151
|
Gut Microbiota and Metabolites in Atrial Fibrillation Patients and Their Changes after Catheter Ablation. Microbiol Spectr 2022; 10:e0107721. [PMID: 35384710 PMCID: PMC9045169 DOI: 10.1128/spectrum.01077-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The gut microbiota has been shown to be associated with multiple cardiovascular diseases, but there is little research on the gut microbiota and atrial fibrillation (AF); thus, how the gut microbiota and metabolites change in AF patients after catheter ablation is unclear. In this study, we used 16S rRNA high-throughput sequencing and nontargeted metabolomic detection to conduct horizontal and longitudinal analyses of the gut microbiota and metabolites of AF patients. Compared with a control group, species richness and diversity increased significantly in AF patients. Among them, opportunistic pathogenic bacteria, such as Klebsiella, Haemophilus, Streptococcus, and Enterococcus, were significantly increased, and symbiotic bacteria, such as Agathobacter and Butyrivibrio, were significantly reduced. After catheter ablation, intestinal symbiotic bacteria (Lactobacillus, Agathobacter, Lachnospira, etc.) were increased in most AF patients, while pathogenic bacteria (Ruminococcus, etc.) were reduced. Moreover, in AF patients, caffeine, which was negatively correlated with Klebsiella, was downregulated, and estradiol and ascorbic acid, which were positively correlated with Agathobacter, were also downregulated. After catheter ablation, citrulline, which was positively correlated with Ralstonia and Lactobacillus, was increased. Oleanolic acid, which was negatively correlated with Ralstonia was downregulated. In conclusion, our results not only show overall changes in the gut microbiota and metabolites in AF patients but also indicate their changes in the short term after catheter ablation. These data will provide novel possibilities for the future clinical diagnosis and treatment of AF. IMPORTANCE Gut microbiota and metabolites play a very important role in human health and can not only assess human health but also treat and prevent diseases. We analyzed the characteristics of the microbiota and metabolites in the human gut and found the effect of disease on gut microbiota and metabolites, which may be of important value in the pathogenesis of atrial fibrillation. At the same time, we also observed dynamic changes in gut microbiota and metabolites with the intervention of catheter ablation, which was not available in previous studies.
Collapse
|
152
|
Louisa M, Patintingan CGH, Wardhani BWK. Moringa Oleifera Lam. in Cardiometabolic Disorders: A Systematic Review of Recent Studies and Possible Mechanism of Actions. Front Pharmacol 2022; 13:792794. [PMID: 35431967 PMCID: PMC9006177 DOI: 10.3389/fphar.2022.792794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/28/2022] [Indexed: 12/18/2022] Open
Abstract
Cardiometabolic disorders (CMD) have become a global emergency and increasing burden on health and economic problems. Due to the increasing need for new drugs for cardiometabolic diseases, many alternative medicines from plants have been considered and studied. Moringa oleifera Lam. (MO), one of the native plants from several Asian countries, has been used empirically by people for various kinds of illnesses. In the present systematic review, we aimed to investigate the recent studies of MO in CMD and its possible mechanism of action. We systematically searched from three databases and summarized the data. This review includes a total of 108 papers in nonclinical studies and clinical trials of MO in cardiometabolic-related disorders. Moringa oleifera, extracts or isolated compound, exerts its effect on CMD through its antioxidative, anti-inflammatory actions resulting in the modulation in glucose and lipid metabolism and the preservation of target organ damage. Several studies supported the beneficial effect of MO in regulating the gut microbiome, which generates the diversity of gut microbiota and reduces the number of harmful bacteria in the caecum. Molecular actions that have been studied include the suppression of NF-kB translocation, upregulation of the Nrf2/Keap1 pathway, stimulation of total antioxidant capacity by reducing PKCζ activation, and inhibiting the Nox4 protein expression and several other proposed mechanisms. The present review found substantial evidence supporting the potential benefits of Moringa oleifera in cardiovascular or metabolic disorders.
Collapse
Affiliation(s)
- Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Bantari W. K. Wardhani
- Department of Pharmacology, Faculty of Military Pharmacy, Indonesia Defense University, West Java, Indonesia
| |
Collapse
|
153
|
Robeson MS, Manna K, Randolph C, Byrum S, Hakkak R. Short-Term Metformin Treatment Enriches Bacteroides dorei in an Obese Liver Steatosis Zucker Rat Model. Front Microbiol 2022; 13:834776. [PMID: 35432282 PMCID: PMC9006818 DOI: 10.3389/fmicb.2022.834776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/07/2022] [Indexed: 11/29/2022] Open
Abstract
Obesity is the leading cause of health-related diseases in the United States and World. Previously, we reported that obesity can change gut microbiota using the Zucker rat model. Metformin is an oral anti-hyperglycemic agent approved by the FDA to treat type 2 diabetes (T2D) in adults and children older than 10 years of age. The correlation of short-term metformin treatment and specific alterations to the gut microbiota in obese models is less known. Short-term metformin has been shown to reduce liver steatosis. Here we investigate the effects of short-term metformin treatment on population of gut microbiota profile in an obese rat model. Five week old obese (n = 12) female Zucker rats after 1 week of acclimation, received AIN-93 G diet for 8 weeks and then rats were randomly assigned into two groups (6 rats/group): (1) obese without metformin (ObC), or (2) obese with metformin (ObMet). Metformin was mixed with AIN-93G diet at 1,000 mg/kg of diet. Rats were weighed twice per week. All rats were sacrificed at the end of metformin treatment at 10 weeks and fecal samples were collected and kept at -80°C. Total microbial DNA was collected directly from the fecal samples used for shotgun-metagenomics sequencing and subsequently analyzed using MetaPlAn and HUMAnN. After stringent data filtering and quality control we found significant differences (p = 0.0007) in beta diversity (Aitchison distances) between the ObC vs. ObMet groups. Supervised and unsupervised analysis of the log-ratios Bacteroides dorei and B. massiliensis vs. all other Bacteroides spp., revealed that B. dorei and B. massiliensis were enriched in the ObMet group, while the remaining Bacteroides spp. where enriched in the ObC group (p = 0.002). The contributional diversity of pathways is also significantly associated by treatment group (p = 0.008). In summary, in the obese Zucker rat model, short-term metformin treatment changes the gut microbiota profile, particularly altering the composition Bacteroides spp. between ObC and ObMet.
Collapse
Affiliation(s)
- Michael S. Robeson
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Kanishka Manna
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Stephanie Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Reza Hakkak
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
154
|
Pagani IS, Poudel G, Wardill HR. A Gut Instinct on Leukaemia: A New Mechanistic Hypothesis for Microbiota-Immune Crosstalk in Disease Progression and Relapse. Microorganisms 2022; 10:microorganisms10040713. [PMID: 35456764 PMCID: PMC9029211 DOI: 10.3390/microorganisms10040713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 02/05/2023] Open
Abstract
Despite significant advances in the treatment of Chronic Myeloid and Acute Lymphoblastic Leukaemia (CML and ALL, respectively), disease progression and relapse remain a major problem. Growing evidence indicates the loss of immune surveillance of residual leukaemic cells as one of the main contributors to disease recurrence and relapse. More recently, there was an appreciation for how the host’s gut microbiota predisposes to relapse given its potent immunomodulatory capacity. This is especially compelling in haematological malignancies where changes in the gut microbiota have been identified after treatment, persisting in some patients for years after the completion of treatment. In this hypothesis-generating review, we discuss the interaction between the gut microbiota and treatment responses, and its capacity to influence the risk of relapse in both CML and ALL We hypothesize that the gut microbiota contributes to the creation of an immunosuppressive microenvironment, which promotes tumour progression and relapse.
Collapse
Affiliation(s)
- Ilaria S. Pagani
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide 5000, Australia; (G.P.); (H.R.W.)
- Faculty of Health and Medical Sciences, School of Medicine, University of Adelaide, Adelaide 5000, Australia
- Correspondence:
| | - Govinda Poudel
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide 5000, Australia; (G.P.); (H.R.W.)
- Faculty of Health and Medical Sciences, School of Medicine, University of Adelaide, Adelaide 5000, Australia
| | - Hannah R. Wardill
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide 5000, Australia; (G.P.); (H.R.W.)
- Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide 5000, Australia
| |
Collapse
|
155
|
Spisni E, Turroni S, Alvisi P, Spigarelli R, Azzinnari D, Ayala D, Imbesi V, Valerii MC. Nutraceuticals in the Modulation of the Intestinal Microbiota: Current Status and Future Directions. Front Pharmacol 2022; 13:841782. [PMID: 35370685 PMCID: PMC8971809 DOI: 10.3389/fphar.2022.841782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Pharmaceutical interest in the human intestinal microbiota has increased considerably, because of the increasing number of studies linking the human intestinal microbial ecology to an increasing number of non-communicable diseases. Many efforts at modulating the gut microbiota have been made using probiotics, prebiotics and recently postbiotics. However, there are other, still little-explored opportunities from a pharmaceutical point of view, which appear promising to obtain modifications of the microbiota structure and functions. This review summarizes all in vitro, in vivo and clinical studies demonstrating the possibility to positively modulate the intestinal microbiota by using probiotics, prebiotics, postbiotics, essential oils, fungus and officinal plants. For the future, clinical studies investigating the ability to impact the intestinal microbiota especially by using fungus, officinal and aromatic plants or their extracts are required. This knowledge could lead to effective microbiome modulations that might support the pharmacological therapy of most non-communicable diseases in a near future.
Collapse
Affiliation(s)
- Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
- *Correspondence: Enzo Spisni,
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Alvisi
- Pediatric Gastroenterology Unit, Maggiore Hospital, Bologna, Italy
| | - Renato Spigarelli
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Demetrio Azzinnari
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | | | - Veronica Imbesi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Chiara Valerii
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
156
|
Barcena ML, Aslam M, Pozdniakova S, Norman K, Ladilov Y. Cardiovascular Inflammaging: Mechanisms and Translational Aspects. Cells 2022; 11:cells11061010. [PMID: 35326461 PMCID: PMC8946971 DOI: 10.3390/cells11061010] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/07/2022] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
Aging is one of the major non-reversible risk factors for several chronic diseases, including cancer, type 2 diabetes, dementia, and cardiovascular diseases (CVD), and it is a key cause of multimorbidity, disability, and frailty (decreased physical activity, fatigue, and weight loss). The underlying cellular mechanisms are complex and consist of multifactorial processes, such as telomere shortening, chronic low-grade inflammation, oxidative stress, mitochondrial dysfunction, accumulation of senescent cells, and reduced autophagy. In this review, we focused on the molecular mechanisms and translational aspects of cardiovascular aging-related inflammation, i.e., inflammaging.
Collapse
Affiliation(s)
- Maria Luisa Barcena
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-525-359
| | - Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine I, Justus Liebig University, Aulweg 129, 35392 Giessen, Germany;
- Department of Cardiology, Kerckhoff Clinic GmbH, 61231 Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Rhein-Main, 61231 Bad Nauheim, Germany
| | - Sofya Pozdniakova
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Barcelona Biomedical Research Park (PRBB), Barcelona Institute for Global Health (ISGlobal), Doctor Aiguader, 88, 08003 Barcelona, Spain
| | - Kristina Norman
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Department of Nutrition & Gerontology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Yury Ladilov
- Department of Geriatrics and Medical Gerontology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (S.P.); (K.N.); (Y.L.)
- Department of Cardiovascular Surgery, Heart Center Brandenburg, Brandenburg Medical School Theodor Fontane, University Hospital, Ladeburger Str. 17, 16321 Bernau, Germany
| |
Collapse
|
157
|
Xu SS, Zhang XL, Liu SS, Feng ST, Xiang GM, Xu CJ, Fan ZY, Xu K, Wang N, Wang Y, Che JJ, Liu ZG, Mu YL, Li K. Multi-Omic Analysis in a Metabolic Syndrome Porcine Model Implicates Arachidonic Acid Metabolism Disorder as a Risk Factor for Atherosclerosis. Front Nutr 2022; 9:807118. [PMID: 35284467 PMCID: PMC8906569 DOI: 10.3389/fnut.2022.807118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/10/2022] [Indexed: 11/25/2022] Open
Abstract
Background The diet-induced gut microbiota dysbiosis has been suggested as a major risk factor for atherothrombosis, however, the detailed mechanism linking these conditions is yet to be fully understood. Methods We established a long-term excessive-energy diet-induced metabolic syndrome (MetS) inbred Wuzhishan minipig model, which is characterized by its genetic stability, small size, and human-like physiology. The metabolic parameters, atherosclerotic lesions, gut microbiome, and host transcriptome were analyzed. Metabolomics profiling revealed a linkage between gut microbiota and atherothrombosis. Results We showed that white atheromatous plaque was clearly visible on abdominal aorta in the MetS model. Furthermore, using metagenome and metatranscriptome sequencing, we discovered that the long-term excessive energy intake altered the local intestinal microbiota composition and transcriptional profile, which was most dramatically illustrated by the reduced abundance of SCFAs-producing bacteria including Bacteroides, Lachnospiraceae, and Ruminococcaceae in the MetS model. Liver and abdominal aorta transcriptomes in the MetS model indicate that the diet-induced gut microbiota dysbiosis activated host chronic inflammatory responses and significantly upregulated the expression of genes related to arachidonic acid-dependent signaling pathways. Notably, metabolomics profiling further revealed an intimate linkage between arachidonic acid metabolism and atherothrombosis in the host-gut microbial metabolism axis. Conclusions These findings provide new insights into the relationship between atherothrombosis and regulation of gut microbiota via host metabolomes and will be of potential value for the treatment of cardiovascular diseases in MetS.
