1
|
Chen X, Yan L, Yang J, Xu C, Yang L. The impact of probiotics on oxidative stress and inflammatory markers in patients with diabetes: a meta-research of meta-analysis studies. Front Nutr 2025; 12:1552358. [PMID: 40123937 PMCID: PMC11926743 DOI: 10.3389/fnut.2025.1552358] [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: 12/27/2024] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
Objective Probiotic supplementation has gained attention for its potential to modulate inflammatory and oxidative stress biomarkers, particularly in metabolic disorders. This meta-analysis evaluates the effects of probiotics on C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), malondialdehyde (MDA), total antioxidant capacity (TAC), glutathione (GSH), and nitric oxide (NO) in patients with diabetes. Methods A Meta-Research was conducted on 15 meta-analyses of unique 33 randomized controlled trials (RCTs) published between 2015 and 2022, involving 26 to 136 participants aged 26 to 66 years. Data were synthesized using standardized mean differences (SMD), with sensitivity analysis using a random-effect model. Results Probiotic supplementation significantly reduced CRP (SMD = -0.79, 95% CI: -1.19, -0.38), TNF-α (SMD = -1.35, 95% CI: -2.05, -0.66), and MDA levels (WMD: -0.82, 95% CI: -1.16, -0.47). Probiotics increased GSH (SMD = 1.00, 95% CI: 0.41, 1.59), TAC (SMD = 0.48, 95% CI: 0.27, 0.69), and NO (SMD = 0.60, 95% CI: 0.30, 0.91). Result on IL-6 was not significant (SMD = -0.29, 95% CI: -0.66, 0.09). Sensitivity analyses confirmed robustness. Conclusion Probiotics significantly improved inflammatory and oxidative stress biomarkers in patients with diabetes, with variations influenced by population and dosage. Future studies should explore novel probiotic strains and longer interventions.
Collapse
Affiliation(s)
- Xi Chen
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Lijun Yan
- Department of Geriatric Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jie Yang
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Chenlong Xu
- Department of Laboratory Medicine, Ningbo Yinzhou No.2 Hospital, Ningbo, China
| | - Lv Yang
- Department of Laboratory Medicine, Ningbo Yinzhou No.2 Hospital, Ningbo, China
| |
Collapse
|
2
|
Ghorbani Z, Dashti F, Grafenauer S, Arami S, Mahdavi-Roshan M, Salari A. Ultra-processed foods and coronary artery disease severity: a cross-sectional study of at-risk normal-weight and overweight patients undergoing elective angiography. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:63. [PMID: 40045397 PMCID: PMC11883926 DOI: 10.1186/s41043-025-00796-4] [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] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/16/2025] [Indexed: 03/09/2025]
Abstract
INTRODUCTION There is growing interest in the connection between ultra-processed food (UPF) and cardiovascular diseases. This study explores how UPF intake relates to the severity of coronary artery disease (CAD) in at-risk patients undergoing elective angiography. METHODS Data covering demographic, and clinical details, and dietary intakes (using a validated food frequency questionnaire) were gathered from the Nutrition Heshmat Registry (NUTHER) in Rasht, Iran. UPF consumption was evaluated using the NOVA food classification system, with the exception of core grain foods. The study comprised 1,015 participants, who were classified based on the severity of CAD using the Gensini score (severe-CAD = Gensini score ≥ 60). Logistic regression was used to analyze the odd ratio (OR) and 95%confidence interval (95%CI) for severe-CAD across UPF quartiles (percentage of energy), and for each 10% increase in UPF intake. Restricted cubic spline (RCS) regression was employed to explore nonlinear relationships between UPF and severe-CAD. RESULTS Following controlling for potential confounders, normal-weight participants in the highest quartile of UPF exhibited about 5 times greater odds of severe-CAD than those in the lowest category (OR(95%CI): 5.01 (1.89, 13.29); P-for-trend = 0.002). Overweight/obese participants in the higher UPF quartiles had approximately 2-3.5 times greater odds for severe-CAD than those in the 1st quartile (ORs (95%CIs): 3rd quartile 1.91 (1.14, 3.21); and 4th quartile: 3.53 (2.07, 5.99); P-for-trend < 0.001). Each 10% increase in daily energy intake from UPF was associated with about 1.6-2 times increased severe-CAD risk among overweight/obese and normal-weight individuals (ORs (95%CIs) of 1.64 (1.28, 2.11), and 2.24 (1.24, 4.05), respectively). RCS analysis showed an upward trend toward higher UPF intake in relation to increased risk of severe-CAD (P-for-overall-trend < 0.0001; P-for-nonlinearity = 0.005). CONCLUSION The findings obtained underscore a direct association between UPF and the risk of CAD progression among at-risk patients, independent of BMI. However, further prospective studies are essential to confirm these results and better understand this relationship.
Collapse
Affiliation(s)
- Zeinab Ghorbani
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Dashti
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Grafenauer
- School of Health Science, Faculty of Medicine and Health, University of New South Wales, Randwick, Australia
| | - Samira Arami
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Marjan Mahdavi-Roshan
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
3
|
Liu Y, Bai Z, Yan R, Ma J, Wang L, Li Y, Liu Y, Ma H, Wang T, Yang L, Liu J, Shen W, Zhang X, Jia S, Wang H. Lactobacillus rhamnosus GG ameliorates atherosclerosis via suppression of oxidative stress and inflammation by reshaping the gut microbiota. Biochem Biophys Res Commun 2025; 751:151417. [PMID: 39929060 DOI: 10.1016/j.bbrc.2025.151417] [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: 09/07/2024] [Revised: 01/09/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE With growing awareness of probiotics' benefits, more studies are exploring their efficacy and mechanisms in reducing atherosclerosis (AS). This study aimed to investigate the potential therapeutic effects of Lactobacillus rhamnosus GG (LGG) on atherosclerotic mice and underlying mechanisms. DESIGN ApoE-/- mice were gavaged with a dose of 2 × 109 CFU LGG per mouse once daily, while both ApoE-/- and C57BL/6J mice received normal saline as controls. After 15 weeks, en face Oil Red O staining and aortic sinus morphometry were used to assess the effects of LGG intervention on AS. The expression of the Nrf2/HO-1 pathway, along with oxidative stress and inflammation, was measured in the aortic sinus, aortas, or plasma. Immune cells were analyzed by flow cytometry. 16S rRNA gene sequencing analysis evaluated structural changes in the intestinal microbiota. RESULTS LGG-treated ApoE-/- mice showed a significant reduction of AS progression by suppressing oxidative stress and inflammation. Mechanistically, LGG intervention significantly increased the levels of Nrf2/HO-1 in the aortic sinus of ApoE-/- mice. Moreover, decreased aortic macrophages and elevated blood regulatory T cells (Tregs) were found with LGG intervention in the murine AS model. Moreover, compared to C57BL/6J mice, ApoE-/- mice exhibited disrupted intestinal flora. Nonetheless, LGG intervention restored their intestinal flora to a composition resembling that of C57BL/6J mice, thereby increasing the abundance of beneficial bacteria. CONCLUSION LGG significantly attenuates AS by reducing oxidative stress and inflammation probably via activating the Nrf2/HO-1 pathway. Remarkably, LGG modulates gut microbiota, further enhancing its protective efficacy against AS.
