501
|
Zhong HJ, Yuan Y, Xie WR, Chen MH, He XX. Type 2 Diabetes Mellitus Is Associated with More Serious Small Intestinal Mucosal Injuries. PLoS One 2016; 11:e0162354. [PMID: 27598308 PMCID: PMC5012602 DOI: 10.1371/journal.pone.0162354] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/29/2016] [Accepted: 08/22/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Clinical and experimental research has revealed that diabetes mellitus (DM) is characterized by intestinal hypomotility, gut microbial dysbiosis, increased gut permeability, microcirculation disorders, circulatory changes, and dysfunction of intestinal stem cells, which may be linked to inflammation of intestinal mucosa. However, the relationship between type 2 DM (T2DM) and macroscopic small intestinal mucosal injuries is still unclear. Therefore, we retrospectively studied capsule endoscopy data to determine the relationship between T2DM and small intestinal mucosal injuries. MATERIALS AND METHODS We compared the records of 38 T2DM patients with those of 152 non-DM patients for small intestinal mucosal injuries. Different types of mucosal injuries and Lewis scores were compared between T2DM and non-DM patients. The relationships between patients with or without different types of diabetic complications and the Lewis score was assessed. Moreover, the relationships between insulin resistance and Lewis score, between HbA1c and Lewis score, were also both assessed. RESULTS The prevalence of a villous edema in subjects with T2DM was significantly higher than in those without DM (P < 0.001), but incidence of ulcers was not different (P = 1.000). With T2DM, the Lewis score was also significantly higher (P = 0.002). In addition, subjects with diabetic nephropathy showed significantly higher Lewis scores than patients without diabetic nephropathy (P = 0.033). In Pearson's correlation tests, the homeostasis model assessment of insulin resistance (HOMA-IR) value was correlated positively with the Lewis score (γ = 0.175, P = 0.015), but no statistical correlation was found between HbA1c level and Lewis score (γ = 0.039, P = 0.697). CONCLUSIONS Subjects with T2DM, especially those with diabetic nephropathy, have higher Lewis scores and more serious small intestinal mucosal lesions.
Collapse
Affiliation(s)
- Hao-Jie Zhong
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yu Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Wen-Rui Xie
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Mei-Hui Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xing-Xiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- * E-mail:
| |
Collapse
|
502
|
Jensen AB, Ajslev TA, Brunak S, Sørensen TIA. Long-term risk of cardiovascular and cerebrovascular disease after removal of the colonic microbiota by colectomy: a cohort study based on the Danish National Patient Register from 1996 to 2014. BMJ Open 2015; 5:e008702. [PMID: 26671951 PMCID: PMC4680005 DOI: 10.1136/bmjopen-2015-008702] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/07/2015] [Revised: 10/13/2015] [Accepted: 10/26/2015] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES The hypothesis of the study was that if the gut microbiota is involved in the development of atherosclerotic cardiovascular and cerebrovascular diseases (CVDs), total colectomy may reduce the long-term risk of CVDs. The aim was therefore to investigate the risk of CVD in patients after a total colectomy compared with patients undergoing other types of surgery, which are not expected to alter the gut microbiota or the CVD risk. SETTING The Danish National Patient Register including all hospital discharges in Denmark from 1996 to 2014. PARTICIPANTS Patients (n=1530) aged 45 years and above and surviving 1000 days after total colectomy without CVDs were selected and matched with five control patients who were also free of CVD 1000 days after other types of surgery. The five control patients were randomly selected from each of the three surgical groups: orthopaedic surgery, surgery in the gastrointestinal tract leaving it intact and other surgeries not related to the gastrointestinal tract or CVD (n=22 950). PRIMARY AND SECONDARY OUTCOME MEASURES The primary outcome was the first occurring CVD event in any of the seven diagnostic domains (hypertensive disorders, acute ischaemic heart diseases, chronic ischaemic heart disease, cardiac arrhythmias, heart failure, cerebrovascular diseases and other arterial diseases) and the secondary outcomes were the first occurring event within each of these domains. RESULTS Estimated by Cox proportional hazard models, the HRs of the composite CVD end point for patients with colectomy compared with the control patients were not significantly reduced (HR=0.94, 95% confidence limits 0.85 to 1.04). Among the seven CVD domains, only the risk of hypertensive disorders was significantly reduced (HR=0.85, 0.73 to 0.98). CONCLUSIONS Colectomy did not reduce the general risk of CVD, but reduced the risk of hypertensive disorders, most likely due to salt and water depletion induced by colectomy. These results encourage a reappraisal of the associations between gut microbiota and CVD.