Collapse
Affiliation(s)
- Song-Song Xu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xiu-Ling Zhang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Sha-Sha Liu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Animal Husbandry and Veterinary Department, Beijing Vocational College of Agriculture, Beijing, China
| | - Shu-Tang Feng
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Guang-Ming Xiang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chang-Jiang Xu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zi-Yao Fan
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kui Xu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Nan Wang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yue Wang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jing-Jing Che
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhi-Guo Liu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yu-Lian Mu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Yu-Lian Mu
| | - Kui Li
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kui Li
| |
Collapse
|
158
|
Padilla P, Andrade MJ, Peña FJ, Rodríguez A, Estévez M. Molecular mechanisms of the disturbance caused by malondialdehyde on probiotic Lactobacillus reuteri PL503. Microb Biotechnol 2022; 15:668-682. [PMID: 33356002 PMCID: PMC8867985 DOI: 10.1111/1751-7915.13723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/16/2020] [Indexed: 11/23/2022] Open
Abstract
This study aimed to provide insight into the molecular and genetic mechanisms implicated in the responses of Lactobacillus reuteri against the oxidative stress induced by malondialdehyde (MDA) by analysing protein oxidation and assessing the uspA and the dhaT genes. Four experimental groups were evaluated depending on the concentration of MDA added in Man, Rogosa and Sharpe (MRS) broth: Control (L. reuteri), 5 µM (L. reuteri + 5 µM MDA), 25 µM (L. reuteri + 25 µM MDA) and 100 µM (L. reuteri + 100 µM MDA). Three replicates were incubated at 37 °C for 24 h in microaerophilic conditions and sampled at 12, 16, 20 and 24 h. The upregulation of the uspA gene by L. reuteri indicates the recognition of MDA as a potential DNA-damaging agent. The dhaT gene, encoding a NADH-dependent-oxidoreductase, was also upregulated at the highest MDA concentrations. This gene was proposed to play a role in the antioxidant response of L. reuteri. The incubation of L. reuteri with MDA increased the production of ROS and caused thiol depletion and protein carbonylation. L. reuteri is proposed to detoxify pro-oxidative species while the underlying mechanism requires further elucidation.
Collapse
Affiliation(s)
- Patricia Padilla
- IPROCAR Research InstituteFood TechnologyUniversity of ExtremaduraCáceres10003Spain
- IPROCAR Research InstituteFood Hygiene and SafetyUniversity of ExtremaduraCáceres10003Spain
| | - María J. Andrade
- IPROCAR Research InstituteFood Hygiene and SafetyUniversity of ExtremaduraCáceres10003Spain
| | - Fernando J. Peña
- Laboratory of Equine Reproduction and Equine SpermatologyUniversity of ExtremaduraCáceres10003Spain
| | - Alicia Rodríguez
- IPROCAR Research InstituteFood Hygiene and SafetyUniversity of ExtremaduraCáceres10003Spain
| | - Mario Estévez
- IPROCAR Research InstituteFood TechnologyUniversity of ExtremaduraCáceres10003Spain
| |
Collapse
|
159
|
Jung E, Romero R, Yeo L, Gomez-Lopez N, Chaemsaithong P, Jaovisidha A, Gotsch F, Erez O. The etiology of preeclampsia. Am J Obstet Gynecol 2022; 226:S844-S866. [PMID: 35177222 PMCID: PMC8988238 DOI: 10.1016/j.ajog.2021.11.1356] [Citation(s) in RCA: 241] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/08/2023]
Abstract
Preeclampsia is one of the "great obstetrical syndromes" in which multiple and sometimes overlapping pathologic processes activate a common pathway consisting of endothelial cell activation, intravascular inflammation, and syncytiotrophoblast stress. This article reviews the potential etiologies of preeclampsia. The role of uteroplacental ischemia is well-established on the basis of a solid body of clinical and experimental evidence. A causal role for microorganisms has gained recognition through the realization that periodontal disease and maternal gut dysbiosis are linked to atherosclerosis, thus possibly to a subset of patients with preeclampsia. The recent reports indicating that SARS-CoV-2 infection might be causally linked to preeclampsia are reviewed along with the potential mechanisms involved. Particular etiologic factors, such as the breakdown of maternal-fetal immune tolerance (thought to account for the excess of preeclampsia in primipaternity and egg donation), may operate, in part, through uteroplacental ischemia, whereas other factors such as placental aging may operate largely through syncytiotrophoblast stress. This article also examines the association between gestational diabetes mellitus and maternal obesity with preeclampsia. The role of autoimmunity, fetal diseases, and endocrine disorders is discussed. A greater understanding of the etiologic factors of preeclampsia is essential to improve treatment and prevention.
Collapse
Affiliation(s)
- Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI; Detroit Medical Center, Detroit, MI.
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Piya Chaemsaithong
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Faculty of Medicine, Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adithep Jaovisidha
- Faculty of Medicine, Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, HaEmek Medical Center, Afula, Israel
| |
Collapse
|
160
|
Khan AA, Sirsat AT, Singh H, Cash P. Microbiota and cancer: current understanding and mechanistic implications. Clin Transl Oncol 2022; 24:193-202. [PMID: 34387847 PMCID: PMC8360819 DOI: 10.1007/s12094-021-02690-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
During last few decades, role of microbiota and its importance in several diseases has been a hot topic for research. The microbiota is considered as an accessory organ for maintaining normal physiology of an individual. These microbiota organisms which normally colonize several epithelial surfaces are known to secrete several small molecules leading to local and systemic effects on normal biological processes. The role of microbiota is also established in carcinogenesis as per several recent findings. The effects of microbiota on cancer is not only limited to their contribution in oncogenesis, but the overall susceptibility for oncogenesis and its subsequent progression, development of coinfections, and response to anticancer therapy is also found to be affected by microbiota. The information about microbiota and subsequent contributions of microbes in anticancer response motivated researchers in development of microbes-based anticancer therapeutics. We provided current status of microbiota contribution in oncogenesis with special reference to their mechanistic implications in different aspects of oncogenesis. In addition, the mechanistic implications of bacteria in anticancer therapy are also discussed. We conclude that several mechanisms of microbiota-mediated regulation of oncogenesis is known, but approaches must be focused on understanding contribution of microbiota as a community rather than single organisms-mediated effects.
Collapse
Affiliation(s)
- A A Khan
- Division of Microbiology, Indian Council of Medical Research-National AIDS Research Institute, Pune, Maharashtra, India.
| | - A T Sirsat
- Division of Microbiology, Indian Council of Medical Research-National AIDS Research Institute, Pune, Maharashtra, India
| | - H Singh
- Division of Molecular Biology, Indian Council of Medical Research-National AIDS Research Institute, Pune, Maharashtra, India
| | - P Cash
- Division of Applied Medicine, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, Scotland
| |
Collapse
|
161
|
Tian L, Liu R, Zhou Z, Xu X, Feng S, Kushmaro A, Marks RS, Wang D, Sun Q. Probiotic Characteristics of Lactiplantibacillus Plantarum N-1 and Its Cholesterol-Lowering Effect in Hypercholesterolemic Rats. Probiotics Antimicrob Proteins 2022; 14:337-348. [PMID: 35064922 DOI: 10.1007/s12602-021-09886-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2021] [Indexed: 11/28/2022]
Abstract
In this study, the probiotic potential and treatment effects of Lactiplantibacillus plantarum N-1 in hypercholesterolemic rats were investigated, and the possible regulatory mechanisms of lipid metabolism via short-chain fatty acids (SCFAs) and 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase were elucidated. The strain N-1 displayed probiotic properties of antioxidant capacity, adhesion to Caco-2 cells, susceptibility to antibiotics in vitro. The results in animal study showed that the total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels in serum and TC in liver declined significantly in both N-1 and simvastatin (Sta) treatment groups compared to the control (P < 0.05), and the extent of these decreases were similar between them. The expression of the HMG-CoA gene in the N-1 group was downregulated significantly by 31.18% compared to the control (P < 0.01), and the contents of butyrate and valerate in N-1 groups were significantly higher than those in both model and Sta group (P < 0.05). Thus, promoting the production of the intestinal SCFAs and inhibiting the expression of HMG-CoA reductase by L. plantarum N-1 may contribute to the improved lipid metabolism and thus lowering cholesterol level in rats. Our investigation indicated that L. plantarum N-1 has the potential to be developed into a functional food supplement for hypercholesterolemia treatment.
Collapse
Affiliation(s)
- Lei Tian
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, People's Republic of China.,Department of Biotechnology Engineering, Faculty of Engineering Sciences, Avram and Stella Goldstein-Goren, Ben Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Rongmei Liu
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, People's Republic of China.,Key Laboratory of Sichuan Province for Dairy Nutrition and Function, New Hope Dairy Co., Ltd., Chengdu, China
| | - Zhiwei Zhou
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, People's Republic of China
| | - Xiaofang Xu
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, People's Republic of China
| | - Su Feng
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, People's Republic of China
| | - Ariel Kushmaro
- Department of Biotechnology Engineering, Faculty of Engineering Sciences, Avram and Stella Goldstein-Goren, Ben Gurion University of the Negev, 84105, Beer-Sheva, Israel.,The Ilse Katz Centre for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Robert S Marks
- Department of Biotechnology Engineering, Faculty of Engineering Sciences, Avram and Stella Goldstein-Goren, Ben Gurion University of the Negev, 84105, Beer-Sheva, Israel.,The Ilse Katz Centre for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Dan Wang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, Sichuan, People's Republic of China.
| | - Qun Sun
- Key Laboratory of Bio-Resource and Eco-Environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, People's Republic of China.
| |
Collapse
|
162
|
Tsaturyan V, Kushugulova A, Mirzabekyan S, Sidamonidze K, Tsereteli D, Torok T, Pepoyan A. Promising Indicators in Probiotic-recommendations in COVID-19 and its Accompanying Diseases. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.7989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Scientific data suggests the possible beneficial role of probiotics in treatments for COVID-19, but the species/strains-specificity and disease-specificity of probiotics need high attention in choosing the appropriate probiotic in diseases, in particularly in the COVID-19. We hope this review will raise awareness of the COVID-19 probiotic recommendations, highlighting the latest scientific information about virus/hydrogen peroxide/probiotics and the importance of finding out of a specific “criterion” for the probiotics’ recommendation in this disease.
Collapse
|
163
|
Mukohda M, Mizuno R, Ozaki H. [Gut microflora and metabolic syndrome: new insight into the pathogenesis of hypertension]. Nihon Yakurigaku Zasshi 2022; 157:311-315. [PMID: 36047142 DOI: 10.1254/fpj.22035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Emerging evidences indicate that a microbial imbalance (dysbiosis) is linked to several diseases including metabolic cardiovascular diseases. A fecal microbiota transplantation from hypertensive human donor to germ-free mice caused blood pressure elevation. In addition, there is a report demonstrating that angiotensin II-induced hypertension and vascular dysfunction were attenuated in germ-free mice, suggesting that gut microbiome may mediate development of hypertension. Although detailed mechanism by which the dysbiosis induces an increased blood pressure remains unknown, changes in microbiome may modify host immune systems and induce inflammatory dysfunction in cardiovascular system, resulting in dysregulation of blood pressure. Some cohort studies demonstrated an association between a higher abundance of Streptococcaceae spp. and blood pressure. One recent report demonstrated that an increasing number of gram-positive Streptococcus was found in the feces of adult spontaneously hypertensive rats with an increased intestinal permeability. We hypothesized that increased bacterial toxin levels derived from gut Streptococcus may be a factor inducing blood pressure dysregulation. In this review, we discuss the possible role of microbiome in cardiovascular disease, especially hypertension.