Collapse
Affiliation(s)
- Yajuan Liu
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Zhixia Bai
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ru Yan
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Junbai Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Liting Wang
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yiwei Li
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yuanyuan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Huiyan Ma
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ting Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Libo Yang
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jian Liu
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Wenke Shen
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Shaobin Jia
- General Hospital of Ningxia Medical University, the First Clinical Medical College of Ningxia Medical University, Yinchuan, 750004, Ningxia, China; Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China; National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Yinchuan, 750004, Ningxia, China
| | - Hao Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
4
|
Huang P, Xiao Y, He Y. The causal relationships between gut microbiota and venous thromboembolism: a Mendelian randomization study. Hereditas 2025; 162:25. [PMID: 39980076 PMCID: PMC11841150 DOI: 10.1186/s41065-025-00389-5] [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: 12/20/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Venous thromboembolism (VTE) is still one of the most severe health issues, increasing mortality and lengthening hospital stays. Different abundances of gut microbiota have been clinically linked to VTE and coagulopathy. However, whether gut microbiota affected VTE formation remained uncertain. METHODS The causative links between VTE and 211 gut microbiota at phylum, class, order, family and genus level were separately investigated using two-sample Mendelian Randomization (MR) analysis. Firstly, single nucleotide polymorphisms (SNPs) locus-wide significantly (P < 1.0 × 10- 5) related with gut microbiome abundance were extracted from large genome-wide analysis (GWAS) meta-analysis summary data. Instrumental variables (IVs) without pleiotropy were selected using the PhenoScanner and MR PRESSO test. Then, the MR analysis was implemented using the inverse variance weighted (IVW) method. Moreover, weighted median method, MR Egger method, simple median method and MR PRESSO were conducted to validate the causal associations. The reliability of the results was also assessed utilizing various sensitivity analyses, reverse MR analysis and multivariate Mendelian Randomization analysis (MVMR). RESULTS We found the phylum Firmicutes was robustly protective against VTE with MR analysis. Moreover, five taxa of Actinobacteria phylum (Bifidobacteriales order, Actinomycetales order, Bifidobacteriaceae family, Actinomycetaceae family, Slackia genus) and two taxa of Firmicutes phylum (Bacillales order, Lachnospiraceae UCG-010 genus) were suggestively protective for VTE. While three taxa of Firmicutes phylum (Bacilli class, Lactobacillales order and Lactococcus genus) might suggestively increase the risk of VTE. Sensitivity analyses indicated no significant horizontal pleiotropy, heterogeneity, or reverse causal associations. Furthermore, MVMR analysis unveiled independently positive causal association of Firmicutes phylum and Lachnospiraceae UCG-010 genus with risk of VTE. CONCLUSION Two taxa of gut microbes (Firmicutes phylum and Lachnospiraceae UCG-010 genus) were independently protective against VTE, which suggests a potential avenue for developing new cost-effective strategies with minor side effects for VTE prevention and treatment.
Collapse
Affiliation(s)
- Pin Huang
- Department of Intensive Care Unit (ICU), Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ying Xiao
- Department of Intensive Care Unit (ICU), Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Ye He
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China.
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
5
|
de la Fuente-Muñoz M, Román-Carmena M, Amor S, González-Hedström D, Martinez-Rios V, Martorell P, Inarejos-García AM, García Bou R, Guilera-Bermell S, García-Villalón ÁL, Granado M. Supplementation with the Postbiotic BPL1™-HT (Heat-Inactivated Bifidobacterium animalis subsp. Lactis) Attenuates the Cardiovascular Alterations Induced by Angiotensin II Infusion in Mice. Antioxidants (Basel) 2025; 14:193. [PMID: 40002381 PMCID: PMC11851978 DOI: 10.3390/antiox14020193] [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: 01/10/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Hypertension is associated with alterations in the composition and diversity of the intestinal microbiota. Indeed, supplementation with probiotics and prebiotics has shown promising results in modulating the gut microbiota and improving cardiovascular health. However, there are no studies regarding the possible beneficial effects of postbiotics on cardiovascular function and particularly on hypertension-induced cardiovascular alterations. Thus, the aim of this study was to analyze the effect of supplementation with the heat-treated Bifidobacterium animalis subsp. lactis CECT 8145 strain (BPL1™ HT), a postbiotic developed by the company ADM-Biopolis, on cardiovascular alterations induced by angiotensin II (AngII) infusion in mice. For this purpose, three groups of C57BL/6J male mice were used: (i) mice infused with saline (control); (ii) mice infused with AngII for 4 weeks (AngII); and (iii) mice supplemented with BPL1™ HT in the drinking water (1010 cells/animal/day) for 8 weeks and infused with AngII for the last 4 weeks (AngII + BPL1™ HT). AngII infusion was associated with heart hypertrophy, hypertension, endothelial dysfunction, and overexpression of proinflammatory cytokines in aortic tissue. BPL1™ HT supplementation reduced systolic blood pressure and attenuated AngII-induced endothelial dysfunction in aortic segments. Moreover, mice supplemented with BPL1™ HT showed a decreased gene expression of the proinflammatory cytokine interleukin 6 (Il-6) and the prooxidant enzymes NADPH oxidases 1 (Nox-1) and 4 (Nox-4), as well as an overexpression of AngII receptor 2 (At2r) and interleukin 10 (Il-10) in arterial tissue. In the heart, BPL1™ HT supplementation increased myocardial contractility and prevented ischemia-reperfusion-induced cardiomyocyte apoptosis. In conclusion, supplementation with the postbiotic BPL1™ HT prevents endothelial dysfunction, lowers blood pressure, and has cardioprotective effects in an experimental model of hypertension induced by AngII infusion in mice.
Collapse
Affiliation(s)
- Mario de la Fuente-Muñoz
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.d.l.F.-M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Marta Román-Carmena
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.d.l.F.-M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Sara Amor
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.d.l.F.-M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Daniel González-Hedström
- R&D Department of Functional Extracts, ADM Valencia, 46740 Carcaixent, Spain; (D.G.-H.); (V.M.-R.); (A.M.I.-G.); (R.G.B.); (S.G.-B.)
| | - Verónica Martinez-Rios
- R&D Department of Functional Extracts, ADM Valencia, 46740 Carcaixent, Spain; (D.G.-H.); (V.M.-R.); (A.M.I.-G.); (R.G.B.); (S.G.-B.)
| | - Patricia Martorell
- Nutrition Archer Daniels Midland (ADM) Health & Wellness, Biopolis S. L. Parc Cientific, Universitat de València, 46980 Paterna, Spain;
| | - Antonio M. Inarejos-García
- R&D Department of Functional Extracts, ADM Valencia, 46740 Carcaixent, Spain; (D.G.-H.); (V.M.-R.); (A.M.I.-G.); (R.G.B.); (S.G.-B.)
| | - Reme García Bou
- R&D Department of Functional Extracts, ADM Valencia, 46740 Carcaixent, Spain; (D.G.-H.); (V.M.-R.); (A.M.I.-G.); (R.G.B.); (S.G.-B.)
| | - Sonia Guilera-Bermell
- R&D Department of Functional Extracts, ADM Valencia, 46740 Carcaixent, Spain; (D.G.-H.); (V.M.-R.); (A.M.I.-G.); (R.G.B.); (S.G.-B.)
| | - Ángel L. García-Villalón
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.d.l.F.-M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
| | - Miriam Granado
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.d.l.F.-M.); (M.R.-C.); (S.A.); (Á.L.G.-V.)