Collapse
Affiliation(s)
- Anders Boeck Jensen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Teresa Adeltoft Ajslev
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| | - Søren Brunak
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark Department of Systems Biology, Center for Biological Sequence Analyses, Technical University of Denmark, Lyngby, Denmark
| | - Thorkild I A Sørensen
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark MRC Integrative Epidemiology Unit, Bristol University, Bristol, UK
| |
Collapse
|
503
|
Dunlop AL, Mulle JG, Ferranti EP, Edwards S, Dunn AB, Corwin EJ. Maternal Microbiome and Pregnancy Outcomes That Impact Infant Health: A Review. Adv Neonatal Care 2015; 15:377-85. [PMID: 26317856 PMCID: PMC4658310 DOI: 10.1097/anc.0000000000000218] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/09/2023]
Abstract
The maternal microbiome is recognized as a key determinant of a range of important maternal and child health outcomes, and together with perinatal factors influences the infant microbiome. This article provides a summary review of research investigating (1) the role of the maternal microbiome in pregnancy outcomes known to adversely influence neonatal and infant health, including preterm birth, cardiometabolic complications of pregnancy such as preeclampsia and gestational diabetes, and excessive gestational weight gain; (2) factors with an established link to adverse pregnancy outcomes that are known to influence the composition of the maternal microbiome; and (3) strategies for promoting a healthy maternal microbiome, recognizing that much more research is needed in this area.
Collapse
Affiliation(s)
- Anne L. Dunlop
- Emory University School of Nursing, 1520 Clifton Road NE, Atlanta, GA 30322, 404-712-8520 (phone); 404-727-6945 (fax)
| | - Jennifer G. Mulle
- Rollins School of Public Health & School of Medicine, Department of Human Genetics, Emory University Atlanta, GA 30322
| | - Erin P. Ferranti
- Nell Hodgson Woodruff School of Nursing, Emory University,Atlanta, GA 30322
| | - Sara Edwards
- Nell Hodgson Woodruff School of Nursing, Emory University,Atlanta, GA 30322
| | - Alexis B. Dunn
- Nell Hodgson Woodruff School of Nursing, Emory University,Atlanta, GA 30322
| | | |
Collapse
|
504
|
Plant polyphenols bioavailability and modulation of the gut microbiota consortium: a paradigm shift in understanding their effects on diseases. ACTA ACUST UNITED AC 2015. [DOI: 10.17660/actahortic.2015.1106.31] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022]
|
505
|
Nutritional Modulation of Non-Alcoholic Fatty Liver Disease and Insulin Resistance. Nutrients 2015; 7:9127-38. [PMID: 26556368 PMCID: PMC4663582 DOI: 10.3390/nu7115454] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/17/2015] [Revised: 10/07/2015] [Accepted: 10/09/2015] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) covers a spectrum of disorders ranging from simple steatosis (non-alcoholic fatty liver, NAFL) to non-alcoholic steatohepatitis (NASH) and cirrhosis. NAFL increases the risk of liver fibrosis. If the liver is fatty due to causes of insulin resistance such as obesity and physical inactivity, it overproduces glucose and triglycerides leading to hyperinsulinemia and a low high-density lipoprotein (HDL) cholesterol concentration. The latter features predispose to type 2 diabetes and cardiovascular disease (CVD). Understanding the impact of nutritional modulation of liver fat content and insulin resistance is therefore of interest for prevention and treatment of NAFLD. Hypocaloric, especially low carbohydrate ketogenic diets rapidly decrease liver fat content and associated metabolic abnormalities. However, any type of caloric restriction seems effective long-term. Isocaloric diets containing 16%-23% fat and 57%-65% carbohydrate lower liver fat compared to diets with 43%-55% fat and 27%-38% carbohydrate. Diets rich in saturated (SFA) as compared to monounsaturated (MUFA) or polyunsaturated (PUFA) fatty acids appear particularly harmful as they increase both liver fat and insulin resistance. Overfeeding either saturated fat or carbohydrate increases liver fat content. Vitamin E supplementation decreases liver fat content as well as fibrosis but has no effect on features of insulin resistance.