Collapse
Affiliation(s)
- Masashi Mukohda
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science
| | - Risuke Mizuno
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science
| | - Hiroshi Ozaki
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science
| |
Collapse
|
164
|
OUP accepted manuscript. FEMS Microbiol Lett 2022; 369:6555450. [DOI: 10.1093/femsle/fnac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/17/2022] [Accepted: 03/26/2022] [Indexed: 11/14/2022] Open
|
165
|
Hill EB, Chen L, Bailey MT, Singh Khalsa A, Maltz R, Kelleher K, Spees CK, Zhu J, Loman BR. Facilitating a high-quality dietary pattern induces shared microbial responses linking diet quality, blood pressure, and microbial sterol metabolism in caregiver-child dyads. Gut Microbes 2022; 14:2150502. [PMID: 36457073 PMCID: PMC9721422 DOI: 10.1080/19490976.2022.2150502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
Low-resource individuals are at increased risk of obesity and cardiovascular disease (CVD), partially attributable to poor dietary patterns and dysfunctional microbiota. Dietary patterns in childhood play critical roles in physiological development and are shaped by caregivers, making caregiver-child dyads attractive targets for dietary interventions to reduce metabolic disease risk. Herein, we targeted low-resource caregiver-child dyads for a 10-week, randomized, controlled, multifaceted lifestyle intervention including: nutrition and physical activity education, produce harvesting, cooking demonstrations, nutrition counseling, and kinetic activites; to evaluate its effects on dietary patterns, CVD risk factors, and microbiome composition. Subjects in the lifestyle intervention group improved total diet quality, increased whole grain intake, decreased energy intake, and enhanced fecal elimination of the microbe-derived metabolite lithocholic acid (LCA) in contrast to control subjects. Microbiomes were highly personalized, similar within dyads, and altered by lifestyle intervention. Differential modeling of microbiome composition identified taxa associated with total diet quality, whole grain intake, and LCA elimination including recognized fiber-degrading bacteria such as Subdoligranulum, and bile acid metabolizing organisms like Bifidobacterium. Inclusion of taxa identified in diet and metabolite modeling within blood pressure models improved prediction accuracy of microbiome-blood pressure associations. Importantly, microbiota-blood pressure relationships were shared between dyads, implying shared host-microbiota responses to lifestyle intervention. Overall, these outcomes provide insight into mechanisms by which dietary interventions impact the gut-cardiovascular axis to reduce future CVD risk. Registered at clinicaltrials.gov: NCT05367674.
Collapse
Affiliation(s)
- Emily B. Hill
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, USA
| | - Li Chen
- Department of Human Sciences and James Comprehensive Cancer Center, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Michael T. Bailey
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Division of Primary Care Pediatrics, Center for Child Health Equity and Outcomes Research, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Amrik Singh Khalsa
- Division of Primary Care Pediatrics, Center for Child Health Equity and Outcomes Research, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ross Maltz
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Kelly Kelleher
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Colleen K. Spees
- Division of Medical Dietetics, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jiangjiang Zhu
- Department of Human Sciences and James Comprehensive Cancer Center, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Brett R. Loman
- Department of Animal Sciences, the University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, the University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
166
|
Wallenborn JT, Vonaesch P. OUP accepted manuscript. Gastroenterol Rep (Oxf) 2022; 10:goac010. [PMID: 35419206 PMCID: PMC8996373 DOI: 10.1093/gastro/goac010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/10/2021] [Accepted: 02/16/2022] [Indexed: 11/15/2022] Open
Abstract
The intestinal microbiota plays a crucial role in health and changes in its composition are linked with major global human diseases. Fully understanding what shapes the human intestinal microbiota composition and knowing ways of modulating the composition are critical for promotion of life-course health, combating diseases, and reducing global health disparities. We aim to provide a foundation for understanding what shapes the human intestinal microbiota on an individual and global scale, and how interventions could utilize this information to promote life-course health and reduce global health disparities. We briefly review experiences within the first 1,000 days of life and how long-term exposures to environmental elements or geographic specific cultures have lasting impacts on the intestinal microbiota. We also discuss major public health threats linked to the intestinal microbiota, including antimicrobial resistance and disappearing microbial diversity due to globalization. In order to promote global health, we argue that the interplay of the larger ecosystem with intestinal microbiota research should be utilized for future research and urge for global efforts to conserve microbial diversity.
Collapse
Affiliation(s)
- Jordyn T Wallenborn
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Pascale Vonaesch
- Department of Fundamental Microbiology, University of Lausanne, Bâtiment Biophore Campus UNIL-Sorge, Lausanne, Switzerland
- Corresponding author. Department of Fundamental Microbiology, University of Lausanne, 1015 Lausanne, Switzerland. Tel: +41-21-692-5600;
| |
Collapse
|
167
|
Coletto E, Latousakis D, Pontifex MG, Crost EH, Vaux L, Perez Santamarina E, Goldson A, Brion A, Hajihosseini MK, Vauzour D, Savva GM, Juge N. The role of the mucin-glycan foraging Ruminococcus gnavus in the communication between the gut and the brain. Gut Microbes 2022; 14:2073784. [PMID: 35579971 PMCID: PMC9122312 DOI: 10.1080/19490976.2022.2073784] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Ruminococcus gnavus is a prevalent member of the human gut microbiota, which is over-represented in inflammatory bowel disease and neurological disorders. We previously showed that the ability of R. gnavus to forage on mucins is strain-dependent and associated with sialic acid metabolism. Here, we showed that mice monocolonized with R. gnavus ATCC 29149 (Rg-mice) display changes in major sialic acid derivatives in their cecum content, blood, and brain, which is accompanied by a significant decrease in the percentage of sialylated residues in intestinal mucins relative to germ-free (GF) mice. Changes in metabolites associated with brain function such as tryptamine, indolacetate, and trimethylamine N-oxide were also detected in the cecal content of Rg-mice when compared to GF mice. Next, we investigated the effect of R. gnavus monocolonization on hippocampus cell proliferation and behavior. We observed a significant decrease of PSA-NCAM immunoreactive granule cells in the dentate gyrus (DG) of Rg-mice as compared to GF mice and recruitment of phagocytic microglia in the vicinity. Behavioral assessments suggested an improvement of the spatial working memory in Rg-mice but no change in other cognitive functions. These results were also supported by a significant upregulation of genes involved in proliferation and neuroplasticity. Collectively, these data provide first insights into how R. gnavus metabolites may influence brain regulation and function through modulation of granule cell development and synaptic plasticity in the adult hippocampus. This work has implications for further understanding the mechanisms underpinning the role of R. gnavus in neurological disorders.
Collapse
Affiliation(s)
- Erika Coletto
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Dimitrios Latousakis
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Matthew G Pontifex
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Emmanuelle H Crost
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Laura Vaux
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Estella Perez Santamarina
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Andrew Goldson
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Arlaine Brion
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Mohammad K Hajihosseini
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - David Vauzour
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - George M Savva
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Nathalie Juge
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| |
Collapse
|
168
|
Preethy S, Ranganathan N, Raghavan K, Dedeepiya VD, Ikewaki N, Abraham SJ. Integrating the Synergy of the Gut Microbiome into Regenerative Medicine: Relevance to Neurological Disorders. J Alzheimers Dis 2022; 87:1451-1460. [PMID: 35466942 PMCID: PMC9277691 DOI: 10.3233/jad-220313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 11/29/2022]
Abstract
A new paradigm of cell therapy-based approaches as a solution to several diseases caused by damage or loss of cells/tissues leading to organ failure heralded the birth of a new branch in medicine called regenerative medicine (RM), which was further fueled by in vitro cell expansion and tissue engineering (TE) technologies, including the ability to grow embryonic stem cells, induce pluripotent stem cells, and so on. RM addresses organ failure by repair, regeneration, or restoration, rejuvenation using cells, stem cells, or progenitor cells as tools having added cell-derived products also as a tool, and extracellular matrix component-based support, either direct or indirect (e.g., matrix induced autologous chondrocyte implantation) using scaffolds. Now, the main objective of RM is to solve the functional loss of cells that have evolved from cells as tools to cell-derived factors and scaffolds per se as tools. In this context, an important yet indispensable group of cells that constitute the major portion of the human body in terms of the number of cells having several essential roles to play, both directly and indirectly, starting from digestion and the immune system to the growing evidence of influencing neuronal function, aging, and carcinogenesis has been ignored. We would like to focus on these in this review as they should essentially be considered as a tool of RM, especially for neurological disorders for their vital role. What we are indicating is the second genome or the gut microbiome.
Collapse
Affiliation(s)
- Senthilkumar Preethy
- Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
| | | | - Kadalraja Raghavan
- Department of Paediatric Neurology, Jesuit Antonyraj memorial Inter-disciplinary Centre for Advanced Recovery and Education (JAICARE), Madurai, India
- Department of Paediatric Neurology, Sarvee Integra Private Limited, Chennai, India
| | | | - Nobunao Ikewaki
- Department of Medical Life Science, Kyushu University of Health and Welfare, Nobeoka, Japan
- Institute of Immunology, Junsei Educational Institute, Nobeoka, Japan
| | - Samuel J.K. Abraham
- Mary-Yoshio Translational Hexagon (MYTH), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
- Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan
- Antony- Xavier Interdisciplinary Scholastics (AXIS), GN Corporation Co. Ltd., Kofu, Japan
| |
Collapse
|
169
|
Phungviwatnikul T, Lee AH, Belchik SE, Suchodolski JS, Swanson KS. Weight loss and high-protein, high-fiber diet consumption impact blood metabolite profiles, body composition, voluntary physical activity, fecal microbiota, and fecal metabolites of adult dogs. J Anim Sci 2021; 100:6490144. [PMID: 34967874 PMCID: PMC8846339 DOI: 10.1093/jas/skab379] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/29/2021] [Indexed: 01/01/2023] Open
Abstract
Canine obesity is associated with reduced lifespan and metabolic dysfunction, but can be managed by dietary intervention. This study aimed to determine the effects of restricted feeding of a high-protein, high-fiber (HPHF) diet and weight loss on body composition, physical activity, blood metabolites, and fecal microbiota and metabolites of overweight dogs. Twelve spayed female dogs (age: 5.5 ± 1.1 yr; body weight [BW]: 14.8 ± 2.0 kg, body condition score [BCS]: 7.9 ± 0.8) were fed a HPHF diet during a 4-wk baseline phase to maintain BW. After baseline (week 0), dogs were first fed 80% of baseline intake and then adjusted to target 1.5% weekly weight loss for 24 wk. Body composition using dual-energy x-ray absorptiometry and blood samples (weeks 0, 6, 12, 18, and 24), voluntary physical activity (weeks 0, 7, 15, and 23), and fresh fecal samples for microbiota and metabolite analysis (weeks 0, 4, 8, 12, 16, 20, and 24) were measured over time. Microbiota data were analyzed using QIIME 2. All data were analyzed statistically over time using SAS 9.4. After 24 wk, dogs lost 31.2% of initial BW and had 1.43 ± 0.73% weight loss per week. BCS decreased (P < 0.0001) by 2.7 units, fat mass decreased (P < 0.0001) by 3.1 kg, and fat percentage decreased (P < 0.0001) by 11.7% with weight loss. Many serum metabolites and hormones were altered, with triglycerides, leptin, insulin, C-reactive protein, and interleukin-6 decreasing (P < 0.05) with weight loss. Relative abundances of fecal Bifidobacterium, Coriobacteriaceae UCG-002, undefined Muribaculaceae, Allobaculum, Eubacterium, Lachnospira, Negativivibacillus, Ruminococcus gauvreauii group, uncultured Erysipelotrichaceae, and Parasutterella increased (P < 0.05), whereas Prevotellaceae Ga6A1 group, Catenibacterium, Erysipelatoclostridium, Fusobacterium, Holdemanella, Lachnoclostridium, Lactobacillus, Megamonas, Peptoclostridium, Ruminococcus gnavus group, and Streptococcus decreased (P < 0.01) with weight loss. Despite the number of significant changes, a state of dysbiosis was not observed in overweight dogs. Fecal ammonia and secondary bile acids decreased, whereas fecal valerate increased with weight loss. Several correlations between gut microbial taxa and biological parameters were observed. Our results suggest that restricted feeding of a HPHF diet and weight loss promotes fat mass loss, minimizes lean mass loss, reduces inflammatory marker and triglyceride concentrations, and modulates fecal microbiota phylogeny and activity in overweight dogs.