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
6
|
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
|
7
|
Jeon H, Lee D, Kim JY, Shim JJ, Lee JH. Limosilactobacillus reuteri HY7503 and Its Cellular Proteins Alleviate Endothelial Dysfunction by Increasing Nitric Oxide Production and Regulating Cell Adhesion Molecule Levels. Int J Mol Sci 2024; 25:11326. [PMID: 39457107 PMCID: PMC11509054 DOI: 10.3390/ijms252011326] [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: 10/02/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Endothelial dysfunction, which is marked by a reduction in nitric oxide (NO) production or an imbalance in relaxing and contracting factor levels, exacerbates atherosclerosis by promoting the production of cell adhesion molecules and cytokines. This study aimed to investigate the effects of Limosilactobacillus reuteri HY7503, a novel probiotic isolated from raw milk, on endothelial dysfunction. Five lactic acid bacterial strains were screened for their antioxidant, anti-inflammatory, and endothelium-protective properties; L. reuteri HY7503 had the most potent effect. In a mouse model of angiotensin II-induced endothelial dysfunction, L. reuteri HY7503 reduced vascular thickening (19.78%), increased serum NO levels (226.70%), upregulated endothelial NO synthase (eNOS) expression in the aortic tissue, and decreased levels of cell adhesion molecules (intercellular adhesion molecule-1 [ICAM-1] and vascular cell adhesion molecule-1 [VCAM-1]) and serum cytokines (tumor necrosis factor-alpha [TNF-α] and interleukin-6 [IL-6]). In TNF-α-treated human umbilical vein endothelial cells (HUVECs), L. reuteri HY7503 enhanced NO production and reduced cell adhesion molecule levels. In HUVECs, surface-layer proteins (SLPs) were more effective than extracellular vesicles (exosomes) in increasing NO production and decreasing cell adhesion molecule levels. These findings suggested that L. reuteri HY7503 may serve as a functional probiotic that alleviates endothelial dysfunction.
Collapse
Affiliation(s)
| | | | - Joo-Yun Kim
- R&BD Center, Hy Co., Ltd., 22 Giheungdanji-ro 24 Beon-gil, Giheung-gu, Yongin-si 17086, Gyeonggi-do, Republic of Korea; (H.J.); (D.L.); (J.-J.S.); (J.-H.L.)
| | | | | |
Collapse
|
8
|
Chen YC, Hsu PY, Su MC, Chen YL, Chang YT, Chin CH, Lin IC, Chen YM, Wang TY, Lin YY, Lee CP, Lin MC, Hsiao CC. Long non-coding RNA FKSG29 regulates oxidative stress and endothelial dysfunction in obstructive sleep apnea. Mol Cell Biochem 2024; 479:2723-2740. [PMID: 37914826 DOI: 10.1007/s11010-023-04880-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/07/2023] [Indexed: 11/03/2023]
Abstract
Altered expressions of pro-/anti-oxidant genes are known to regulate the pathophysiology of obstructive sleep apnea (OSA).We aim to explore the role of a novel long non-coding (lnc) RNA FKSG29 in the development of intermittent hypoxia with re-oxygenation (IHR)-induced endothelial dysfunction in OSA. Gene expression levels of key pro-/anti-oxidant genes, vasoactive genes, and the FKSG29 were measured in peripheral blood mononuclear cells from 12 subjects with primary snoring (PS) and 36 OSA patients. Human monocytic THP-1 cells and human umbilical vein endothelial cells (HUVEC) were used for gene knockout and double luciferase under IHR exposure. Gene expression levels of the FKSG29 lncRNA, NOX2, NOX5, and VEGFA genes were increased in OSA patients versus PS subjects, while SOD2 and VEGFB gene expressions were decreased. Subgroup analysis showed that gene expression of the miR-23a-3p, an endogenous competitive microRNA of the FKSG29, was decreased in sleep-disordered breathing patients with hypertension versus those without hypertension. In vitro IHR experiments showed that knock-down of the FKSG29 reversed IHR-induced ROS overt production, early apoptosis, up-regulations of the HIF1A/HIF2A/NOX2/NOX4/NOX5/VEGFA/VEGFB genes, and down-regulations of the VEGFB/SOD2 genes, while the protective effects of FKSG29 knock-down were abolished by miR-23a-3p knock-down. Dual-luciferase reporter assays confirmed that FKSG29 was a sponge of miR-23a-3p, which regulated IL6R directly. Immunofluorescence stain further demonstrated that FKSGH29 knock-down decreased IHR-induced uptake of oxidized low density lipoprotein and reversed IHR-induced IL6R/STAT3/GATA6/ICAM1/VCAM1 up-regulations. The findings indicate that the combined RNA interference may be a novel therapy for OSA-related endothelial dysfunction via regulating pro-/anti-oxidant imbalance or targeting miR-23a-IL6R-ICAM1/VCAM1 signaling.
Collapse
Affiliation(s)
- Yung-Che Chen
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan.
- Sleep Center, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan.
- Department of Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan.
| | - Po-Yuan Hsu
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Mao-Chang Su
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
- Sleep Center, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
- Chang Gung University of Science and Technology, Chia-Yi, Taiwan
| | - Yung-Lung Chen
- Division of Cardiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Ya-Ting Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Chien-Hung Chin
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
- Sleep Center, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - I-Chun Lin
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Yu-Mu Chen
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Ting-Ya Wang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Yong-Yong Lin
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Chiu-Ping Lee
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan.
- Sleep Center, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan.
| | - Chang-Chun Hsiao
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan.
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Kaohsiung City, Taiwan.
| |
Collapse
|
9
|
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
|
10
|
Hassan A, Luqman A, Zhang K, Ullah M, Din AU, Xiaoling L, Wang G. Impact of Probiotic Lactiplantibacillus plantarum ATCC 14917 on atherosclerotic plaque and its mechanism. World J Microbiol Biotechnol 2024; 40:198. [PMID: 38727952 DOI: 10.1007/s11274-024-04010-1] [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: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 06/22/2024]
Abstract
Atherosclerosis is viewed as not just as a problem of lipid build-up in blood vessels, but also as a chronic inflammatory disease involving both innate and acquired immunity. In atherosclerosis, the inflammation of the arterial walls is the key characteristic that significantly contributes to both the instability of plaque and the occlusion of arteries by blood clots. These events ultimately lead to stroke and acute coronary syndrome. Probiotics are living microorganisms that, when consumed in the right quantities, offer advantages for one's health. The primary objective of this study was to investigate the influence of Lactiplantibacillus plantarum ATCC 14917 (ATCC 14917) on the development of atherosclerotic plaques and its underlying mechanism in Apo lipoprotein E-knockout (Apoe-/- mice). In this study, Apoe-/- mice at approximately 8 weeks of age were randomly assigned to three groups: a Normal group that received a normal chow diet, a high fat diet group that received a gavage of PBS, and a Lactiplantibacillus plantarum ATCC 14917 group that received a high fat diet and a gavage of 0.2 ml ATCC 14917 (2 × 109 CFU/mL) per day for a duration of 12 weeks. Our strain effectively reduced the size of plaques in Apoe-/- mice by regulating the expression of inflammatory markers, immune cell markers, chemokines/chemokine receptors, and tight junction proteins (TJPs). Specifically, it decreased the levels of inflammatory markers (ICAM-1, CD-60 MCP-1, F4/80, ICAM-1, and VCAM-1) in the thoracic aorta, (Ccr7, cd11c, cd4, cd80, IL-1β, TNF-α) in the colon, and increased the activity of ROS-scavenging enzymes (SOD-1 and SOD-2). It also influenced the expression of TJPs (occludin, ZO-1, claudin-3, and MUC-3). In addition, the treatment of ATCC 14917 significantly reduced the level of lipopolysaccharide in the mesenteric adipose tissue. The findings of our study demonstrated that our strain effectively decreased the size of atherosclerotic plaques by modulating inflammation, oxidative stress, intestinal integrity, and intestinal immunity.