Collapse
|
506
|
Ungefroren H, Gieseler F, Fliedner S, Lehnert H. Obesity and cancer. Horm Mol Biol Clin Investig 2015; 21:5-15. [PMID: 25719336 DOI: 10.1515/hmbci-2014-0046] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/23/2014] [Accepted: 01/14/2015] [Indexed: 11/15/2022]
Abstract
The incidence of obesity in the Western world has increased dramatically during the recent decades. Epidemiological data suggest that obesity is associated with an increased risk of several but not all types of cancers, with clear sex-specific differences. The underlying mechanisms are still a matter of debate. In this review, we discuss the potential factors linking obesity to cancer, with a focus on hormone-dependent cancer types. Current experimental evidence suggests that insulin resistance and a chronic, subclinical inflammation in the visceral fat are the major metabolic events, causing alterations in the levels of insulin, glucose, free fatty acids, insulin-like growth factor-1 and -2, adipose tissue-derived proinflammatory factors (tumor necrosis factor-α and interleukin-1, -6, -12, and -23), and other bioactive molecules such as adipokines (adiponectin and leptin), vascular endothelial growth factor, sex hormones, gut microbiota, and secondary bile acids. All these factors may act directly or indirectly on the tumor microenvironment to drive tumor progression via the stimulation of cell survival/antiapoptosis, cell proliferation, angiogenesis, and invasion/metastasis of cancer cells. Therapeutic strategies (including bariatric surgery) that target dysfunctional or inflamed fat have been shown to benefit patients, whereas other cell- or hormone-directed interventions (such as the conversion of visceral fat macrophages to an anti-inflammatory M2 phenotype or the pharmacological modulation of serum adipokine levels) are still theoretical and need to be clinically evaluated for their ability to successfully treat or prevent obesity-related cancers.
Collapse
|
507
|
Qian LL, Li HT, Zhang L, Fang QC, Jia WP. Effect of the Gut Microbiota on Obesity and Its Underlying Mechanisms: an Update. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2015; 28:839-847. [PMID: 26695364 DOI: 10.1016/s0895-3988(15)30116-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/23/2015] [Accepted: 10/27/2015] [Indexed: 01/05/2025]
Affiliation(s)
- Ling Ling Qian
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China; Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hua Ting Li
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China
| | - Lei Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China; Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qi Chen Fang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China
| | - Wei Ping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China; Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
508
|
Ahn SY, Sohn SH, Lee SY, Park HL, Park YW, Kim H, Nam JH. The effect of lipopolysaccharide-induced obesity and its chronic inflammation on influenza virus-related pathology. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:924-930. [PMID: 26509733 DOI: 10.1016/j.etap.2015.09.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/01/2015] [Revised: 09/25/2015] [Accepted: 09/27/2015] [Indexed: 06/05/2023]
Abstract
Obese individuals show increased susceptibility to infection, low vaccine efficacy, and worse pathophysiology. However, it is unclear how obesity affects these events. The aim of this study was to investigate the effect of obesity-triggered chronic inflammation on immune cells after influenza virus infection. Control and lipopolysaccharide mice, in which an osmotic pump continually released Tween saline or lipopolysaccharide, were prepared and 3 weeks later were infected with pandemic H1N1 2009 influenza A virus. In lipopolysaccharide mice, we found a reduction in macrophage activation markers in the steady state, and reduced production of pro-inflammatory cytokines including tumor necrosis factor-α, interleukin-1β, and interleukin-6, in restimulated peritoneal macrophages. Interestingly, lipopolysaccharide-triggered chronic inflammation exacerbated the severity of pathological symptoms in the lungs after challenge with influenza virus. Taken together, the increased severity of virus-induced symptoms in obese individuals with chronic inflammation may be, at least partially, caused by macrophage dysfunction.
Collapse
Affiliation(s)
- Sun-Young Ahn
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, South Korea
| | - Sung-Hwa Sohn
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, South Korea
| | - Sang-Yeon Lee
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, South Korea
| | - Hye-Lim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, South Korea
| | - Yong-Wook Park
- SK Chemical, Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Hun Kim
- SK Chemical, Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, South Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, South Korea.