Collapse
Affiliation(s)
| | - Anne H Lee
- Department of Animal Sciences, University of Illinois at Urbana – Champaign, Urbana, IL 61801, USA
| | - Sara E Belchik
- Department of Animal Sciences, University of Illinois at Urbana – Champaign, Urbana, IL 61801, USA
| | - Jan S Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana – Champaign, Urbana, IL 61801, USA,Department of Veterinary Clinical Medicine, University of Illinois at Urbana – Champaign, Urbana, IL 61801, USA,Division of Nutritional Sciences, University of Illinois at Urbana – Champaign, Urbana, IL 61801, USA,Corresponding author:
| |
Collapse
|
170
|
Zhu WJ, Liu Y, Cao YN, Peng LX, Yan ZY, Zhao G. Insights into Health-Promoting Effects of Plant MicroRNAs: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14372-14386. [PMID: 34813309 DOI: 10.1021/acs.jafc.1c04737] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Plant-derived microRNAs (miRNAs) play a significant role in human health and are "dark nutrients", as opposed to traditional plant nutrients, as well as important components of food diversification. Studies have revealed that multiple plant-derived miRNA pathways affect human health. First, plant miRNAs regulate plant growth and development and accumulation of metabolites, which alters the food quality and thus indirectly interferes with the health of the host. Moreover, when absorbed in vivo, some miRNAs may target the host cell mRNAs to affect protein expression. In addition, plant miRNAs target and reshape the human gut microbiota (GM), which interferes with the physiology and metabolism of the host. Therefore, miRNAs play a significant role in the cross-kingdom communication of plants, GM, and the host and in maintaining a balance of the three. Future contributions of plant miRNAs can bring new perspectives and opportunities to better understand food nutrition and health care research, which will facilitate the right exploitation of plant resources.
Collapse
Affiliation(s)
- Wen-Jing Zhu
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs; Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Yu Liu
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs; Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Ya-Nan Cao
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs; Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Lian-Xin Peng
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs; Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Zhu-Yun Yan
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, People's Republic of China
| | - Gang Zhao
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs; Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| |
Collapse
|
171
|
Yang C, Starnecker F, Pang S, Chen Z, Güldener U, Li L, Heinig M, Schunkert H. Polygenic risk for coronary artery disease in the Scottish and English population. BMC Cardiovasc Disord 2021; 21:586. [PMID: 34876023 PMCID: PMC8650538 DOI: 10.1186/s12872-021-02398-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/22/2021] [Indexed: 01/01/2023] Open
Abstract
Background Epidemiological studies have repeatedly observed a markedly higher risk for coronary artery disease (CAD) in Scotland as compared to England. Up to now, it is unclear whether environmental or genetic factors might explain this phenomenon. Methods Using UK Biobank (UKB) data, we assessed CAD risk, based on the Framingham risk score (FRS) and common genetic variants, to explore the respective contribution to CAD prevalence in Scotland (n = 31,963) and England (n = 317,889). We calculated FRS based on sex, age, body mass index (BMI), total cholesterol (TC), high density lipoprotein cholesterol (HDL-C), systolic blood pressure (SBP), antihypertensive medication, smoking status, and diabetes. We determined the allele frequency of published genome-wide significant risk CAD alleles and a weighted genetic risk score (wGRS) for quantifying genetic CAD risk. Results Prevalence of CAD was 16% higher in Scotland as compared to England (8.98% vs. 7.68%, P < 0.001). However, the FRS only predicted a marginally higher CAD risk (less than 1%) in Scotland (12.5 ± 10.5 vs.12.6 ± 10.6, P = 0.03). Likewise, the overall number of genome-wide significant variants affecting CAD risk (157.6 ± 7.7 and 157.5 ± 7.7; P = 0.12) and a wGRS for CAD (2.49 ± 0.25 in both populations, P = 0.14) were remarkably similar in the English and Scottish population. Interestingly, we observed substantial differences in the allele frequencies of individual risk variants. Of the previously described 163 genome-wide significant variants studied here, 35 variants had higher frequencies in Scotland, whereas 37 had higher frequencies in England (P < 0.001 each). Conclusions Neither the traditional risk factors included in the FRS nor a genetic risk score (GRS) based on established common risk alleles explained the higher CAD prevalence in Scotland. However, we observed marked differences in the distribution of individual risk alleles, which emphasizes that even geographically and ethnically closely related populations may display relevant differences in the genetic architecture of a common disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02398-4.
Collapse
Affiliation(s)
- Chuhua Yang
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,Deutsches Zentrum Für Herz- Und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Medical Graduate Center, Technische Universität München, Munich, Germany
| | - Fabian Starnecker
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,Deutsches Zentrum Für Herz- Und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Shichao Pang
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,Deutsches Zentrum Für Herz- Und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Zhifen Chen
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,Deutsches Zentrum Für Herz- Und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Ulrich Güldener
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Ling Li
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,Deutsches Zentrum Für Herz- Und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Informatics, Technische Universität München, Munich, Germany
| | - Matthias Heinig
- Department of Informatics, Technische Universität München, Munich, Germany.,Institute of Computational Biology ICB, Helmholtz Zentrum München (HMGU), Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany. .,Deutsches Zentrum Für Herz- Und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, Germany. .,Medical Graduate Center, Technische Universität München, Munich, Germany. .,German Heart Center Munich, Technical University Munich, Lazarettstraße 36, 80636, Munich, Germany.
| |
Collapse
|
172
|
RRM2 Improves Cardiomyocyte Proliferation after Myocardial Ischemia Reperfusion Injury through the Hippo-YAP Pathway. DISEASE MARKERS 2021; 2021:5089872. [PMID: 34868394 PMCID: PMC8639268 DOI: 10.1155/2021/5089872] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/09/2021] [Indexed: 01/13/2023]
Abstract
Objective Ribonucleotide reductase M2 (RRM2) as an enzyme that catalyzes the deoxyreduction of nucleosides to deoxyribonucleoside triphosphate (dNTP) has been extensively studied, and it plays a crucial role in regulating cell proliferation. However, its role in ischemia-reperfusion injury (I/RI) is still unclear. Methods SD rats were used as the research object to detect the expression of RRM2 in the myocardium by constructing an I/RI model. At the same time, primary SD neonatal rat cardiomyocytes were extracted, and hypoxia/reoxygenation (H/R) treatment simulated the I/RI model. Using transfection technology to overexpress RRM2 in cardiomyocytes, quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) was used to detect the expression of RRM2, Cell Counting Kit-8 (CCK-8) assay was used to detect cell viability, and immunofluorescence staining was used to detect Ki67 and EdU-positive cells. Western blot (WB) technology was used to detect YAP and its phosphorylation expression. Results qRT-PCR results indicated that the expression of RRM2 was inhibited in the model group, and cardiomyocytes overexpressing RRM2 can obviously promote the proliferation of primary cardiomyocytes and improve the damage of cardiac structure and function caused by I/R. At the same time, RRM2 can promote the increase of YAP protein expression and the increase of Cyclin D1 mRNA expression. Conclusion RRM2 expression was downregulated in myocardial tissue with I/R. After overexpression of RRM2, cardiomyocyte proliferation was upregulated and the Hippo-YAP signaling pathway was activated.
Collapse
|
173
|
Abadi B, Yazdanpanah N, Nokhodchi A, Rezaei N. Smart biomaterials to enhance the efficiency of immunotherapy in glioblastoma: State of the art and future perspectives. Adv Drug Deliv Rev 2021; 179:114035. [PMID: 34740765 DOI: 10.1016/j.addr.2021.114035] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022]
Abstract
Glioblastoma multiform (GBM) is considered as the most lethal tumor among CNS malignancies. Although immunotherapy has achieved remarkable advances in cancer treatment, it has not shown satisfactory results in GBM patients. Biomaterial science, along with nanobiotechnology, is able to optimize the efficiency of immunotherapy in these patients. They can be employed to provide the specific activation of immune cells in tumor tissue and combinational therapy as well as preventing systemic adverse effects resulting from hyperactivation of immune responses and off-targeting effect. Advance biomaterials in this field are classified into targeting nanocarriers and localized delivery systems. This review will offer an overview of immunotherapy strategies for glioblastoma and advance delivery systems for immunotherapeutics that may have a high potential in glioblastoma treatment.
Collapse
|
174
|
Liu L, Zhao Y, Ming J, Chen J, Zhao G, Chen ZY, Wang Y, Lei L. Polyphenol extract and essential oil of Amomum tsao-ko equally alleviate hypercholesterolemia and modulate gut microbiota. Food Funct 2021; 12:12008-12021. [PMID: 34755750 DOI: 10.1039/d1fo03082e] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
This study explored the effects of polyphenol extract (TKP) and essential oil (TKO) from Amomum tsao-ko Crevost et Lemaire (tsao-ko) on plasma total cholesterol and gut microbiota. Four groups of hamsters (n = 8 each) were fed one of four diets, respectively, namely a high-cholesterol diet (HCD) containing 0.1% cholesterol, a HCD containing 0.5% cholestyramine (PCD), a HCD with daily oral administration of 1000 mg per kg body weight TKP, and a HCD with daily oral administration of 200 mg per kg body weight TKO for 6 weeks. TKP and TKO equally lowered plasma total cholesterol (TC) by 13-18% via increasing the fecal elimination of total acidic sterols by 50-191%. This might be due to up-regulation of liver cholesterol 7α-hydroxylase (CYP7A1) at both transcriptional and translational levels. At a family level, TKP and TKO diets favorably modified the relative abundance of Ruminococcaceae, Erysipelotrichaceae, and Desulfovibrionaceae associated with acidic sterols and CYP7A1. It was therefore concluded that TKP and TKO were equally effective in alleviating hypercholesterolemia in hamsters via the interaction between gut microbiota and bile acid metabolism.
Collapse
Affiliation(s)
- Lijun Liu
- College of Food Science, Southwest University, Chongqing, People's Republic of China.
| | - Yimin Zhao
- School of Life Sciences, The Chinese University of Hong Kong, Shatin NT, Hong Kong, China.
| | - Jian Ming
- College of Food Science, Southwest University, Chongqing, People's Republic of China.
| | - Jia Chen
- College of Food Science, Southwest University, Chongqing, People's Republic of China.
| | - Guohua Zhao
- College of Food Science, Southwest University, Chongqing, People's Republic of China.
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin NT, Hong Kong, China.
| | - Yujie Wang
- Department of Chemistry, College of Resource and Environment, Baoshan University, Baoshan 678000, P. R. China.
| | - Lin Lei
- College of Food Science, Southwest University, Chongqing, People's Republic of China.
| |
Collapse
|
175
|
Zheng S, Piao C, Liu Y, Liu X, Liu T, Zhang X, Ren J, Liu Y, Zhu B, Du J. Glycan Biosynthesis Ability of Gut Microbiota Increased in Primary Hypertension Patients Taking Antihypertension Medications and Potentially Promoted by Macrophage-Adenosine Monophosphate-Activated Protein Kinase. Front Microbiol 2021; 12:719599. [PMID: 34803940 PMCID: PMC8600050 DOI: 10.3389/fmicb.2021.719599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/11/2021] [Indexed: 01/10/2023] Open
Abstract
Increasing evidences suggest that the gut microbiota have their contributions to the hypertension, but the metagenomic characteristics and potential regulating mechanisms in primary hypertension patients taking antihypertension drugs are not clear yet. We carried out a metagenomic analysis in 30 primary hypertension patients taking antihypertension medications and eight healthy adults without any medication. We found that bacterial strains from species, such as Bacteroides fragilis, Bacteroides vulgatus, Escherichia coli, Klebsiella pneumoniae, and Streptococcus vestibularis, were highly increased in patients; and these strains were reported to generate glycan, short-chain fatty acid (SCFA) and trimethylamine (TMA) or be opportunistic pathogens. Meanwhile, Dorea longicatena, Eubacterium hallii, Clostridium leptum, Faecalibacterium prausnitzii, and some other strains were greatly decreased in the patient group. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis found that ortholog groups and pathways related to glycan biosynthesis and multidrug resistance were significantly increased in the patient group, and some of the hub genes related to N-glycan biosynthesis were increased in the patient group, while those related to TMA precursor metabolism and amino acid metabolism both increased and decreased in the patient group. Metabolites tested by untargeted liquid chromatography–mass spectrometry (LC-MS) proved the decrease of acetic acid, choline, betaine, and several amino acids in patients’ fecal samples. Moreover, meta-analysis of recent studies found that almost all patients were taking at least one kind of drugs that were reported to regulate adenosine monophosphate-activated protein kinase (AMPK) pathway, so we further investigated if AMPK regulated the metagenomic changes by using angiotensin II-induced mouse hypertensive model on wild-type and macrophage-specific AMPK-knockout mice. We found that the changes in E. coli and Dorea and glycan biosynthesis-related orthologs and pathways were similar in our cohort and hypertensive wild-type mice but reversed after AMPK knockout. These results suggest that the gut microbiota-derived glycan, SCFA, TMA, and some other metabolites change in medication-taking primary hypertension patients and that medications might promote gut microbiota glycan biosynthesis through activating macrophage-AMPK.