Collapse
Affiliation(s)
- Adil Hassan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China
| | - Ameer Luqman
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Mehtab Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Ahmad Ud Din
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Liao Xiaoling
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- JinFeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
11
|
Khan S, Ahmad F, Khalid N. Applications of Strain-Specific Probiotics in the Management of Cardiovascular Diseases: A Systemic Review. Mol Nutr Food Res 2024; 68:e2300675. [PMID: 38549453 DOI: 10.1002/mnfr.202300675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/14/2024] [Indexed: 05/08/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of global mortality and novel approaches for prevention and management are needed. The human gastrointestinal tract hosts a diverse microbiota that is crucial in maintaining metabolic homeostasis. The formulation of effective probiotics, alone or in combination, has been under discussion due to their impact on cardiovascular and metabolic diseases. Probiotics have been shown to impact cardiovascular health positively. An imbalance in the presence of Firmicutes and Bacteroidetes has been linked to the progression of CVDs due to their impact on bile acid and cholesterol metabolism. The probiotics primarily help in the reduction of plasma low-density lipoprotein levels and attenuation of the proinflammatory markers. These beneficial microorganisms contribute to lowering cholesterol levels and produce essential short-chain fatty acids. The impact of lipid-regulating probiotic strains on human health is quite significant. However, only a few have been tested for potential beneficial efficacy, and ambiguity exists regarding strain dosages, interactions with confounding factors, and potential adverse effects. Hence, more comprehensive studies and randomized trials are needed to understand the mechanisms of probiotics on CVDs and to ensure human health. This review assesses the evidence and highlights the roles of strain-specific probiotics in the management of CVDs.
Collapse
Affiliation(s)
- Saleha Khan
- Department of Human Nutrition and Dietetics, School of Food and Agricultural Sciences, University of Management and Technology, Lahore, 54000, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Nauman Khalid
- Department of Human Nutrition and Dietetics, School of Food and Agricultural Sciences, University of Management and Technology, Lahore, 54000, Pakistan
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, 59911, United Arab Emirates
| |
Collapse
|
12
|
Liu X, Chen X, Wang C, Song J, Xu J, Gao Z, Huang Y, Suo H. Mechanisms of probiotic modulation of ovarian sex hormone production and metabolism: a review. Food Funct 2024; 15:2860-2878. [PMID: 38433710 DOI: 10.1039/d3fo04345b] [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: 03/05/2024]
Abstract
Sex hormones play a pivotal role in the growth and development of the skeletal, neurological, and reproductive systems. In women, the dysregulation of sex hormones can result in various health complications such as acne, hirsutism, and irregular menstruation. One of the most prevalent diseases associated with excess androgens is polycystic ovary syndrome with a hyperandrogenic phenotype. Probiotics have shown the potential to enhance the secretion of ovarian sex hormones. However, the underlying mechanism of action remains unclear. Furthermore, comprehensive reviews detailing how probiotics modulate ovarian sex hormones are scarce. This review seeks to shed light on the potential mechanisms through which probiotics influence the production of ovarian sex hormones. The role of probiotics across various biological axes, including the gut-ovarian, gut-brain-ovarian, gut-liver-ovarian, gut-pancreas-ovarian, and gut-fat-ovarian axes, with a focus on the direct impact of probiotics on the ovaries via the gut and their effects on brain gonadotropins is discussed. It is also proposed herein that probiotics can significantly influence the onset, progression, and complications of ovarian sex hormone abnormalities. In addition, this review provides a theoretical basis for the therapeutic application of probiotics in managing sex hormone-related health conditions.
Collapse
Affiliation(s)
- Xiao Liu
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Xiaoyong Chen
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| | - Jiahui Xu
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Zhen Gao
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
| | - Yechuan Huang
- College of Bioengineering, Jingchu University of Technology, Jingmen 448000, P. R. China.
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, P. R. China.
- Citrus Research Institute, National Citrus Engineering Research Center, Southwest University, Chongqing 400715, P. R. China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, P. R. China
| |
Collapse
|
13
|
Tang J, Wei Y, Pi C, Zheng W, Zuo Y, Shi P, Chen J, Xiong L, Chen T, Liu H, Zhao Q, Yin S, Ren W, Cao P, Zeng N, Zhao L. The therapeutic value of bifidobacteria in cardiovascular disease. NPJ Biofilms Microbiomes 2023; 9:82. [PMID: 37903770 PMCID: PMC10616273 DOI: 10.1038/s41522-023-00448-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
There has been an increase in cardiovascular morbidity and mortality over the past few decades, making cardiovascular disease (CVD) the leading cause of death worldwide. However, the pathogenesis of CVD is multi-factorial, complex, and not fully understood. The gut microbiome has long been recognized to play a critical role in maintaining the physiological and metabolic health of the host. Recent scientific advances have provided evidence that alterations in the gut microbiome and its metabolites have a profound influence on the development and progression of CVD. Among the trillions of microorganisms in the gut, bifidobacteria, which, interestingly, were found through the literature to play a key role not only in regulating gut microbiota function and metabolism, but also in reducing classical risk factors for CVD (e.g., obesity, hyperlipidemia, diabetes) by suppressing oxidative stress, improving immunomodulation, and correcting lipid, glucose, and cholesterol metabolism. This review explores the direct and indirect effects of bifidobacteria on the development of CVD and highlights its potential therapeutic value in hypertension, atherosclerosis, myocardial infarction, and heart failure. By describing the key role of Bifidobacterium in the link between gut microbiology and CVD, we aim to provide a theoretical basis for improving the subsequent clinical applications of Bifidobacterium and for the development of Bifidobacterium nutritional products.
Collapse
Affiliation(s)
- Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Shi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Qianjiao Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Cao
- The Affiliated Hospital of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, P.R. China.
| | - Nan Zeng
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| |
Collapse
|
14
|
Ghorbani Z, Mirmohammadali SN, Shoaibinobarian N, Rosenkranz SK, Arami S, Hekmatdoost A, Mahdavi-Roshan M. Insulin resistance surrogate markers and risk of hyperuricemia among patients with and without coronary artery disease: a cross-sectional study. Front Nutr 2023; 10:1048675. [PMID: 37671195 PMCID: PMC10475726 DOI: 10.3389/fnut.2023.1048675] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
Background Although emerging evidence emphasizes the associations between both insulin resistance and hyperuricemia with coronary artery disease (CAD) risk, no definite relationship has yet been established. In this respect, time-efficient and affordable methods to estimate insulin resistance (IR) status, and to predict risk of hyperuricemia, are needed. Thus, the goal of this investigation was to examine the associations between IR, as assessed by novel surrogate markers [triglyceride-glucose (TyG) and TyG-body mass index (TyG-BMI)], and risk of hyperuricemia in patients with and without diagnosed CAD. Methods This cross-sectional study used data from the medical records of 1,170 patients who were referred to the cardiology outpatient clinic. Medical records, anthropometrics, and serum analytes were determined at the initial visit. Hyperuricemia was defined as serum uric acid ≥ 5.6 mg/dL. IR was estimated through surrogate markers (TyG and TyG-BMI). Multiple regression analysis was performed to assess the relationship between these indices and odds of hyperuricemia among patients with and without CAD. Results Overall, 814 angiographically-confirmed CAD cases (mean age (SD) = 52 (8)yrs) were compared with 356 patients without CAD (mean age (SD) = 48 (8)yr). There were positive associations between TyG and TyG-BMI indices and odds of hyperuricemia in CAD patients after controlling for confounders (adjusted odds ratio (aOR) = 1.60; 95%CI: 1.02-2.51; p-value = 0.036; and aOR = 1.83; 95%CI: 1.24-2.70; p-value = 0.002, third tertiles for TYG and TYG-BMI, respectively). Conclusion The present findings suggest that higher levels of the IR surrogate markers, TyG and TyG-BMI, are associated with higher odds of hyperuricemia in patients with CAD. However, given the cross-sectional design of this study, the sensitivity and specificity of these novel markers could not be determined for confirming the diagnosis of IR and hyperuricemia, further studies are needed to determine such outcomes and to confirm the current findings.