| |
Collapse
|
509
|
Palau-Rodriguez M, Tulipani S, Isabel Queipo-Ortuño M, Urpi-Sarda M, Tinahones FJ, Andres-Lacueva C. Metabolomic insights into the intricate gut microbial-host interaction in the development of obesity and type 2 diabetes. Front Microbiol 2015; 6:1151. [PMID: 26579078 PMCID: PMC4621279 DOI: 10.3389/fmicb.2015.01151] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
Gut microbiota has recently been proposed as a crucial environmental factor in the development of metabolic diseases such as obesity and type 2 diabetes, mainly due to its contribution in the modulation of several processes including host energy metabolism, gut epithelial permeability, gut peptide hormone secretion, and host inflammatory state. Since the symbiotic interaction between the gut microbiota and the host is essentially reflected in specific metabolic signatures, much expectation is placed on the application of metabolomic approaches to unveil the key mechanisms linking the gut microbiota composition and activity with disease development. The present review aims to summarize the gut microbial-host co-metabolites identified so far by targeted and untargeted metabolomic studies in humans, in association with impaired glucose homeostasis and/or obesity. An alteration of the co-metabolism of bile acids, branched fatty acids, choline, vitamins (i.e., niacin), purines, and phenolic compounds has been associated so far with the obese or diabese phenotype, in respect to healthy controls. Furthermore, anti-diabetic treatments such as metformin and sulfonylurea have been observed to modulate the gut microbiota or at least their metabolic profiles, thereby potentially affecting insulin resistance through indirect mechanisms still unknown. Despite the scarcity of the metabolomic studies currently available on the microbial-host crosstalk, the data-driven results largely confirmed findings independently obtained from in vitro and animal model studies, putting forward the mechanisms underlying the implication of a dysfunctional gut microbiota in the development of metabolic disorders.
Collapse
Affiliation(s)
- Magali Palau-Rodriguez
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Sara Tulipani
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain ; Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain
| | - Maria Isabel Queipo-Ortuño
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Mireia Urpi-Sarda
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Francisco J Tinahones
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| |
Collapse
|
510
|
Chassaing B, Miles-Brown J, Pellizzon M, Ulman E, Ricci M, Zhang L, Patterson AD, Vijay-Kumar M, Gewirtz AT. Lack of soluble fiber drives diet-induced adiposity in mice. Am J Physiol Gastrointest Liver Physiol 2015; 309:G528-41. [PMID: 26185332 PMCID: PMC4593822 DOI: 10.1152/ajpgi.00172.2015] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/28/2015] [Accepted: 07/08/2015] [Indexed: 01/31/2023]
Abstract
Diet-induced obesity is often modeled by comparing mice fed high-fat diet (HFD), which is made from purified ingredients, vs. normal chow diet (NCD), which is a low-fat assemblage of relatively unrefined plant and animal products. The mechanism by which HFD promotes adiposity is complex but thought to involve low-grade inflammation and altered gut microbiota. The goal of this study was to investigate the extent to which HFD-induced adiposity is driven by fat content vs. other factors that differentiate HFD vs. NCD. Mice were fed NCD, HFD, or other compositionally defined diets (CDD), designed to mimic NCD and/or explore the role of HFD components. A range of metabolic parameters reflecting low-grade inflammation and adiposity were assayed. Relative to NCD, HFD, and to a lesser, but, nonetheless, significant extent, CDD induced increased adiposity, indicating both lipid content and other aspects of HFD are obesogenic. Moreover, HFD and CDD induced a rapid and marked loss of cecal and colonic mass. Such CDD-induced effects were not affected by adjusting dietary protein levels/types but could be largely eliminated by exchanging insoluble fiber (cellulose) for soluble fiber (inulin). Replacing cellulose with inulin in HFD also protected mice against decreased intestinal mass, hyperphagia, and increased adiposity. Such beneficial effects of inulin were microbiota dependent, correlated with elevated fecal short-chain fatty acid levels analyzed via (1)H-NMR-based metabolomics and were partially recapitulated by administration of short-chain fatty acid. HFD-induced obesity is strongly promoted by its lack of soluble fiber, which supports microbiota-mediated intestinal tissue homeostasis that prevents inflammation driving obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Benoit Chassaing
- 1Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia;
| | - Jennifer Miles-Brown
- 1Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia;
| | | | - Edward Ulman
- 2Research Diets, Inc., New Brunswick, New Jersey;
| | | | - Limin Zhang
- 3Departments of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania; and
| | - Andrew D. Patterson
- 3Departments of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania; and
| | - Matam Vijay-Kumar
- 4Departments of Nutritional Sciences & Medicine, Pennsylvania State University, University Park, Pennsylvania
| | - Andrew T. Gewirtz
- 1Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia;
| |
Collapse
|
511
|
Shin JH, Lee JS, Seo JG. Assessment of cell adhesion, cell surface hydrophobicity, autoaggregation, and lipopolysaccharide-binding properties of live and heat-killed Lactobacillus acidophilus CBT LA1. ACTA ACUST UNITED AC 2015. [DOI: 10.7845/kjm.2015.5029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/05/2022]
|
512
|
Balasubramanian H, Patole S. Early probiotics to prevent childhood metabolic syndrome: A systematic review. World J Methodol 2015; 5:157-163. [PMID: 26413489 PMCID: PMC4572029 DOI: 10.5662/wjm.v5.i3.157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/27/2015] [Revised: 04/05/2015] [Accepted: 06/19/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To conduct a systematic review of studies on early probiotic supplementation to prevent childhood metabolic syndrome (MS).