Collapse
Affiliation(s)
- Shuai Zheng
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Chunmei Piao
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yan Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Xuxia Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Tingting Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Xiaoping Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Jingyuan Ren
- Department of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yulei Liu
- Department of Clinic Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Baoli Zhu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Collaborative Innovation Centre for Cardiovascular Disorders, Beijing, China.,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
176
|
Hu X, Fan Y, Li H, Zhou R, Zhao X, Sun Y, Zhang S. Impacts of Cigarette Smoking Status on Metabolomic and Gut Microbiota Profile in Male Patients With Coronary Artery Disease: A Multi-Omics Study. Front Cardiovasc Med 2021; 8:766739. [PMID: 34778417 PMCID: PMC8581230 DOI: 10.3389/fcvm.2021.766739] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Cigarette smoking has been considered a modifiable risk factor for coronary artery disease (CAD). Changes in gut microbiota and microbe-derived metabolites have been shown to influence atherosclerotic pathogenesis. However, the effect of cigarette smoking on the gut microbiome and serum metabolites in CAD remains unclear. Method: We profiled the gut microbiota and serum metabolites of 113 male participants with diagnosed CAD including 46 current smokers, 34 former smokers, and 33 never smokers by 16S ribosomal RNA (rRNA) gene sequencing and untargeted metabolomics study. A follow-up study was conducted. PICRUSt2 was used for metagenomic functional prediction of important bacterial taxa. Results: In the analysis of the microbial composition, the current smokers were characterized with depleted Bifidobacterium catenulatum, Akkermansia muciniphila, and enriched Enterococcus faecium, Haemophilus parainfluenzae compared with the former and never smokers. In the untargeted serum metabolomic study, we observed and annotated 304 discriminant metabolites, uniquely including ceramides, acyl carnitines, and glycerophospholipids. Pathway analysis revealed a significantly changed sphingolipids metabolism related to cigarette smoking. However, the change of the majority of the discriminant metabolites is possibly reversible after smoking cessation. While performing PICRUSt2 metagenomic prediction, several key enzymes (wbpA, nadM) were identified to possibly explain the cross talk between gut microbiota and metabolomic changes associated with smoking. Moreover, the multi-omics analysis revealed that specific changes in bacterial taxa were associated with disease severity or outcomes by mediating metabolites such as glycerophospholipids. Conclusions: Our results indicated that both the gut microbiota composition and metabolomic profile of current smokers are different from that of never smokers. The present study may provide new insights into understanding the heterogenic influences of cigarette smoking on atherosclerotic pathogenesis by modulating gut microbiota as well as circulating metabolites.
Collapse
Affiliation(s)
- Xiaomin Hu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.,Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yue Fan
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Hanyu Li
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Ruilin Zhou
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xinyue Zhao
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yueshen Sun
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shuyang Zhang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| |
Collapse
|
177
|
Antimicrobial and Antioxidant Potential of Vernonia Cinerea Extract Coated AuNPs. Indian J Microbiol 2021; 61:506-518. [PMID: 34744206 DOI: 10.1007/s12088-021-00976-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022] Open
Abstract
Abstract Green synthesis of nanoparticles is an important tool to reduce the harmful effects associated with traditional methods. In the present investigation, we have synthesised gold nanoparticles (AuNPs) using aqueous extract prepared from fresh aerial parts (leaf and stem) of Vernonia cinerea as bioreducing agent. The visual indication of change in colour from pale yellow to brown to ruby-red indicated the successful formation of the AuNPs. Characterization of nanoparticles was carried out by UV-visible spectroscopy, X-ray crystallography (XRD), Transmission electron microscopy (TEM) and Energy dispersive X-ray analysis (EDX). UV-Vis spectra showed a specific peak at 546 nm which was the initial confirmation of the biosynthesized AuNPs. TEM images showed spherical and triangular shape of AuNPs with an average size of 25 nm. From FTIR spectrum, different functional groups were identified that could be responsible for the formation, stabilization, and capping of biosynthesized AuNPs. Aqueous plant extract and biosynthesised AuNPs were separately tested for their antimicrobial activity against six bacterial strains and four fungal strains. Biosynthesised AuNPs (2 mg/ml) showed significantly high zone of inhibition against the selected bacterial strains as compared to the aqueous plant extract. Maximum zone of inhibition (18.2 mm) was observed with AuNPs against Streptococcus pyogenes whereas comparatively less value (12.5 mm) was recorded with the plant extract. Interestingly, the inhibitory activity observed against bacterial strains was even better than ampicillin. Antifungal activity recorded with AuNPs (5 mg/ml) was maximum (17.4 mm) against R. oryzae and it was higher than positive control (17.00 mm) and plant extract (13.2 mm).The present study clearly showed that AuNPs coated with Vernonia cinerea extract were as good as positive control in inhibiting bacterial and fungal growth. In addition, these AuNPs also showed good antioxidant potential which was comparable to ascorbic acid. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s12088-021-00976-w.
Collapse
|
178
|
Hajipour MJ, Saei AA, Walker ED, Conley B, Omidi Y, Lee K, Mahmoudi M. Nanotechnology for Targeted Detection and Removal of Bacteria: Opportunities and Challenges. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100556. [PMID: 34558234 PMCID: PMC8564466 DOI: 10.1002/advs.202100556] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 08/06/2021] [Indexed: 05/04/2023]
Abstract
The emergence of nanotechnology has created unprecedented hopes for addressing several unmet industrial and clinical issues, including the growing threat so-termed "antibiotic resistance" in medicine. Over the last decade, nanotechnologies have demonstrated promising applications in the identification, discrimination, and removal of a wide range of pathogens. Here, recent insights into the field of bacterial nanotechnology are examined that can substantially improve the fundamental understanding of nanoparticle and bacteria interactions. A wide range of developed nanotechnology-based approaches for bacterial detection and removal together with biofilm eradication are summarized. The challenging effects of nanotechnologies on beneficial bacteria in the human body and environment and the mechanisms of bacterial resistance to nanotherapeutics are also reviewed.
Collapse
Affiliation(s)
- Mohammad J. Hajipour
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LansingMI48824USA
| | - Amir Ata Saei
- Division of Physiological Chemistry IDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholm171 65Sweden
| | - Edward D. Walker
- Department of EntomologyMichigan State UniversityEast LansingMI48824USA
- Department of Microbiology and Molecular GeneticsMichigan State UniversityEast LansingMI48824USA
| | - Brian Conley
- Department of Chemistry and Chemical BiologyRutgersThe State University of New JerseyPiscatawayNJ08854USA
| | - Yadollah Omidi
- Department of Pharmaceutical SciencesCollege of PharmacyNova Southeastern UniversityFort LauderdaleFL33328USA
| | - Ki‐Bum Lee
- Department of Chemistry and Chemical BiologyRutgersThe State University of New JerseyPiscatawayNJ08854USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LansingMI48824USA
| |
Collapse
|
179
|
Zeibich L, Koebele SV, Bernaud VE, Ilhan ZE, Dirks B, Northup-Smith SN, Neeley R, Maldonado J, Nirmalkar K, Files JA, Mayer AP, Bimonte-Nelson HA, Krajmalnik-Brown R. Surgical Menopause and Estrogen Therapy Modulate the Gut Microbiota, Obesity Markers, and Spatial Memory in Rats. Front Cell Infect Microbiol 2021; 11:702628. [PMID: 34660336 PMCID: PMC8515187 DOI: 10.3389/fcimb.2021.702628] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023] Open
Abstract
Menopause in human females and subsequent ovarian hormone deficiency, particularly concerning 17β-estradiol (E2), increase the risk for metabolic dysfunctions associated with obesity, diabetes type 2, cardiovascular diseases, and dementia. Several studies indicate that these disorders are also strongly associated with compositional changes in the intestinal microbiota; however, how E2 deficiency and hormone therapy affect the gut microbial community is not well understood. Using a rat model, we aimed to evaluate how ovariectomy (OVX) and subsequent E2 administration drive changes in metabolic health and the gut microbial community, as well as potential associations with learning and memory. Findings indicated that OVX-induced ovarian hormone deficiency and E2 treatment had significant impacts on several health-affecting parameters, including (a) the abundance of some intestinal bacterial taxa (e.g., Bifidobacteriaceae and Porphyromonadaceae), (b) the abundance of microbial short-chain fatty acids (SCFAs) (e.g., isobutyrate), (c) weight/BMI, and (d) high-demand spatial working memory following surgical menopause. Furthermore, exploratory correlations among intestinal bacteria abundance, cognition, and BMI underscored the putative influence of surgical menopause and E2 administration on gut-brain interactions. Collectively, this study showed that surgical menopause is associated with physiological and behavioral changes, and that E2-linked compositional changes in the intestinal microbiota might contribute to some of its related negative health consequences. Overall, this study provides novel insights into interactions among endocrine and gastrointestinal systems in the post-menopausal life stage that collectively alter the risk for the development and progression of cardiovascular, metabolic, and dementia-related diseases.
Collapse
Affiliation(s)
- Lydia Zeibich
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Victoria E Bernaud
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Zehra Esra Ilhan
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Blake Dirks
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Steven N Northup-Smith
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Rachel Neeley
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Juan Maldonado
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States.,Genomics Core, Arizona State University, Tempe, AZ, United States
| | - Khemlal Nirmalkar
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| | - Julia A Files
- Division of Women's Health Internal Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Anita P Mayer
- Division of Women's Health Internal Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ, United States.,Arizona Alzheimer's Consortium, Phoenix, AZ, United States
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
180
|
Arun KB, Madhavan A, Sindhu R, Emmanual S, Binod P, Pugazhendhi A, Sirohi R, Reshmy R, Awasthi MK, Gnansounou E, Pandey A. Probiotics and gut microbiome - Prospects and challenges in remediating heavy metal toxicity. JOURNAL OF HAZARDOUS MATERIALS 2021; 420:126676. [PMID: 34329091 DOI: 10.1016/j.jhazmat.2021.126676] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 07/15/2021] [Indexed: 05/26/2023]
Abstract
The gut microbiome, often referred to as "super organ", comprises up to a hundred trillion microorganisms, and the species diversity may vary from person to person. They perform a decisive role in diverse biological functions related to metabolism, immunity and neurological responses. However, the microbiome is sensitive to environmental pollutants, especially heavy metals. There is continuous interaction between heavy metals and the microbiome. Heavy metal exposure retards the growth and changes the structure of the phyla involved in the gut microbiome. Meanwhile, the gut microbiome tries to detoxify the heavy metals by altering the physiological conditions, intestinal permeability, enhancing enzymes for metabolizing heavy metals. This review summarizes the effect of heavy metals in altering the gut microbiome, the mechanism by which gut microbiota detoxifies heavy metals, diseases developed due to heavy metal-induced dysbiosis of the gut microbiome, and the usage of probiotics along with advancements in developing improved recombinant probiotic strains for the remediation of heavy metal toxicity.
Collapse
Affiliation(s)
- K B Arun
- Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, Kerala, India
| | - Aravind Madhavan
- Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, Kerala, India
| | - Raveendran Sindhu
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Trivandrum 695019, Kerala, India
| | - Shibitha Emmanual
- Department of Zoology, St. Joseph's College, Thrissur 680121, Kerala, India
| | - Parameswaran Binod
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Trivandrum 695019, Kerala, India
| | - Arivalagan Pugazhendhi
- School of Renewable Energy, Maejo University, Chiang Mai 50290, Thailand; College of Medical and Health Science, Asia University, Taichung, Taiwan ROC
| | - Ranjna Sirohi
- Department of Chemical & Biological Engineering, Korea University, Seoul 136713, Republic of Korea; Centre for Energy and Environmental Sustainability, Lucknow 226029, Uttar Pradesh, India
| | - R Reshmy
- Post Graduate and Research Department of Chemistry, Bishop Moore College, Mavelikara 690110, Kerala, India
| | - Mukesh Kumar Awasthi
- College of Natural Resources and Environment, North West A & F University, Yangling, Shaanxi 712100, China
| | - Edgard Gnansounou
- Ecole Polytechnique Federale de Lausanne, ENAC GR-GN, CH-1015 Lausanne, Switzerland
| | - Ashok Pandey
- Centre for Innovation and Translational Research, CSIR, Indian Institute for Toxicology Research, Lucknow 226001, Uttar Pradesh, India; Centre for Energy and Environmental Sustainability, Lucknow 226029, Uttar Pradesh, India.
| |
Collapse
|
181
|
Sharma V, Sharma V, Shahjouei S, Li J, Chaudhary D, Khan A, Wolk DM, Zand R, Abedi V. At the Intersection of Gut Microbiome and Stroke: A Systematic Review of the Literature. Front Neurol 2021; 12:729399. [PMID: 34630304 PMCID: PMC8498333 DOI: 10.3389/fneur.2021.729399] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Ischemic and hemorrhagic stroke are associated with a high rate of long-term disability and death. Recent investigations focus efforts to better understand how alterations in gut microbiota composition influence clinical outcomes. A key metabolite, trimethylamine N-oxide (TMAO), is linked to multiple inflammatory, vascular, and oxidative pathways. The current biochemical underpinnings of microbial effects on stroke remain largely understudied. The goal of our study is to explore the current literature to explain the interactions between the human gut microbiome and stroke progression, recovery, and outcome. We also provide a descriptive review of TMAO. Methods: A systematic literature search of published articles between January 1, 1990, and March 22, 2020, was performed on the PubMed database to identify studies addressing the role of the microbiome and TMAO in the pathogenesis and recovery of acute stroke. Our initial investigation focused on human subject studies and was further expanded to include animal studies. Relevant articles were included, regardless of study design. The analysis included reviewers classifying and presenting selected articles by study design and sample size in a chart format. Results: A total of 222 titles and abstracts were screened. A review of the 68 original human subject articles resulted in the inclusion of 24 studies in this review. To provide further insight into TMAO as a key player, an additional 40 articles were also reviewed and included. Our findings highlighted that alterations in richness and abundance of gut microbes and increased plasma TMAO play an important role in vascular events and outcomes. Our analysis revealed that restoration of a healthy gut, through targeted TMAO-reducing therapies, could provide alternative secondary prevention for at-risk patients. Discussion: Biochemical interactions between the gut microbiome and inflammation, resulting in metabolic derangements, can affect stroke progression and outcomes. Clinical evidence supports the importance of TMAO in modulating underlying stroke risk factors. Lack of standardization and distinct differences in sample sizes among studies are major limitations.