Collapse
Affiliation(s)
- Zeinab Ghorbani
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Nargeskhatoon Shoaibinobarian
- Department of Nutrition, School of Medical Sciences and Technologies, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sara K. Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Samira Arami
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Azita Hekmatdoost
- Department of Nutrition Research, National Nutrition and Food Technology Research Institute, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Mahdavi-Roshan
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
15
|
Chen Z, Liang W, Liang J, Dou J, Guo F, Zhang D, Xu Z, Wang T. Probiotics: functional food ingredients with the potential to reduce hypertension. Front Cell Infect Microbiol 2023; 13:1220877. [PMID: 37465757 PMCID: PMC10351019 DOI: 10.3389/fcimb.2023.1220877] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/15/2023] [Indexed: 07/20/2023] Open
Abstract
Hypertension is an increasingly pressing public health concern across the globe. It can be triggered by a variety of factors such as age and diet, as well as the stress of modern life. The traditional treatment of hypertension includes calcium ion blockers, angiotensin II receptor inhibitors and β-receptor blockers, but these drugs have at least some side effects. Recent studies have revealed that intestinal flora plays a vital role in maintaining and promoting human health. This is due to the type and amount of probiotics present in the flora. Probiotics can reduce hypertension symptoms through four mechanisms: regulating vascular oxidative stress, producing short-chain fatty acids, restoring endothelial cell function, and reducing inflammation. It has been reported that certain functional foods, using probiotics as their raw material, can modify the composition of intestinal flora, thus regulating hypertension symptoms. Consequently, utilizing the probiotic function of probiotics in conjunction with the properties of functional foods to treat hypertension is a novel, side-effect-free treatment method. This study seeks to summarize the various factors that contribute to hypertension, the mechanism of probiotics in mitigating hypertension, and the fermented functional foods with probiotic strains, in order to provide a basis for the development of functional foods which utilize probiotics as their raw material and may have the potential to reduce hypertension.
Collapse
Affiliation(s)
- Zouquan Chen
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Wanjie Liang
- Research and Development Department(R&D), Shandong Ande Healthcare Apparatus Co., Ltd., Zibo, China
| | - Jie Liang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Jiaxin Dou
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Fangyu Guo
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Daolei Zhang
- School of Bioengineering, Shandong Polytechnic, Jinan, China
- Henan Province Key Laboratory of Water Pollution Control and Rehabilitation Technology, Henan University of Urban Construction, Pingdingshan, China
| | - Zhenshang Xu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| | - Ting Wang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Science, Jinan, China
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Science, Jinan, China
| |
Collapse
|
16
|
Shi Y, Jiang M, Zhang Y, Diao Y, Li N, Liu W, Qiu Z, Qiu Y, Jia A. Hyperoside Nanomicelles Alleviate Atherosclerosis by Modulating the Lipid Profile and Intestinal Flora Structure in High-Fat-Diet-Fed Apolipoprotein-E-Deficient Mice. Molecules 2023; 28:5088. [PMID: 37446750 DOI: 10.3390/molecules28135088] [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: 05/12/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Atherosclerosis (AS) is a serious threat to human health and the main pathological basis of cardiovascular disease. Hyperoside (Hyp), a flavonoid found mainly in traditional Chinese herbs, can exert antitumor, anti-inflammatory, antioxidant, and cardiovascular-protective effects. Herein, we prepared hybrid nanomicelles (HFT) comprising Hyp loaded into pluronic F-127 and polyethylene glycol 1000 vitamin E succinate and assessed their effects on AS. To establish an AS model, apolipoprotein-E-deficient (ApoE-/-) mice were fed a high-fat diet. We then analyzed the effects of HFT on AS-induced changes in aortic tissues and metabolic markers, simultaneously assessing changes in gut flora community structure. In mice with AS, HFT significantly reduced the aortic plaque area; decreased levels of total cholesterol, triglyceride, low-density lipoprotein cholesterol, inflammatory factors, and inducible nitric oxide synthase (NOS); increased high-density lipoprotein cholesterol, endothelial NOS, superoxide dismutase, catalase, and glutathione levels; and promoted the proliferation of beneficial gut bacteria. HFT could regulate intestinal flora structure and lipid metabolism and inhibit inflammatory responses. These beneficial effects may be mediated by inhibiting nuclear factor kappa B signal activation, reducing inflammatory factor expression and improving gut microflora structure and dyslipidemia. The present study provides an empirical basis for the development and clinical application of new dosage forms of Hyp.
Collapse
Affiliation(s)
- Yuwen Shi
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Mengcheng Jiang
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yuhang Zhang
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yuanyuan Diao
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Na Li
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Weipeng Liu
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Zhidong Qiu
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Ye Qiu
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Ailing Jia
- Pharmacy College, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
17
|
Banerjee A, Somasundaram I, Das D, Jain Manoj S, Banu H, Mitta Suresh P, Paul S, Bisgin A, Zhang H, Sun XF, Duttaroy AK, Pathak S. Functional Foods: A Promising Strategy for Restoring Gut Microbiota Diversity Impacted by SARS-CoV-2 Variants. Nutrients 2023; 15:nu15112631. [PMID: 37299594 DOI: 10.3390/nu15112631] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Natural herbs and functional foods contain bioactive molecules capable of augmenting the immune system and mediating anti-viral functions. Functional foods, such as prebiotics, probiotics, and dietary fibers, have been shown to have positive effects on gut microbiota diversity and immune function. The use of functional foods has been linked to enhanced immunity, regeneration, improved cognitive function, maintenance of gut microbiota, and significant improvement in overall health. The gut microbiota plays a critical role in maintaining overall health and immune function, and disruptions to its balance have been linked to various health problems. SARS-CoV-2 infection has been shown to affect gut microbiota diversity, and the emergence of variants poses new challenges to combat the virus. SARS-CoV-2 recognizes and infects human cells through ACE2 receptors prevalent in lung and gut epithelial cells. Humans are prone to SARS-CoV-2 infection because their respiratory and gastrointestinal tracts are rich in microbial diversity and contain high levels of ACE2 and TMPRSS2. This review article explores the potential use of functional foods in mitigating the impact of SARS-CoV-2 variants on gut microbiota diversity, and the potential use of functional foods as a strategy to combat these effects.