METHODS: Using the Cochrane systematic review strategy we searched PubMed, EMBASE, CENTRAL, CINAHL, and the conference proceedings of the Pediatric American Society meetings and trial registries in December 2014. Randomised controlled trials (RCTs) and non RCTs of probiotic supplementation to the mother and/or infant for a minimum duration of 4 wk were selected. Of these, studies that reported on MS or its components (obesity, raised blood pressure, hyperglycemia, dyslipidemia) in children between 2-19 years were to be eligible for inclusion in the review. Risk of bias (ROB) in selected RCTs and quality assessment of non-RCT studies were to be assessed by the Cochrane ROB assessment table and New Castle Ottawa scale.
RESULTS: There were no studies on early probiotic administration for prevention of childhood MS (CMS). Follow up studies of two placebo controlled RCTs (n = 233) reported on the effects of early probiotics on one or more components of MS in children aged 2-19 years. Meta-analysis of those two studies could not be performed due to differences in the patient population, type of outcomes studied and the timing of their assessment. Assessment of childhood metabolic outcomes was not the primary objective of these studies. The first study that assessed the effects of prenatal and postnatal supplementation of Lactobacillus rhamnosus GG on body mass index till 10 years, did not report a significant benefit. In the second study, Lactobacillus paracasei 19 was supplemented to healthy term infants from 4-13 mo. No significant effect on body mass index, body composition or metabolic markers was detected.
CONCLUSION: Current evidence on early probiotic administration to prevent CMS is inadequate. Gaps in knowledge need to be addressed before large RCTs can be planned.
Collapse
|
513
|
Han SJ, Ha KH, Jeon JY, Kim HJ, Lee KW, Kim DJ. Impact of Cadmium Exposure on the Association between Lipopolysaccharide and Metabolic Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:11396-409. [PMID: 26378560 PMCID: PMC4586682 DOI: 10.3390/ijerph120911396] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Academic Contribution Register] [Received: 07/30/2015] [Revised: 08/21/2015] [Accepted: 09/06/2015] [Indexed: 02/06/2023]
Abstract
Cadmium (Cd) is an environmental contaminant that has a direct impact on the gut microbiome. Perturbations in the gut microbiome have been linked to metabolic disorders associated with inflammation generated by lipopolysaccharide (LPS). We investigated the impact of Cd on the association between LPS and metabolic syndrome. The study population consisted of 200 apparently healthy subjects (30–64 years of age; 96 men, 104 women). Serum LPS and blood Cd concentrations were measured by ELISA and graphite furnace-atomic absorption spectrophotometry (GF-AAS), respectively. The highest LPS quartile was associated with a greater prevalence of metabolic syndrome in men. There was a significant association between LPS activity and metabolic syndrome in men with blood Cd concentrations higher than the 50th percentile (OR = 3.05, 95% CI = 1.39–6.70); however, this relationship was not significant in men with blood Cd concentrations lower than the 50th percentile. The results of this study provide evidence for a strong association between high LPS activity and the prevalence of metabolic syndrome in men with relatively high blood Cd concentrations. Therefore, exposure to Cd may potentiate the association between LPS and metabolic syndrome in men.
Collapse
Affiliation(s)
- Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
| | - Kyoung Hwa Ha
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
- Cardiovascular and Metabolic Disease Etiology Research Center, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
| | - Ja Young Jeon
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
| | - Hae Jin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
| | - Kwan Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
- Cardiovascular and Metabolic Disease Etiology Research Center, Ajou University School of Medicine, 164, World Cup-ro, Yeongtong-gu, Suwon 443-380, Korea.