Collapse
Affiliation(s)
- Vishakha Sharma
- Kansas City University College of Osteopathic Medicine, Kansas City, MO, United States
| | - Vaibhav Sharma
- Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | - Shima Shahjouei
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States
| | - Jiang Li
- Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA, United States
| | - Durgesh Chaudhary
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States
| | - Ayesha Khan
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States.,Geisinger Health System, Geisinger Northeast Internal Medicine Residency, Wilkes Barre, PA, United States
| | - Donna M Wolk
- Department of Laboratory Medicine, Geisinger Health System, Diagnostic Medicine Institute, Danville, PA, United States
| | - Ramin Zand
- Geisinger Health System, Geisinger Neuroscience Institute, Danville, PA, United States
| | - Vida Abedi
- Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA, United States.,Department of Public Health Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
182
|
Kassan A, Ait-Aissa K, Kassan M. Hypothalamic miR-204 Induces Alteration of Heart Electrophysiology and Neurogenic Hypertension by Regulating the Sympathetic Nerve Activity: Potential Role of Microbiota. Cureus 2021; 13:e18783. [PMID: 34692262 PMCID: PMC8523185 DOI: 10.7759/cureus.18783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 02/06/2023] Open
Abstract
There is abundant evidence demonstrating the association between gut dysbiosis and neurogenic diseases such as hypertension. A common characteristic of resistant hypertension is the chronic elevation in sympathetic nervous system (SNS) activity accompanied by increased release of norepinephrine (NE), indicating a neurogenic component that contributes to the development of hypertension. Factors that modulate the sympathetic tone to the cardiovascular system in hypertensive patients are still poorly understood. Research has identified an interaction between the brain and the gut, and this interaction plays a possible role in the mechanism of heart damage-induced hypertension. Data, however, remain scarce, and further study is required to define the role of microbiota in sympathetic neural function and its relationship with heart damage and blood pressure (BP) control. Experimental evidence has pointed toward a bidirectional relationship between alterations in the types of bacteria present in the gut and neurogenic diseases, such as hypertension. Our published data showed that miR-204, a microRNA that plays an important role in the CNS function, is affected by gut dysbiosis. Therefore, miR-204 could be a key element that regulates normal sinus rhythm and neuronal hypertension. In this review, we will shed light on the potential mechanism by which microbiota affects hypothalamic miR-204, which in turn, could hinder the sympathetic nerve drive to the cardiovascular system leading to arrhythmia and hypertension.
Collapse
Affiliation(s)
- Adam Kassan
- School of Pharmacy, West Coast University, Los Angeles, USA
| | | | - Modar Kassan
- Physiology, The University of Tennessee Health Science Center, Memphis, USA
| |
Collapse
|
183
|
Cheng CK, Huang Y. The gut-cardiovascular connection: new era for cardiovascular therapy. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:23-46. [PMID: 37724079 PMCID: PMC10388818 DOI: 10.1515/mr-2021-0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/02/2021] [Indexed: 09/20/2023]
Abstract
Our gut microbiome is constituted by trillions of microorganisms including bacteria, archaea and eukaryotic microbes. Nowadays, gut microbiome has been gradually recognized as a new organ system that systemically and biochemically interact with the host. Accumulating evidence suggests that the imbalanced gut microbiome contributes to the dysregulation of immune system and the disruption of cardiovascular homeostasis. Specific microbiome profiles and altered intestinal permeability are often observed in the pathophysiology of cardiovascular diseases. Gut-derived metabolites, toxins, peptides and immune cell-derived cytokines play pivotal roles in the induction of inflammation and the pathogenesis of dysfunction of heart and vasculature. Impaired crosstalk between gut microbiome and multiple organ systems, such as gut-vascular, heart-gut, gut-liver and brain-gut axes, are associated with higher cardiovascular risks. Medications and strategies that restore healthy gut microbiome might therefore represent novel therapeutic options to lower the incidence of cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science; The Chinese University of Hong Kong, Hong Kong SAR999077, China
- Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR999077, China
| | - Yu Huang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science; The Chinese University of Hong Kong, Hong Kong SAR999077, China
- Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR999077, China
| |
Collapse
|
184
|
Bin-Jumah MN, Gilani SJ, Hosawi S, Al-Abbasi FA, Zeyadi M, Imam SS, Alshehri S, Ghoneim MM, Nadeem MS, Kazmi I. Pathobiological Relationship of Excessive Dietary Intake of Choline/L-Carnitine: A TMAO Precursor-Associated Aggravation in Heart Failure in Sarcopenic Patients. Nutrients 2021; 13:3453. [PMID: 34684454 PMCID: PMC8540684 DOI: 10.3390/nu13103453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 02/04/2023] Open
Abstract
The microecological environment of the gastrointestinal tract is altered if there is an imbalance between the gut microbiota phylases, resulting in a variety of diseases. Moreover, progressive age not only slows down physical activity but also reduces the fat metabolism pathway, which may lead to a reduction in the variety of bacterial strains and bacteroidetes' abundance, promoting firmicutes and proteobacteria growth. As a result, dysbiosis reduces physiological adaptability, boosts inflammatory markers, generates ROS, and induces the destruction of free radical macromolecules, leading to sarcopenia in older patients. Research conducted at various levels indicates that the microbiota of the gut is involved in pathogenesis and can be considered as the causative agent of several cardiovascular diseases. Local and systematic inflammatory reactions are caused in patients with heart failure, as ischemia and edema are caused by splanchnic hypoperfusion and enable both bacterial metabolites and bacteria translocation to enter from an intestinal barrier, which is already weakened, to the blood circulation. Multiple diseases, such as HF, include healthy microbe-derived metabolites. These key findings demonstrate that the gut microbiota modulates the host's metabolism, either specifically or indirectly, by generating multiple metabolites. Currently, the real procedures that are an analogy to the symptoms in cardiac pathologies, such as cardiac mass dysfunctions and modifications, are investigated at a minimum level in older patients. Thus, the purpose of this review is to summarize the existing knowledge about a particular diet, including trimethylamine, which usually seems to be effective for the improvement of cardiac and skeletal muscle, such as choline and L-carnitine, which may aggravate the HF process in sarcopenic patients.
Collapse
Affiliation(s)
- May Nasser Bin-Jumah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
- Environment and Biomaterial Unit, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Sadaf Jamal Gilani
- Department of Basic Health Sciences, Preparatory Year, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.H.); (F.A.A.-A.); (M.Z.); (M.S.N.)
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.H.); (F.A.A.-A.); (M.Z.); (M.S.N.)
| | - Mustafa Zeyadi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.H.); (F.A.A.-A.); (M.Z.); (M.S.N.)
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.S.I.); (S.A.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.S.I.); (S.A.)
| | - Mohammed M Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.H.); (F.A.A.-A.); (M.Z.); (M.S.N.)
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.H.); (F.A.A.-A.); (M.Z.); (M.S.N.)
| |
Collapse
|
185
|
Alhajri N, Khursheed R, Ali MT, Abu Izneid T, Al-Kabbani O, Al-Haidar MB, Al-Hemeiri F, Alhashmi M, Pottoo FH. Cardiovascular Health and The Intestinal Microbial Ecosystem: The Impact of Cardiovascular Therapies on The Gut Microbiota. Microorganisms 2021; 9:2013. [PMID: 34683334 PMCID: PMC8541580 DOI: 10.3390/microorganisms9102013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/14/2023] Open
Abstract
It has become evident over the past several years that the intestinal microbial ecosystem plays a critical role in the development and prevention of cardiovascular diseases (CVDs) and other metabolic disorders, such as hypertension, obesity, diabetes mellitus, and metabolic syndrome. The intestinal microbiota ecosystem functions as a major virtual endocrine organ that interacts and responds to molecules' signals within the host. Several meta-organismal pathways are involved in the gut-host interaction, including trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFA). Host phenotype and cardiovascular diseases (CVDs) varying from hypertension, insulin resistance, and obesity to more specific inflammatory processes, such as atherosclerosis and hypercoagulability, have shown to be affected by the gut-host interaction. Additionally, several studies that involved animals and humans demonstrated a striking connection between the development of new CVDs and an imbalance in the gut microbiota composition along with the presence of their derived metabolites. Through this review article, we aim to evaluate the role of the normal gut microbiota ecosystem, its association with CVDs, effects of the therapies used to control and manage CVDs in the gut microbiota environment and explore potential therapeutic interventions to amplify disease outcomes in patients with CVDs.
Collapse
Affiliation(s)
- Noora Alhajri
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
- Department of Medicine, Sheikh Shakhbout Medical City (SSMC), Abu Dhabi P.O. Box 11001, United Arab Emirates
| | - Rubiya Khursheed
- Department of Pharmaceutical Sciences, Lovely Professional University, Punjab 144403, India;
| | - Mohammad Taher Ali
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia; (M.T.A.); (F.H.P.)
| | - Tareq Abu Izneid
- Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, Abu Dhabi P.O. Box 112612, United Arab Emirates;
| | - Oumaima Al-Kabbani
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Mahdia B. Al-Haidar
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Fatima Al-Hemeiri
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Mohamed Alhashmi
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia; (M.T.A.); (F.H.P.)
| |
Collapse
|
186
|
Ke S, Mitchell SJ, MacArthur MR, Kane AE, Sinclair DA, Venable EM, Chadaideh KS, Carmody RN, Grodstein F, Mitchell JR, Liu Y. Gut Microbiota Predicts Healthy Late-Life Aging in Male Mice. Nutrients 2021; 13:3290. [PMID: 34579167 PMCID: PMC8467910 DOI: 10.3390/nu13093290] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022] Open
Abstract
Calorie restriction (CR) extends lifespan and retards age-related chronic diseases in most species. There is growing evidence that the gut microbiota has a pivotal role in host health and age-related pathological conditions. Yet, it is still unclear how CR and the gut microbiota are related to healthy aging. Here, we report findings from a small longitudinal study of male C57BL/6 mice maintained on either ad libitum or mild (15%) CR diets from 21 months of age and tracked until natural death. We demonstrate that CR results in a significantly reduced rate of increase in the frailty index (FI), a well-established indicator of aging. We observed significant alterations in diversity, as well as compositional patterns of the mouse gut microbiota during the aging process. Interrogating the FI-related microbial features using machine learning techniques, we show that gut microbial signatures from 21-month-old mice can predict the healthy aging of 30-month-old mice with reasonable accuracy. This study deepens our understanding of the links between CR, gut microbiota, and frailty in the aging process of mice.
Collapse
Affiliation(s)
- Shanlin Ke
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (S.K.); (F.G.)
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Sarah J. Mitchell
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland;
| | - Michael R. MacArthur
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland;
| | - Alice E. Kane
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; (A.E.K.); (D.A.S.)
| | - David A. Sinclair
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; (A.E.K.); (D.A.S.)
| | - Emily M. Venable
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; (E.M.V.); (K.S.C.); (R.N.C.)
| | - Katia S. Chadaideh
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; (E.M.V.); (K.S.C.); (R.N.C.)
| | - Rachel N. Carmody
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; (E.M.V.); (K.S.C.); (R.N.C.)
| | - Francine Grodstein
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (S.K.); (F.G.)