Collapse
Affiliation(s)
- Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, Tamil Nadu, India
| | - Indumathi Somasundaram
- Department of Biotechnology Engineering, Kolhapur Institute of Technology's College of Engineering, Kolhapur 416012, Maharashtra, India
| | - Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, Tamil Nadu, India
| | - Samatha Jain Manoj
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, Tamil Nadu, India
| | - Husaina Banu
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, Tamil Nadu, India
| | - Pavane Mitta Suresh
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, Tamil Nadu, India
| | - Sujay Paul
- School of Engineering and Sciences, Tecnologico de Monterrey, Campus Queretaro, San Pablo 76130, Mexico
| | - Atil Bisgin
- Department of Medical Genetics, Medical Faculty, Cukurova University, Adana 01250, Turkey
| | - Hong Zhang
- Department of Medical Sciences, School of Medicine, Orebro University, SE-701 82 Orebro, Sweden
| | - Xiao-Feng Sun
- Division of Ocology, Department of Biomedical and Clinical Sciences, Linkoping University, SE-581 83 Linkoping, Sweden
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0313 Oslo, Norway
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai 603103, Tamil Nadu, India
| |
Collapse
|
18
|
Keathley J, White J, Reid G. The Impact of Nutrition, Physical Activity, Beneficial Microbes, and Fecal Microbiota Transplant for Improving Health. Life (Basel) 2023; 13:life13051124. [PMID: 37240769 DOI: 10.3390/life13051124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
The recognition that microbes are integral to human life has led to studies on how to manipulate them in favor of health outcomes. To date, there has been no conjoint recommendation for the intake of dietary compounds that can complement the ingested organisms in terms of promoting an improved health outcome. The aim of this review is to discuss how beneficial microbes in the form of probiotics, fermented foods, and donor feces are being used to manage health. In addition, we explore the rationale for selecting beneficial microbial strains and aligning diets to accommodate their propagation in the gut. A pilot clinical trial design is presented to examine the effects of probiotics and exercise in patients with phenylketonuria (PKU); it is the most common inborn error of amino acid metabolism, and it is a complication that requires lifelong dietary intervention. The example design is provided to illustrate the importance of using omics technology to see if the intervention elevates neuroactive biogenic amines in the plasma; increases the abundance of Eubacterium rectale, Coprococcus eutactus, Akkermansia muciniphila, or Butyricicoccus; and increases Escherichia/Shigella in the gut, all as markers of improved health. By emphasizing the combined importance of diet, microbial supplements, and the gut microbiome, we hope that future studies will better align these components, not only to improve outcomes, but also to enhance our understanding of the mechanisms.
Collapse
Affiliation(s)
- Justine Keathley
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Jessica White
- Department of Food and Nutritional Sciences, Brescia College, 1285 Western Road, London, ON N6G 1H2, Canada
| | - Gregor Reid
- Departments of Microbiology & Immunology and Surgery, The University of Western Ontario, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, 268 Grosvenor Street, London, ON N6A 4V2, Canada
| |
Collapse
|
19
|
Yntema T, Koonen DPY, Kuipers F. Emerging Roles of Gut Microbial Modulation of Bile Acid Composition in the Etiology of Cardiovascular Diseases. Nutrients 2023; 15:nu15081850. [PMID: 37111068 PMCID: PMC10141989 DOI: 10.3390/nu15081850] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Despite advances in preventive measures and treatment options, cardiovascular disease (CVD) remains the number one cause of death globally. Recent research has challenged the traditional risk factor profile and highlights the potential contribution of non-traditional factors in CVD, such as the gut microbiota and its metabolites. Disturbances in the gut microbiota have been repeatedly associated with CVD, including atherosclerosis and hypertension. Mechanistic studies support a causal role of microbiota-derived metabolites in disease development, such as short-chain fatty acids, trimethylamine-N-oxide, and bile acids, with the latter being elaborately discussed in this review. Bile acids represent a class of cholesterol derivatives that is essential for intestinal absorption of lipids and fat-soluble vitamins, plays an important role in cholesterol turnover and, as more recently discovered, acts as a group of signaling molecules that exerts hormonal functions throughout the body. Studies have shown mediating roles of bile acids in the control of lipid metabolism, immunity, and heart function. Consequently, a picture has emerged of bile acids acting as integrators and modulators of cardiometabolic pathways, highlighting their potential as therapeutic targets in CVD. In this review, we provide an overview of alterations in the gut microbiota and bile acid metabolism found in CVD patients, describe the molecular mechanisms through which bile acids may modulate CVD risk, and discuss potential bile-acid-based treatment strategies in relation to CVD.
Collapse
Affiliation(s)
- Tess Yntema
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Debby P Y Koonen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
20
|
Zheng Y, Zhang L, Bonfili L, de Vivo L, Eleuteri AM, Bellesi M. Probiotics Supplementation Attenuates Inflammation and Oxidative Stress Induced by Chronic Sleep Restriction. Nutrients 2023; 15:nu15061518. [PMID: 36986248 PMCID: PMC10054086 DOI: 10.3390/nu15061518] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Background: Insufficient sleep is a serious public health problem in modern society. It leads to increased risk of chronic diseases, and it has been frequently associated with cellular oxidative damage and widespread low-grade inflammation. Probiotics have been attracting increasing interest recently for their antioxidant and anti-inflammatory properties. Here, we tested the ability of probiotics to contrast oxidative stress and inflammation induced by sleep loss. Methods: We administered a multi-strain probiotic formulation (SLAB51) or water to normal sleeping mice and to mice exposed to 7 days of chronic sleep restriction (CSR). We quantified protein, lipid, and DNA oxidation as well as levels of gut-brain axis hormones and pro and anti-inflammatory cytokines in the brain and plasma. Furthermore, we carried out an evaluation of microglia morphology and density in the mouse cerebral cortex. Results: We found that CSR induced oxidative stress and inflammation and altered gut-brain axis hormones. SLAB51 oral administration boosted the antioxidant capacity of the brain, thus limiting the oxidative damage provoked by loss of sleep. Moreover, it positively regulated gut-brain axis hormones and reduced peripheral and brain inflammation induced by CSR. Conclusions: Probiotic supplementation can be a possible strategy to counteract oxidative stress and inflammation promoted by sleep loss.
Collapse
Affiliation(s)
- Yadong Zheng
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy
- Center for Neuroscience, University of Camerino, 62032 Camerino, MC, Italy
| | - Luyan Zhang
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy
- Center for Neuroscience, University of Camerino, 62032 Camerino, MC, Italy
- School of Food and Biological Engineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy
- Center for Neuroscience, University of Camerino, 62032 Camerino, MC, Italy
| | - Luisa de Vivo
- Center for Neuroscience, University of Camerino, 62032 Camerino, MC, Italy
- School of Pharmacy, University of Camerino, 62032 Camerino, MC, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy
- Center for Neuroscience, University of Camerino, 62032 Camerino, MC, Italy
| | - Michele Bellesi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy
- Center for Neuroscience, University of Camerino, 62032 Camerino, MC, Italy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
21
|
Gut Microbiota and Coronary Artery Disease: Current Therapeutic Perspectives. Metabolites 2023; 13:metabo13020256. [PMID: 36837875 PMCID: PMC9963624 DOI: 10.3390/metabo13020256] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The human gut microbiota is the community of microorganisms living in the human gut. This microbial ecosystem contains bacteria beneficial to their host and plays important roles in human physiology, participating in energy harvest from indigestible fiber, vitamin synthesis, and regulation of the immune system, among others. Accumulating evidence suggests a possible link between compositional and metabolic aberrations of the gut microbiota and coronary artery disease in humans. Manipulating the gut microbiota through targeted interventions is an emerging field of science, aiming at reducing the risk of disease. Among the interventions with the most promising results are probiotics, prebiotics, synbiotics, and trimethylamine N-oxide (TMAO) inhibitors. Contemporary studies of probiotics have shown an improvement of inflammation and endothelial cell function, paired with attenuated extracellular matrix remodeling and TMAO production. Lactobacilli, Bifidobacteria, and Bacteroides are some of the most well studied probiotics in experimental and clinical settings. Prebiotics may also decrease inflammation and lead to reductions in blood pressure, body weight, and hyperlipidemia. Synbiotics have been associated with an improvement in glucose homeostasis and lipid abnormalities. On the contrary, no evidence yet exists on the possible benefits of postbiotic use, while the use of antibiotics is not warranted, due to potentially deleterious effects. TMAO inhibitors such as 3,3-dimethyl-1-butanol, iodomethylcholine, and fluoromethylcholine, despite still being investigated experimentally, appear to possess anti-inflammatory, antioxidant, and anti-fibrotic properties. Finally, fecal transplantation carries conflicting evidence, mandating the need for further research. In the present review we summarize the links between the gut microbiota and coronary artery disease and elaborate on the varied therapeutic measures that are being explored in this context.