| |
Collapse
|
514
|
Gómez-Guzmán M, Toral M, Romero M, Jiménez R, Galindo P, Sánchez M, Zarzuelo MJ, Olivares M, Gálvez J, Duarte J. Antihypertensive effects of probioticsLactobacillusstrains in spontaneously hypertensive rats. Mol Nutr Food Res 2015; 59:2326-36. [DOI: 10.1002/mnfr.201500290] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/15/2015] [Revised: 07/21/2015] [Accepted: 07/29/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Manuel Gómez-Guzmán
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
| | - Marta Toral
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
| | - Miguel Romero
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA); Granada Spain
| | - Rosario Jiménez
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA); Granada Spain
| | - Pilar Galindo
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
| | - Manuel Sánchez
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
| | - María José Zarzuelo
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
| | - Mónica Olivares
- Laboratorio de Descubrimiento y Preclínica; Departamento de Investigación BIOSEARCH S.A; Granada Spain
| | - Julio Gálvez
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA); Granada Spain
- CIBER-ehd; Departamento de Farmacología; CIBM; Universidad de Granada; Armilla (Granada) Spain
| | - Juan Duarte
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Granada; Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA); Granada Spain
| |
Collapse
|
515
|
Zhang C, Li Y, Wang L, Sun S, Liu G, Leng J, Guo J, Lv L, Li W, Zhang C, Hu G, Yu Z, Yang X. Blood group AB is protective factor for gestational diabetes mellitus: a prospective population-based study in Tianjin, China. Diabetes Metab Res Rev 2015; 31:627-37. [PMID: 25820620 DOI: 10.1002/dmrr.2650] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/04/2014] [Accepted: 03/16/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND The ABO blood types are associated with cancers, cardiovascular diseases and type 2 diabetes mellitus but whether they are also associated with gestational diabetes mellitus (GDM) is unknown. We examined the relationship between the ABO blood types and the risk of GDM in a prospective population-based Chinese cohort. METHODS From 2010 to 2012, we recruited 14,198 pregnant women within the first 12 weeks of gestation in Tianjin, China. All women had a glucose challenge test (GCT) at 24-28 gestational weeks, followed by a 75-g 2-h oral glucose tolerance test if the results from GCT were ≥7.8 mmol/L. GDM was diagnosed based on the glucose cut-points of the International Association of Diabetes and Pregnancy Study Group criteria. Logistic regression was used to obtain odds ratios (ORs) and 95% confidence intervals (CIs) adjusted for traditional risk factors. Stratified analysis was performed by family history of diabetes (yes versus no). Sensitivity analyses were also performed by using the World Health Organization (WHO) criteria for GDM. RESULTS Women with blood groups A, B or O (i.e. non-AB) were associated with increased risk of GDM as compared with those with blood group AB (adjusted OR: 1.44, 95% CI: 1.13-1.83). Sensitivity analyses showed that the result was consistent using WHO criteria. The adjusted OR of blood group non-AB versus AB for GDM was enhanced among women with a family history of diabetes (2.69, 1.21-5.96) and attenuated among those without (1.33, 1.03-1.71). CONCLUSIONS Blood group AB was a protective factor against GDM in pregnant Chinese women.
Collapse
Affiliation(s)
- Cuiping Zhang
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Yi Li
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Leishen Wang
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Shurong Sun
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Gongshu Liu
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Junhong Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Jia Guo
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Li Lv
- Tianjin Women and Children's Health Centre, Tianjin, China
| | - Weidong Li
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Cuilin Zhang
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, Canada
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| |
Collapse
|
516
|
Roopchand DE, Carmody RN, Kuhn P, Moskal K, Rojas-Silva P, Turnbaugh PJ, Raskin I. Dietary Polyphenols Promote Growth of the Gut Bacterium Akkermansia muciniphila and Attenuate High-Fat Diet-Induced Metabolic Syndrome. Diabetes 2015; 64:2847-58. [PMID: 25845659 PMCID: PMC4512228 DOI: 10.2337/db14-1916] [Citation(s) in RCA: 475] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/19/2014] [Accepted: 03/28/2015] [Indexed: 12/12/2022]
Abstract
Dietary polyphenols protect against metabolic syndrome, despite limited absorption and digestion, raising questions about their mechanism of action. We hypothesized that one mechanism may involve the gut microbiota. To test this hypothesis, C57BL/6J mice were fed a high-fat diet (HFD) containing 1% Concord grape polyphenols (GP). Relative to vehicle controls, GP attenuated several effects of HFD feeding, including weight gain, adiposity, serum inflammatory markers (tumor necrosis factor [TNF]α, interleukin [IL]-6, and lipopolysaccharide), and glucose intolerance. GP lowered intestinal expression of inflammatory markers (TNFα, IL-6, inducible nitric oxide synthase) and a gene for glucose absorption (Glut2). GP increased intestinal expression of genes involved in barrier function (occludin) and limiting triglyceride storage (fasting-induced adipocyte factor). GP also increased intestinal gene expression of proglucagon, a precursor of proteins that promote insulin production and gut barrier integrity. 16S rRNA gene sequencing and quantitative PCR of cecal and fecal samples demonstrated that GP dramatically increased the growth of Akkermansia muciniphila and decreased the proportion of Firmicutes to Bacteroidetes, consistent with prior reports that similar changes in microbial community structure can protect from diet-induced obesity and metabolic disease. These data suggest that GP act in the intestine to modify gut microbial community structure, resulting in lower intestinal and systemic inflammation and improved metabolic outcomes. The gut microbiota may thus provide the missing link in the mechanism of action of poorly absorbed dietary polyphenols.