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - James R. Mitchell
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland;
| | - Yangyu Liu
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (S.K.); (F.G.)
| |
Collapse
|
187
|
Tryptophan: From Diet to Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms22189904. [PMID: 34576067 PMCID: PMC8472285 DOI: 10.3390/ijms22189904] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/02/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the major causes of mortality worldwide. Inflammation is the underlying common mechanism involved in CVD. It has been recently related to amino acid metabolism, which acts as a critical regulator of innate and adaptive immune responses. Among different metabolites that have emerged as important regulators of immune and inflammatory responses, tryptophan (Trp) metabolites have been shown to play a pivotal role in CVD. Here, we provide an overview of the fundamental aspects of Trp metabolism and the interplay between the dysregulation of the main actors involved in Trp metabolism such as indoleamine 2, 3-dioxygenase 1 (IDO) and CVD, including atherosclerosis and myocardial infarction. IDO has a prominent and complex role. Its activity, impacting on several biological pathways, complicates our understanding of its function, particularly in CVD, where it is still under debate. The discrepancy of the observed IDO effects could be potentially explained by its specific cell and tissue contribution, encouraging further investigations regarding the role of this enzyme. Thus, improving our understanding of the function of Trp as well as its derived metabolites will help to move one step closer towards tailored therapies aiming to treat CVD.
Collapse
|
188
|
Jie Z, Liang S, Ding Q, Li F, Sun X, Lin Y, Chen P, Cai K, Zhou H, Lu H, Wang X, Zhang T, Xiao L, Yang H, Wang J, Hou Y, Kristiansen K, Jia H, Xu X. Disease trends in a young Chinese cohort according to fecal metagenome and plasma metabolites. MEDICINE IN MICROECOLOGY 2021. [DOI: 10.1016/j.medmic.2021.100037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
189
|
Abstract
Given the participation of the microbiome in human health and disease, understanding the context of host-microbe interactions involved in vascular pathophysiology is now evolving through identifying microbial communities, specific taxa, and metabolic profiling which can be coupled to human health outcomes. Exercise has been used to define mechanisms related to improved vascular health, which may involve the microbiome. Motivated by the clinical significance that both exercise and the gut microbiome have; the objective of our work is to assist in defining the gut-vascular axis while identifying biomarkers of gut microbial health linked to vascular function. In this commentary, we will provide context to the mechanistic perspectives of exercise-induced improvements in gut microbial characteristics coupled to vascular health outcomes and offer insight on necessary future prospective investigations.
Collapse
|
190
|
Perrelli A, Retta SF. Polymorphisms in genes related to oxidative stress and inflammation: Emerging links with the pathogenesis and severity of Cerebral Cavernous Malformation disease. Free Radic Biol Med 2021; 172:403-417. [PMID: 34175437 DOI: 10.1016/j.freeradbiomed.2021.06.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Cerebral Cavernous Malformation (CCM) is a cerebrovascular disease of genetic origin affecting 0.5% of the population and characterized by abnormally enlarged and leaky capillaries that predispose to seizures, neurological deficits, and intracerebral hemorrhage (ICH). CCM occurs sporadically or is inherited as dominant condition with incomplete penetrance and highly variable expressivity. Three disease genes have been identified: KRIT1 (CCM1), CCM2 and CCM3. Previous results demonstrated that loss-of-function mutations of CCM genes cause pleiotropic effects, including defective autophagy, altered reactive oxygen species (ROS) homeostasis, and enhanced sensitivity to oxidative stress and inflammatory events, suggesting a novel unifying pathogenetic mechanism, and raising the possibility that CCM disease onset and severity are influenced by the presence of susceptibility and modifier genes. Consistently, genome-wide association studies (GWAS) in large and homogeneous cohorts of patients sharing the familial form of CCM disease and identical mutations in CCM genes have led to the discovery of distinct genetic modifiers of major disease severity phenotypes, such as development of numerous and large CCM lesions, and susceptibility to ICH. This review deals with the identification of genetic modifiers with a significant impact on inter-individual variability in CCM disease onset and severity, including highly polymorphic genes involved in oxidative stress, inflammatory and immune responses, such as cytochrome P450 monooxygenases (CYP), matrix metalloproteinases (MMP), and Toll-like receptors (TLR), pointing to their emerging prognostic value, and opening up new perspectives for risk stratification and personalized medicine strategies.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy; CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy.
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy; CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy.
| |
Collapse
|
191
|
Manosso LM, Arent CO, Borba LA, Ceretta LB, Quevedo J, Réus GZ. Microbiota-Gut-Brain Communication in the SARS-CoV-2 Infection. Cells 2021; 10:1993. [PMID: 34440767 PMCID: PMC8391332 DOI: 10.3390/cells10081993] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease of 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome 2 (SARS-CoV-2). In addition to pneumonia, individuals affected by the disease have neurological symptoms. Indeed, SARS-CoV-2 has a neuroinvasive capacity. It is known that the infection caused by SARS-CoV-2 leads to a cytokine storm. An exacerbated inflammatory state can lead to the blood-brain barrier (BBB) damage as well as to intestinal dysbiosis. These changes, in turn, are associated with microglial activation and reactivity of astrocytes that can promote the degeneration of neurons and be associated with the development of psychiatric disorders and neurodegenerative diseases. Studies also have been shown that SARS-CoV-2 alters the composition and functional activity of the gut microbiota. The microbiota-gut-brain axis provides a bidirectional homeostatic communication pathway. Thus, this review focuses on studies that show the relationship between inflammation and the gut microbiota-brain axis in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Luana M. Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| | - Camila O. Arent
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| | - Laura A. Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| | - Luciane B. Ceretta
- Programa de Pós-Graduação em Saúde Coletiva, Universidade do Extremo Sul Catarinense, Criciúma 88806-000, SC, Brazil;
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA
| | - Gislaine Z. Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma 77054-000, SC, Brazil; (L.M.M.); (C.O.A.); (L.A.B.); (J.Q.)
| |
Collapse
|
192
|
The Role of Gut Microbiota on Cholesterol Metabolism in Atherosclerosis. Int J Mol Sci 2021; 22:ijms22158074. [PMID: 34360839 PMCID: PMC8347163 DOI: 10.3390/ijms22158074] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Hypercholesterolemia plays a causal role in the development of atherosclerosis and is one of the main risk factors for cardiovascular disease (CVD), the leading cause of death worldwide especially in developed countries. Current data show that the role of microbiota extends beyond digestion by being implicated in several metabolic and inflammatory processes linked to several diseases including CVD. Studies have reported associations between bacterial metabolites and hypercholesterolemia. However, such associations remain poorly investigated and characterized. In this review, the mechanisms of microbial derived metabolites such as primary and secondary bile acids (BAs), trimethylamine N-oxide (TMAO), and short-chain fatty acids (SCFAs) will be explored in the context of cholesterol metabolism. These metabolites play critical roles in maintaining cardiovascular health and if dysregulated can potentially contribute to CVD. They can be modulated via nutritional and pharmacological interventions such as statins, prebiotics, and probiotics. However, the mechanisms behind these interactions also remain unclear, and mechanistic insights into their impact will be provided. Therefore, the objectives of this paper are to present current knowledge on potential mechanisms whereby microbial metabolites regulate cholesterol homeostasis and to discuss the feasibility of modulating intestinal microbes and metabolites as a novel therapeutic for hypercholesterolemia.
Collapse
|
193
|
Tuganbaev T, Honda K. Non-zero-sum microbiome immune system interactions. Eur J Immunol 2021; 51:2120-2136. [PMID: 34242413 PMCID: PMC8457126 DOI: 10.1002/eji.202049065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/01/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
Fundamental asymmetries between the host and its microbiome in enzymatic activities and nutrient storage capabilities have promoted mutualistic adaptations on both sides. As a result, the enteric immune system has evolved so as not to cause a zero‐sum sterilization of non‐self, but rather achieve a non‐zero‐sum self‐reinforcing cooperation with its evolutionary partner the microbiome. In this review, we attempt to integrate the accumulated knowledge of immune—microbiome interactions into an evolutionary framework and trace the pattern of positive immune—microbiome feedback loops across epithelial, enteric nervous system, innate, and adaptive immune circuits. Indeed, the immune system requires commensal signals for its development and function, and reciprocally protects the microbiome from nutrient shortage and pathogen outgrowth. In turn, a healthy microbiome is the result of immune system curatorship as well as microbial ecology. The paradigms of host–microbiome asymmetry and the cooperative nature of their interactions identified in the gut are applicable across all tissues influenced by microbial activities. Incorporation of immune system influences into models of microbiome ecology will be a step forward toward defining what constitutes a healthy human microbiome and guide discoveries of novel host–microbiome mutualistic adaptations that may be harnessed for the promotion of human health.
Collapse
Affiliation(s)
- Timur Tuganbaev
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.,RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
194
|
Meng L, Song Z, Liu A, Dahmen U, Yang X, Fang H. Effects of Lipopolysaccharide-Binding Protein (LBP) Single Nucleotide Polymorphism (SNP) in Infections, Inflammatory Diseases, Metabolic Disorders and Cancers. Front Immunol 2021; 12:681810. [PMID: 34295331 PMCID: PMC8290185 DOI: 10.3389/fimmu.2021.681810] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/08/2021] [Indexed: 01/11/2023] Open
Abstract
Inflammation, which is induced by the immune response, is recognized as the driving factor in many diseases, including infections and inflammatory diseases, metabolic disorders and cancers. Genetic variations in pivotal genes associated with the immune response, particularly single nucleotide polymorphisms (SNPs), may account for predisposition and clinical outcome of diseases. Lipopolysaccharide (LPS)-binding protein (LBP) functions as an enhancer of the host response to LPS, the main component of the outer membrane of gram-native bacteria. Given the crucial role of LBP in inflammation, we will review the impact of SNPs in the LBP gene on infections and inflammatory diseases, metabolic disorders and cancers.
Collapse
Affiliation(s)
- Leilei Meng
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zichen Song
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Friedrich-Schiller-University Jena, Jena, Germany
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoshu Fang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
195
|
Pontes KSDS, Guedes MR, Cunha MRD, Mattos SDS, Barreto Silva MI, Neves MF, Marques BCAA, Klein MRST. Effects of probiotics on body adiposity and cardiovascular risk markers in individuals with overweight and obesity: A systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2021; 40:4915-4931. [PMID: 34358838 DOI: 10.1016/j.clnu.2021.06.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Evidence suggests that gut microbiota is a potential factor in the pathophysiology of both obesity and related metabolic disorders. While individual randomized controlled trials (RCTs) have evaluated the effects of probiotics on adiposity and cardiovascular disease (CVD) risk factors in subjects with overweight and obesity, the results are inconsistent. Thus, this systematic review and meta-analysis aimed to evaluate the effects of probiotic supplementation on body weight, body adiposity and CVD risk markers in overweight and obese subjects. METHODS A systematic search for RCTs published up to December 2020 was conducted in MEDLINE (via PubMed), EMBASE, Scopus and LILACS. Meta-analysis using a random-effects model was chosen to analyze the impact of combined trials. RESULTS Twenty-six RCTs (n = 1720) were included. Data pooling showed a significant effect of probiotics in reducing body weight (MD:-0.70 kg; 95%CI:-1.04,-0.35 kg; P < 0.0001), body mass index (BMI) (MD:-0.24 kg/m2; 95%CI:-0.35,-0.12 kg/m2; P = 0.0001), waist circumference (WC) (MD:-1.13 cm; 95%CI:-1.54,-0.73 cm; P < 0.0001), fat mass (MD:-0.71 kg; 95%CI:-1.10,-0.32 kg; P = 0.0004), tumor necrosis factor-α (MD:-0.16 pg/ml; 95%CI:-0.24,-0.08 pg/ml; P = 0.0001), insulin (MD:-0.85mcU/ml; 95%CI:-1.50,-0.21mcU/ml; P = 0.010), total cholesterol (MD:-0.16 mmol/l; 95%CI:-0.26,-0.05 mmol/l; P = 0.003) and LDL (MD:-0.09 mmol/l; 95%CI:-0.16,-0.03 mmol/l; P = 0.006) compared with control groups. There was a significant decrease in body weight, BMI and WC in studies using both single and multi-bacterial species. Decreases in body adiposity parameters were only observed in studies using a probiotic dose of ≥ 1010 CFU and for ≥8 weeks duration. CONCLUSIONS The present meta-analysis suggests that probiotics consumption may be helpful for improving body weight, body adiposity and some CVD risk markers in individuals with overweight and obesity. The review was registered on PROSPERO (International prospective register of systematic reviews): CRD42020183136.