Collapse
|
22
|
Noormohammadi M, Ghorbani Z, Löber U, Mahdavi-Roshan M, Bartolomaeus TUP, Kazemi A, Shoaibinobarian N, Forslund SK. The effect of probiotic and synbiotic supplementation on appetite-regulating hormones and desire to eat: A systematic review and meta-analysis of clinical trials. Pharmacol Res 2023; 187:106614. [PMID: 36538981 DOI: 10.1016/j.phrs.2022.106614] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/11/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Recent studies have demonstrated the effect of probiotics, prebiotics, and synbiotics on adiponectin and leptin levels; however, those findings remain contested. The present study aimed to explore the impact of probiotics/synbiotics on appetite-regulating hormones and the desire to eat. METHODS A systematic review was conducted by searching the Medline (PubMed) and Scopus databases from inception to December 2021, using relevant keywords and MeSH terms, and appropriate randomized controlled trials (RCTs) were extracted. The standardized mean differences (SMD) and 95% confidence intervals (95%CIs) were calculated as part of the meta-analysis using a random-effect model to determine the mean effect sizes. Analysis of Galbraith plots and the Cochrane Chi-squared test were conducted to examine heterogeneity. RESULTS Meta-analysis of data from a total of 26 RCTs (n = 1536) showed a significant decrease in serum/plasma leptin concentration following probiotic/synbiotic supplementation (SMD: -0.38, 95%CI= -0.638, -0.124); P-value= 0.004; I2= 69.4%; P heterogeneity < 0.001). The leptin level decrease from probiotic/synbiotic supplementation was higher in patients with NAFLD than those with overweight/obesity or type 2 diabetes mellitus/ metabolic syndrome/ prediabetes. Probiotic/synbiotic supplementation was associated with a trending increase in adiponectin levels, stronger in patients with type 2 diabetes mellitus, metabolic syndrome, and prediabetes (SMD: 0.25, 95%CI= 0.04, 0.46) µg/mL; P-value= 0.021; I2 = 16.8%; P heterogeneity= 0.30). Additionally, supplementation with probiotic/synbiotic was linked to a slight increase in desire to eat (SMD: 0.34, 95%CI= 0.03, 0.66) P-value = 0.030; I2 = 39.4%; P heterogeneity= 0.16). CONCLUSION Our meta-analysis indicates a favorable impact of probiotic/synbiotic supplementation on regulating leptin and adiponectin secretion.
Collapse
Affiliation(s)
- Morvarid Noormohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Ghorbani
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Ulrike Löber
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Marjan Mahdavi-Roshan
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Theda U P Bartolomaeus
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Asma Kazemi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nargeskhatoon Shoaibinobarian
- Department of Nutrition, School of Medical Sciences and Technologies, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sofia K Forslund
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany; Structural and Computational Biology Unit, European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany.
| |
Collapse
|
23
|
Mahdavi-Roshan M, Mozafarihashjin M, Shoaibinobarian N, Ghorbani Z, Salari A, Savarrakhsh A, Hekmatdoost A. Evaluating the use of novel atherogenicity indices and insulin resistance surrogate markers in predicting the risk of coronary artery disease: a case‒control investigation with comparison to traditional biomarkers. Lipids Health Dis 2022; 21:126. [DOI: 10.1186/s12944-022-01732-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/03/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
Due to the contribution of coronary artery disease (CAD) to serious cardiovascular events, determining biomarkers that could robustly predict its risk would be of utmost importance. Thus, this research was designed to assess the value of traditional cardio-metabolic indices, and more novel atherogenicity indices and insulin resistance surrogate markers in the identification of individuals at risk of CAD.
Methods
A case‒control survey was conducted, in which 3085 individuals were enrolled. Their clinical and biochemical data were gathered at baseline. The investigated indices included the atherogenic index of plasma (AIP), triglyceride-glucose (TyG) index, TyG-body mass index (TyG-BMI), lipoprotein combine index (LCI), cholesterol index (CHOLINDEX), Castelli’s risk indices-I, II (CRI-I, CRI-II), and metabolic score for insulin resistance (METS − IR). To examine the relationship between these variables and CAD risk, multiple regression analyses adjusted for potential confounders were conducted.
Results
Overall, 774 angiographically confirmed CAD patients (mean age = 54 years) were compared with 3085 controls (mean age = 51 years). Higher triglyceride, total cholesterol and fasting blood sugar levels and lower HDL-C levels were related to an elevated risk of CAD (P-for-trend < 0.001), while the direct association between increased serum LDL-C concentrations and a greater risk of CAD only became apparent when excluding those with diabetes, and statin users. Among novel indices, greater values of the majority of these markers, including AIP, CRI-I, and -II, CHOLINDEX, LCI, and TyG-index, in comparison to the lower values, significantly elevated CAD risk (P-for-trend < 0.001).
Conclusion
According to the current findings, novel atherogenicity indices and insulin resistance surrogate markers, in particular, AIP, CRI-I and II, CHOLINDEX, LCI, and TyG-index, may be useful in predicting CAD risk.
Collapse
|
24
|
Noormohammadi M, Eslamian G, Kazemi SN, Rashidkhani B. Dietary acid load, alternative healthy eating index score, and bacterial vaginosis: is there any association? A case-control study. BMC Infect Dis 2022; 22:803. [PMID: 36303106 PMCID: PMC9615225 DOI: 10.1186/s12879-022-07788-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Changing the dietary pattern may be an alternative treatment for bacterial vaginosis, the prevalent vaginal infection in women. Methods: One hundred and forty-three bacterial vaginosis-affected women diagnosed by Amsel criteria and 151 healthy controls aged 18 to 45 entered the current case-control research. To calculate the alternative healthy eating index and dietary acid load score, food consumption was recorded with an accurate and precise food frequency questionnaire. The dietary acid load was measured by potential renal acid load (PRAL) and net endogenous acid production (NEAP) indices. Using logistic regression models, the association between the alternative healthy eating index and dietary acid load score with bacterial vaginosis was investigated. Results: The last tertile of the alternative healthy eating index had a 75% decreased odds of experiencing bacterial vaginosis in the adjusted model (adjusted odds ratio (aOR) = 0.25, 95% confidence interval (CI) = 0.12–0.53, P for trend = 0.001). Besides, vegetables (aOR = 0.34, 95% CI = 0.17–0.69, P for trend = 0.003), nuts and legumes (aOR = 0.44, 95% CI = 0.23–0.87, P for trend = 0.028), and meats (aOR = 0.31, 95% CI = 0.16–0.60, P for trend = 0.001) intake was linked to a decreased bacterial vaginosis odds. However, sugar-sweetened beverages and fruit juice (aOR = 3.47, 95% CI = 1.68–7.17, P for trend < 0.001), trans fatty acids (aOR = 2.29, 95% CI = 1.18–4.43, P for trend = 0.005), and sodium (aOR = 3.44, 95% CI = 1.67–7.06, P for trend = 0.002) intake were directly associated with bacterial vaginosis odds. There was no evidence of a link between dietary acid load and bacterial vaginosis. Conclusion: According to the present study’s findings, there is no correlation between dietary acid load and the likelihood of developing bacterial vaginosis. However, following a plant-based dietary pattern based on the healthy eating index may lead to a lower odds of bacterial vaginosis.
