spacecraft and microbial culturing methods in space. The implications of this review are critical, particularly with NASA's ongoing implementation of the Moon to Mars Architecture, which will include weeks or months of living in space and new habitats.
Collapse
Affiliation(s)
- Sofia Etlin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
- BioAstra Inc., New York, New York, USA
| | - Julianna Rose
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
- BioAstra Inc., New York, New York, USA
| | - Luca Bielski
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
| | - Claire Walter
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Department of Biology, Cornell University, Ithaca, New York, USA
- BioAstra Inc., New York, New York, USA
| | - Ashley S Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- BioAstra Inc., New York, New York, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
- Tri-Institutional Biology and Medicine program, Weill Cornell Medicine, New York, New York, USA
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
47
|
Senthong V, Kiatchoosakun S, Wongvipaporn C, Phetcharaburanin J, Sritara P, Phrommintikul A. Trimethylamine-N-oxide and 5-year mortality: the role of gut microbiota-generated metabolite from the CORE-Thailand cohort. Sci Rep 2024; 14:21264. [PMID: 39261513 PMCID: PMC11391081 DOI: 10.1038/s41598-024-71479-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024] Open
Abstract
The gut microbiota metabolite trimethylamine-N-oxide (TMAO)-derived from dietary phosphatidylcholine-is mechanistically linked to cardiovascular disease (CVD) and increased cardiovascular risk. This study examined the relationship between fasting plasma TMAO levels and 5-year all-cause mortality in a cohort of patients at high risk of cardiovascular events (CORE-Thailand Registry). Of the 134 patients, 123 (92%) had established cardiovascular disease, and 11 (8%) had multiple risk factors. Fasting plasma TMAO levels were measured using nuclear magnetic resonance spectroscopy. Within this prospective cohort study, the median TMAO was 3.81 μM [interquartile range (IQR) 2.89-5.50 μM], with a mean age of 65 ± 11 years; 61% were men, and 39.6% had type II diabetes. Among 134 patients, 65 (49%) were identified as the high-TMAO group (≥ 3.8 μM), and 69 (51%) were identified as the low-TMAO group (< 3.8 μM). After a median follow-up of 58.8 months, the high-TMAO group was associated with a 2.88-fold increased mortality risk. Following adjustment for traditional risk factors, high-sensitivity cardiac troponin-T, estimated glomerular filtration rate, angiotensin-converting enzyme (ACEI), or angiotensin-receptor blocker (ARB) use, the high-TMAO group remained predictive of 5-year all-cause mortality risk (the high-TMAO vs. the low-TMAO group, adjusted hazard ratio 2.73, 95% CI 1.13-6.54; P = 0.025). Among Thai patients at high risk of cardiovascular events, increased plasma TMAO levels portended greater long-term mortality risk.
Collapse
Affiliation(s)
- Vichai Senthong
- Cardiovascular Unit, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Songsak Kiatchoosakun
- Cardiovascular Unit, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chaiyasith Wongvipaporn
- Cardiovascular Unit, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jutarop Phetcharaburanin
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University National Phenome Center, Khon Kaen University, Khon Kaen, Thailand
| | - Piyamitr Sritara
- Cardiovascular Unit, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Arintaya Phrommintikul
- Cardiovascular Unit, Department of Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50002, Thailand.
| |
Collapse
|
48
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
49
|
Korn LL, Kutyavin VI, Bachtel ND, Medzhitov R. Adverse Food Reactions: Physiological and Ecological Perspectives. Annu Rev Nutr 2024; 44:155-178. [PMID: 38724028 DOI: 10.1146/annurev-nutr-061021-022909] [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] [Indexed: 05/15/2024]
Abstract
While food is essential for survival, it can also cause a variety of harmful effects, ranging from intolerance to specific nutrients to celiac disease and food allergies. In addition to nutrients, foods contain myriads of substances that can have either beneficial or detrimental effects on the animals consuming them. Consequently, all animals evolved defense mechanisms that protect them from harmful food components. These "antitoxin" defenses have some parallels with antimicrobial defenses and operate at a cost to the animal's fitness. These costs outweigh benefits when defense responses are exaggerated or mistargeted, resulting in adverse reactions to foods. Additionally, pathological effects of foods can stem from insufficient defenses, due to unabated toxicity of harmful food components. We discuss the structure of antitoxin defenses and how their failures can lead to a variety of adverse food reactions.
Collapse
Affiliation(s)
- Lisa L Korn
- Department of Medicine, Section of Rheumatology, Allergy, and Clinical Immunology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Vassily I Kutyavin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Nathaniel D Bachtel
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Ruslan Medzhitov
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| |
Collapse
|
50
|
Turkistani A, Al‐Kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Bahaa MM, Al‐Windy S, Batiha GE. Pharmacological characterization of the antidiabetic drug metformin in atherosclerosis inhibition: A comprehensive insight. Immun Inflamm Dis 2024; 12:e1346. [PMID: 39092773 PMCID: PMC11295104 DOI: 10.1002/iid3.1346] [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: 11/16/2023] [Revised: 05/05/2024] [Accepted: 07/06/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a progressive disease that interferes with blood flow, leading to cardiovascular complications such as hypertension, ischemic heart disease, ischemic stroke, and vascular ischemia. The progression of AS is correlated with inflammation, oxidative stress, and endothelial dysfunction. Various signaling pathways, like nuclear erythroid-related factor 2 (Nrf2) and Kruppel-like factor 2 (KLF2), are involved in the pathogenesis of AS. Nrf2 and KLF2 have anti-inflammatory and antioxidant properties. Thus, activation of these pathways may reduce the development of AS. Metformin, an insulin-sensitizing drug used in the management of type 2 diabetes mellitus (T2DM), increases the expression of Nrf2 and KLF2. AS is a common long-term macrovascular complication of T2DM. Thus, metformin, through its pleiotropic anti-inflammatory effect, may attenuate the development and progression of AS. AIMS Therefore, this review aims to investigate the possible role of metformin in AS concerning its effect on Nrf2 and KLF2 and inhibition of reactive oxygen species (ROS) formation. In addition to its antidiabetic effect, metformin can reduce cardiovascular morbidities and mortalities compared to other antidiabetic agents, even with similar blood glucose control by the Nrf2/KLF2 pathway activation. CONCLUSION In conclusion, metformin is an effective therapeutic strategy against the development and progression of AS, mainly through activation of the KLF2/Nrf2 axis.
Collapse
Affiliation(s)
- Areej Turkistani
- Department of Pharmacology and Toxicology, College of MedicineTaif UniversityTaifSaudi Arabia
| | - Haydar M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
- Department of Clinical Pharmacology and MedicineJabir ibn Hayyan Medical UniversityKufaIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
- Department of Research & DevelopmentFunogenAthensGreece
- University Centre for Research & DevelopmentChandigarh UniversityPunjabIndia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Mostafa M. Bahaa
- Pharmacy Practice Department, Faculty of PharmacyHorus UniversityNew DamiettaEgypt
| | - Salah Al‐Windy
- Department of Biology, College of ScienceBaghdad UniversityBaghdadIraq
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|