Collapse
Affiliation(s)
- Karine Scanci da Silva Pontes
- Post-Graduation Program in Clinical and Experimental Pathophysiology, State University of Rio de Janeiro (UERJ), Av. Professor Manuel de Abreu, 444, Térreo - Rio de Janeiro, RJ, 20550-170, Brazil.
| | - Marcella Rodrigues Guedes
- Post-Graduation Program in Clinical and Experimental Pathophysiology, State University of Rio de Janeiro (UERJ), Av. Professor Manuel de Abreu, 444, Térreo - Rio de Janeiro, RJ, 20550-170, Brazil.
| | - Michelle Rabello da Cunha
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil.
| | - Samanta de Souza Mattos
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil.
| | - Maria Inês Barreto Silva
- Department of Applied Nutrition, Nutrition Institute, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524 - Pavilhão João Lyra Filho, 12º Andar, Bloco D, Rio de Janeiro, RJ, 20559-900, Brazil; Department of Applied Nutrition, Nutrition School, Federal University of the State of Rio de Janeiro (UNIRIO), Av. Pasteur, 296, Botafogo, 3º Andar, Rio de Janeiro, RJ, 22290-250, Brazil.
| | - Mario Fritsch Neves
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil.
| | - Bianca Cristina Antunes Alves Marques
- Department of Clinical Medicine, State University of Rio de Janeiro (UERJ), Av.Vinte e Oito de Setembro, 77 Sala 329, Rio de Janeiro, RJ, 20551-030, Brazil; Department of Nutrition and Dietetics, National Cancer Institute (INCA), Av. Binário do Porto, 831, Rio de Janeiro, RJ, 20081-250, Brazil.
| | - Márcia Regina Simas Torres Klein
- Department of Applied Nutrition, Nutrition Institute, State University of Rio de Janeiro (UERJ), Rua São Francisco Xavier, 524 - Pavilhão João Lyra Filho, 12º Andar, Bloco D, Rio de Janeiro, RJ, 20559-900, Brazil.
| |
Collapse
|
196
|
Matsuoka T, Shimizu T, Minagawa T, Hiranuma W, Takeda M, Kakuta R, Kawamoto S. First case of an invasive Bacteroides dorei infection detected in a patient with a mycotic aortic aneurysm-raising a rebellion of major indigenous bacteria in humans: a case report and review. BMC Infect Dis 2021; 21:625. [PMID: 34193073 PMCID: PMC8247135 DOI: 10.1186/s12879-021-06345-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Background Bacteroides dorei is an anaerobic gram-negative bacterium first described in 2006. Because of the high similarity in mass spectra between B. dorei and Bacteroides vulgatus, discriminating between these species is arduous in clinical practice. In recent decades, 16S rRNA gene sequencing has been a complementary method for distinguishing taxonomically close bacteria, including B. dorei and B. vulgatus, at the genus and species levels. Consequently, B. dorei has been shown to contribute to some diseases, including type 1 autoimmune diabetes mellitus and atherosclerotic diseases. However, there are no reports on invasive infectious diseases caused by B. dorei. This report describes the first case of direct invasion and colonisation of human tissue by B. dorei, thus providing a warning regarding the previously proposed application of B. dorei as a live biotherapeutic for atherosclerotic diseases. Case presentation A 78-year-old Japanese man complained of intermittent chest/back pain and was diagnosed with a mycotic thoracic aortic aneurysm by enhanced computed tomography on admission. Despite strict blood pressure control and empirical antibiotic therapy, the patient’s condition worsened. To prevent aneurysmal rupture and eliminate infectious foci, the patient underwent surgical treatment. The resected specimen was subjected to tissue culture and 16S rRNA gene sequencing analysis to identify pathogenic bacteria. A few days after the surgery, culture and sequencing results revealed that the pathogen was B. dorei/B. vulgatus and B. dorei, respectively. The patient was successfully treated with appropriate antibacterial therapy and after improvement, was transferred to another hospital for rehabilitation on postoperative day 34. There was no recurrence of infection or aneurysm after the patient transfer. Conclusions This report describes the first case of invasive infectious disease caused by B. dorei, casting a shadow over its utilisation as a probiotic for atherosclerotic diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06345-8.
Collapse
Affiliation(s)
- Takayuki Matsuoka
- Department of Cardiovascular Surgery, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino Ward, Sendai, Miyagi, 983-8512, Japan.
| | - Takuya Shimizu
- Department of Cardiovascular Surgery, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino Ward, Sendai, Miyagi, 983-8512, Japan
| | - Tadanori Minagawa
- Department of Cardiovascular Surgery, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino Ward, Sendai, Miyagi, 983-8512, Japan
| | - Wakiko Hiranuma
- Department of Cardiovascular Surgery, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino Ward, Sendai, Miyagi, 983-8512, Japan
| | - Miki Takeda
- Department of Cardiovascular Surgery, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino Ward, Sendai, Miyagi, 983-8512, Japan
| | - Risako Kakuta
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shunsuke Kawamoto
- Department of Cardiovascular Surgery, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino Ward, Sendai, Miyagi, 983-8512, Japan
| |
Collapse
|
197
|
Song W, Sun LY, Zhu ZJ, Wei L, Qu W, Zeng ZG, Yang YS. Characteristics of Gut Microbiota in Children With Biliary Atresia After Liver Transplantation. Front Physiol 2021; 12:704313. [PMID: 34262484 PMCID: PMC8273867 DOI: 10.3389/fphys.2021.704313] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Background and Aims Biliary atresia (BA) is an idiopathic neonatal cholestasis and is the most common indication in pediatric liver transplantation (LT). Previous studies have suggested that the gut microbiota (GM) in BA is disordered. However, the effect of LT on gut dysbiosis in patients with BA has not yet been elucidated. Methods Patients with BA (n = 16) and healthy controls (n = 10) were recruited. In the early life of children with BA, Kasai surgery is a typical procedure for restoring bile flow. According to whether BA patients had previously undergone Kasai surgery, we divided the post-LT patients into the with-Kasai group (n = 8) and non-Kasai group (n = 8). Fecal samples were collected in both the BA and the control group; among BA patients, samples were obtained again 6 months after LT. A total of 40 fecal samples were collected, of which 16 were pre-LT, 14 were post-LT (8 were with-Kasai, 6 were non-Kasai), and 10 were from the control group. Metagenomic sequencing was performed to evaluate the GM. Results The Kruskal-Wallis test showed a statistically significant difference in the number of genes between the pre-LT and the control group, the pre-LT and the post-LT group (P < 0.05), but no statistical difference between the post-LT and the control group. Principal coordinate analysis also showed that the microbiome structure was similar between the post-LT and control group (P > 0.05). Analysis of the GM composition showed a significant decrease in Serratia, Enterobacter, Morganella, Skunalikevirus, and Phifllikevirus while short chain fatty acid (SCFA)-producing bacteria such as Roseburia, Blautia, Clostridium, Akkermansia, and Ruminococcus were increased after LT (linear discriminant analysis > 2, P < 0.05). However, they still did not reach the normal control level. Concerning functional profiles, lipopolysaccharide metabolism, multidrug resistance, polyamine biosynthesis, GABA biosynthesis, and EHEC/EPEC pathogenicity signature were more enriched in the post-LT group compared with the control group. Prior Kasai surgery had a specific influence on the postoperative GM. Conclusion LT partly improved the GM in patients with BA, which provided new insight into understanding the role of LT in BA.
Collapse
Affiliation(s)
- Wei Song
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Li-Ying Sun
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China.,Department of Intensive Care Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhi-Jun Zhu
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Lin Wei
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Wei Qu
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Zhi-Gui Zeng
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Yun-Sheng Yang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
198
|
Ciaravolo S, Martínez-López LM, Allcock RJN, Woodward AP, Mansfield C. Longitudinal Survey of Fecal Microbiota in Healthy Dogs Administered a Commercial Probiotic. Front Vet Sci 2021; 8:664318. [PMID: 34235200 PMCID: PMC8255976 DOI: 10.3389/fvets.2021.664318] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this longitudinal microbiome study was to investigate the effects of a commercially available veterinary synbiotic product (Blackmore's® Paw DigestiCare 60™) on the fecal microbiome of healthy dogs using 16S rRNA gene microbial profiling. Fifteen healthy, privately-owned dogs participated in a 2-week trial administration of the product. Fecal samples were collected at different time points, including baseline (prior to treatment), during administration and after discontinuation of product. Large intra- and inter-individual variation was observed throughout the study, but microbiome composition at higher phylogenetic levels, alpha and beta diversity were not significantly altered after 2 weeks of probiotic administration, suggesting an absence of probiotic impact on microbial diversity. Administration of the synbiotic preparation did, however, result in transient increases in probiotic species from Enterococacceae and Streptococacceae families as well as an increase in Fusobacteria; with the fecal microbiota partially reverting to its baseline state 3-weeks after cessation of probiotic administration.
Collapse
Affiliation(s)
- Susan Ciaravolo
- Department of Veterinary Clinical Sciences, Melbourne Veterinary School, The University of Melbourne, Werribee, VIC, Australia.,Peninsula Vet, Emergency and Referral Hospital, Mornington, VIC, Australia
| | - Lina María Martínez-López
- Department of Veterinary Clinical Sciences, Melbourne Veterinary School, The University of Melbourne, Werribee, VIC, Australia
| | - Richard J N Allcock
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Andrew P Woodward
- Department of Veterinary Clinical Sciences, Melbourne Veterinary School, The University of Melbourne, Werribee, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Werribee, VIC, Australia
| | - Caroline Mansfield
- Department of Veterinary Clinical Sciences, Melbourne Veterinary School, The University of Melbourne, Werribee, VIC, Australia
| |
Collapse
|
199
|
Silva CBP, Elias-Oliveira J, McCarthy CG, Wenceslau CF, Carlos D, Tostes RC. Ethanol: striking the cardiovascular system by harming the gut microbiota. Am J Physiol Heart Circ Physiol 2021; 321:H275-H291. [PMID: 34142885 DOI: 10.1152/ajpheart.00225.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ethanol consumption represents a significant public health problem, and excessive ethanol intake is a risk factor for cardiovascular disease (CVD), one of the leading causes of death and disability worldwide. The mechanisms underlying the effects of ethanol on the cardiovascular system are complex and not fully comprehended. The gut microbiota and their metabolites are indispensable symbionts essential for health and homeostasis and therefore, have emerged as potential contributors to ethanol-induced cardiovascular system dysfunction. By mechanisms that are not completely understood, the gut microbiota modulates the immune system and activates several signaling pathways that stimulate inflammatory responses, which in turn, contribute to the development and progression of CVD. This review summarizes preclinical and clinical evidence on the effects of ethanol in the gut microbiota and discusses the mechanisms by which ethanol-induced gut dysbiosis leads to the activation of the immune system and cardiovascular dysfunction. The cross talk between ethanol consumption and the gut microbiota and its implications are detailed. In summary, an imbalance in the symbiotic relationship between the host and the commensal microbiota in a holobiont, as seen with ethanol consumption, may contribute to CVD. Therefore, manipulating the gut microbiota, by using antibiotics, probiotics, prebiotics, and fecal microbiota transplantation might prove a valuable opportunity to prevent/mitigate the deleterious effects of ethanol and improve cardiovascular health and risk prevention.
Collapse
Affiliation(s)
- Carla B P Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jefferson Elias-Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Camilla F Wenceslau
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
200
|
In Vitro Fecal Fermentation Patterns of Arabinoxylan from Rice Bran on Fecal Microbiota from Normal-Weight and Overweight/Obese Subjects. Nutrients 2021; 13:nu13062052. [PMID: 34203983 PMCID: PMC8232586 DOI: 10.3390/nu13062052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/01/2021] [Accepted: 06/10/2021] [Indexed: 01/01/2023] Open
Abstract
Arabinoxylan (AX) is a structural polysaccharide found in wheat, rice and other cereal grains. Diets high in AX-containing fiber may promote gut health in obesity through prebiotic function. Thus, the impact of soluble AX isolated from rice bran fiber on human gut microbiota phylogenetic composition and short-chain fatty acid (SCFA) production patterns from normal-weight and overweight/obese subjects was investigated through in vitro fecal fermentation. Results showed that rice bran arabinoxylan modified the microbiota in fecal samples from both weight classes compared to control, significantly increasing Collinsella, Blautia and Bifidobacterium, and decreasing Sutterella, Bilophila and Parabacteroides. Rice bran AX also significantly increased total and individual SCFA contents (p < 0.05). This study suggests that rice bran AX may beneficially impact gut health in obesity through prebiotic activities.
Collapse
|