Collapse
Affiliation(s)
- Morvarid Noormohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Faculty of Public Health Branch, Iran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Eslamian
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, No 7, Hafezi St., Farahzadi Blvd, P.O.Box: 19395-4741, 1981619573, Tehran, Iran.
| | - Seyyedeh Neda Kazemi
- Department of Obstetrics and Gynecology, School of Medicine, Preventative Gynecology Research Center, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Rashidkhani
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Wang L, Wang S, Zhang Q, He C, Fu C, Wei Q. The role of the gut microbiota in health and cardiovascular diseases. MOLECULAR BIOMEDICINE 2022; 3:30. [PMID: 36219347 PMCID: PMC9554112 DOI: 10.1186/s43556-022-00091-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota is critical to human health, such as digesting nutrients, forming the intestinal epithelial barrier, regulating immune function, producing vitamins and hormones, and producing metabolites to interact with the host. Meanwhile, increasing evidence indicates that the gut microbiota has a strong correlation with the occurrence, progression and treatment of cardiovascular diseases (CVDs). In patients with CVDs and corresponding risk factors, the composition and ratio of gut microbiota have significant differences compared with their healthy counterparts. Therefore, gut microbiota dysbiosis, gut microbiota-generated metabolites, and the related signaling pathway may serve as explanations for some of the mechanisms about the occurrence and development of CVDs. Several studies have also demonstrated that many traditional and latest therapeutic treatments of CVDs are associated with the gut microbiota and its generated metabolites and related signaling pathways. Given that information, we summarized the latest advances in the current research regarding the effect of gut microbiota on health, the main cardiovascular risk factors, and CVDs, highlighted the roles and mechanisms of several metabolites, and introduced corresponding promising treatments for CVDs regarding the gut microbiota. Therefore, this review mainly focuses on exploring the role of gut microbiota related metabolites and their therapeutic potential in CVDs, which may eventually provide better solutions in the development of therapeutic treatment as well as the prevention of CVDs.
Collapse
Affiliation(s)
- Lu Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Shiqi Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Qing Zhang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chengqi He
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chenying Fu
- grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,grid.412901.f0000 0004 1770 1022Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Quan Wei
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| |
Collapse
|
26
|
Mahdavi-Roshan M, Shoaibinobarian N, Noormohammadi M, Fakhr Mousavi A, Savar Rakhsh A, Salari A, Ghorbani Z. Inflammatory Markers and Atherogenic Coefficient: Early Markers of Metabolic Syndrome. Int J Endocrinol Metab 2022; 20:e127445. [PMID: 36714188 PMCID: PMC9871968 DOI: 10.5812/ijem-127445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/11/2022] [Accepted: 08/09/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Considering the close link between metabolic syndrome (MetSyn) and cardiovascular diseases, considerable attention has been devoted to the identification of their shared underlying pathological mechanisms in recent decades. OBJECTIVES This study aimed to investigate the association between pro-inflammatory factors and newly-diagnosed MetSyn. METHODS This case-control study recruited obese and nonobese individuals who were newly diagnosed with MetSyn (cases, n = 84) and healthy individuals (controls, n = 83). The medical and sociodemographic data of the participants were collected on enrollment. Serum analysis was performed to ascertain the concentrations of tumor necrosis factor-alpha (TNF-α), C-reactive protein (CRP), fasting blood sugar (FBS), total cholesterol, triglyceride, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and atherogenic coefficient (AC). Multiple regression analysis was carried out to explore the relationship between inflammatory markers and AC with MetSyn odds. The Pearson correlation test was also performed to investigate the correlations between metabolic and inflammatory parameters. RESULTS Positive relationships were observed between the serum levels of TNF-α and CRP with the odds of MetSyn following controlling for confounders (adjusted odds ratio [AOR] = 1.32; 95% confidence interval [CI]: 1.01 - 1.72; AOR = 1.29; 95% CI: 1.18 - 1.41; respectively, P ≤ 0.03). Additionally, higher AC was accompanied by increased odds of MetSyn (AOR = 1.98; 95% CI: 1.31 - 2.98; P = 0.001). The Pearson correlation analysis also showed positive correlations between TNF-α levels and serum metabolic abnormalities, including elevated LDL-C, FBS, and AC and lowered HDL-C levels (P ≤ 0.02). CONCLUSIONS The present results revealed that higher serum concentrations of pro-inflammatory and atherogenic indices, including CRP, TNF-α, and AC, might be associated with elevated odds of newly diagnosed MetSyn regardless of potential confounders, particularly body mass index. The obtained findings might be moderated by the positive correlations observed between serum TNF-α, as the chronic inflammatory state indicator, and impaired lipid and glycemic markers.
Collapse
Affiliation(s)
- Marjan Mahdavi-Roshan
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nargeskhatoon Shoaibinobarian
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Morvarid Noormohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Aboozar Fakhr Mousavi
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amir Savar Rakhsh
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zeinab Ghorbani
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Corresponding Author: Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
27
|
Ghorbani Z, Kazemi A, Bartolomaeus TUP, Martami F, Noormohammadi M, Salari A, Löber U, Balou HA, Forslund SK, Mahdavi-Roshan M. The effect of probiotic and synbiotic supplementation on lipid parameters among patients with cardiometabolic risk factors: a systematic review and meta-analysis of clinical trials. Cardiovasc Res 2022; 119:933-956. [PMID: 35934838 DOI: 10.1093/cvr/cvac128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
Although the available evidence emphasizes the beneficial effects of probiotics in normalizing various cardiometabolic markers, there is still substantial uncertainty in this regard. Thus, we set out to determine the effect sizes of probiotics on blood lipid parameters more coherently. A systematic literature search of the Medline (PubMed) and Scopus databases was conducted from inception to February 12, 2021, applying both MeSH terms and free text terms to find the relevant randomized controlled trials (RCTs). The meta-analysis was conducted based on a random-effect model to calculate the mean effect sizes demonstrated as weighted mean differences (WMD) and the 95% confidence intervals (95%CI). To explore the heterogeneity, the Cochrane Chi-squared test, and analysis of Galbraith plots were performed. Meta-analysis of data from 40 RCTs (n = 2795) indicated a significant decrease in serum/plasma triglyceride (WMD (95%CI) -12.26 (-17.11- -7.41) mg/dL; P-value <0.001; I2 (%)= 29.9; P heterogeneity = 0.034)), total cholesterol (with high heterogeneity) (WMD (95%CI) -8.43 (-11.90- -4.95) mg/dL; P-value <0.001; I2 (%) =56.8; P heterogeneity < 0.001), LDL-C (WMD (95%CI) -5.08 (-7.61, -2.56) mg/dL; P-value <0.001; I2 (%) =42.7; P heterogeneity =0.002), and HDL-C (with high heterogeneity) (WMD (95%CI) 1.14 (0.23, 2.05) mg/dL; P-value =0.014; I2 (%) = 59.8; P heterogeneity < 0.001) following receiving probiotic/synbiotic supplements. Collectively, the current preliminary evidence supports the effectiveness of probiotics/synbiotics in improving dyslipidemia and various lipid parameters more prominently among subjects with hyperlipidemia, diabetes, and metabolic syndrome. However, large and well conducted RCTs are required to provide further convincing support for these results.
Collapse
Affiliation(s)
- Zeinab Ghorbani
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Asma Kazemi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Theda U P Bartolomaeus
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany
| | - Fahimeh Martami
- School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Morvarid Noormohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ulrike Löber
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany
| | - Heydar Ali Balou
- Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sofia K Forslund
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
| | - Marjan Mahdavi-Roshan
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|