Collapse
Affiliation(s)
- Diana E Roopchand
- School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ Nutrasorb, LLC, North Brunswick, NJ
| | - Rachel N Carmody
- G.W. Hooper Research Foundation, University of California, San Francisco, San Francisco, CA
| | - Peter Kuhn
- School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ
| | | | - Patricio Rojas-Silva
- School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ
| | - Peter J Turnbaugh
- G.W. Hooper Research Foundation, University of California, San Francisco, San Francisco, CA
| | - Ilya Raskin
- School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ
| |
Collapse
|
517
|
Castanon N, Luheshi G, Layé S. Role of neuroinflammation in the emotional and cognitive alterations displayed by animal models of obesity. Front Neurosci 2015; 9:229. [PMID: 26190966 PMCID: PMC4490252 DOI: 10.3389/fnins.2015.00229] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/31/2015] [Accepted: 06/11/2015] [Indexed: 12/15/2022] Open
Abstract
Obesity is associated with a high prevalence of mood disorders and cognitive dysfunctions in addition to being a significant risk factor for important health complications such as cardiovascular diseases and type 2 diabetes. Identifying the pathophysiological mechanisms underlying these health issues is a major public health challenge. Based on recent findings, from studies conducted on animal models of obesity, it has been proposed that inflammatory processes may participate in both the peripheral and brain disorders associated with the obesity condition including the development of emotional and cognitive alterations. This is supported by the fact that obesity is characterized by peripheral low-grade inflammation, originating from increased adipose tissue mass and/or dysbiosis (changes in gut microbiota environment), both of which contribute to increased susceptibility to immune-mediated diseases. In this review, we provide converging evidence showing that obesity is associated with exacerbated neuroinflammation leading to dysfunction in vulnerable brain regions associated with mood regulation, learning, and memory such as the hippocampus. These findings give new insights to the pathophysiological mechanisms contributing to the development of brain disorders in the context of obesity and provide valuable data for introducing new therapeutic strategies for the treatment of neuropsychiatric complications often reported in obese patients.
Collapse
Affiliation(s)
- Nathalie Castanon
- Nutrition and Integrative Neurobiology, INRA, UMR 1286, Université de Bordeaux Bordeaux, France
| | - Giamal Luheshi
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University Montreal, Canada
| | - Sophie Layé
- Nutrition and Integrative Neurobiology, INRA, UMR 1286, Université de Bordeaux Bordeaux, France
| |
Collapse
|
518
|
Sellmann C, Priebs J, Landmann M, Degen C, Engstler AJ, Jin CJ, Gärttner S, Spruss A, Huber O, Bergheim I. Diets rich in fructose, fat or fructose and fat alter intestinal barrier function and lead to the development of nonalcoholic fatty liver disease over time. J Nutr Biochem 2015; 26:1183-92. [PMID: 26168700 DOI: 10.1016/j.jnutbio.2015.05.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/22/2014] [Revised: 05/08/2015] [Accepted: 05/13/2015] [Indexed: 02/07/2023]
Abstract
General overnutrition but also a diet rich in certain macronutrients, age, insulin resistance and an impaired intestinal barrier function may be critical factors in the development of nonalcoholic fatty liver disease (NAFLD). Here the effect of chronic intake of diets rich in different macronutrients, i.e. fructose and/or fat on liver status in mice, was studied over time. C57BL/6J mice were fed plain water, 30% fructose solution, a high-fat diet or a combination of both for 8 and 16 weeks. Indices of liver damage, toll-like receptor 4 (TLR-4) signaling cascade, macrophage polarization and insulin resistance in the liver and intestinal barrier function were analyzed. Chronic exposure to a diet rich in fructose and/or fat was associated with the development of hepatic steatosis that progressed with time to steatohepatitis in mice fed a combination of macronutrients. The development of NAFLD was also associated with a marked reduction of the mRNA expression of insulin receptor, whereas hepatic expressions of TLR-4, myeloid differentiation primary response gene 88 and markers of M1 polarization of macrophages were induced in comparison to controls. Bacterial endotoxin levels in portal plasma were found to be increased while levels of the tight junction protein occludin and zonula occludens 1 were found to be significantly lower in the duodenum of all treated groups after 8 and 16 weeks. Our data suggest that chronic intake of fructose and/or fat may lead to the development of NAFLD over time and that this is associated with an increased translocation of bacterial endotoxin.
Collapse
Affiliation(s)
- Cathrin Sellmann
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Josephine Priebs
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Marianne Landmann
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Christian Degen
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Anna Janina Engstler
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Cheng Jun Jin
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Stefanie Gärttner
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Astrid Spruss
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Otmar Huber
- Department of Biochemistry II, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany; Center of Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Ina Bergheim
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.
| |
Collapse
|
519
|
Li Z, Summanen PH, Komoriya T, Henning SM, Lee RP, Carlson E, Heber D, Finegold SM. Pomegranate ellagitannins stimulate growth of gut bacteria in vitro: Implications for prebiotic and metabolic effects. Anaerobe 2015; 34:164-8. [PMID: 26051169 DOI: 10.1016/j.anaerobe.2015.05.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2015] [Revised: 04/24/2015] [Accepted: 05/12/2015] [Indexed: 11/26/2022]
Abstract
The present study investigated the effect of pomegranate extract (POMx) and pomegranate juice (POM juice) on the growth of major groups of intestinal bacteria: Enterobacteriaceae, Bacteroides fragilis group, clostridia, bifidobacteria, and lactobacilli, and the utilization of pomegranate polyphenols by Bifidobacterium and Lactobacillus. The total phenolic content of the pomegranate extract and juice was determined using the Folin-Ciocalteau colorimetric method and reported as gallic acid equivalent (GAE). The polyphenol composition was determined by HPLC. Stool specimens were incubated with 400, 100, and 25 μg/ml GAE POMx and POM juice and subjected to selective culture. Bifidobacterium and Lactobacillus strains were incubated with 400 μg/ml GAE POMx and POM juice and metabolites were analyzed. POMx and POM juice increased the mean counts of Bifidobacterium and Lactobacillus and significantly inhibited the growth of B. fragilis group, clostridia, and Enterobacteriaceae in a dose-response manner. Bifidobacterium and Lactobacillus utilized ellagic acid and glycosyl ellagic acid but little or no punicalin was utilized. Neither POMx nor POM juice was converted to urolithins by the test bacteria or the in vitro stool cultures. The effect of pomegranate on the gut bacteria considered to be beneficial (Bifidobacterium and Lactobacillus) suggests that pomegranate may potentially work as a prebiotic. The concept that polyphenols such as those in pomegranate impact gut microbiota populations may establish a new role for polyphenols in human health.
Collapse
Affiliation(s)
- Zhaoping Li
- Department of Medicine, VA Medical Center West Los Angeles, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA; Department of Research Service, VA Medical Center West Los Angeles, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA; Department of Medicine, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA; UCLA Center for Human Nutrition, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA.
| | - Paula H Summanen
- Department of Research Service, VA Medical Center West Los Angeles, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA
| | - Tomoe Komoriya
- Department of Research Service, VA Medical Center West Los Angeles, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA; Department of Sustainable Engineering, College of Industrial Technology, Nihon University, Japan
| | - Susanne M Henning
- UCLA Center for Human Nutrition, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA
| | - Ru-Po Lee
- UCLA Center for Human Nutrition, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA
| | - Eliisa Carlson
- UCLA Center for Human Nutrition, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA
| | - David Heber
- UCLA Center for Human Nutrition, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA
| | - Sydney M Finegold
- Department of Medicine, VA Medical Center West Los Angeles, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA; Department of Research Service, VA Medical Center West Los Angeles, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA; Department of Microbiology, Immunology and Molecular Genetics, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA; Department of Medicine, UCLA School of Medicine, 405 Hilgard Ave., Los Angeles, CA 90095, USA
| |
Collapse
|
520
|
Potgieter M, Bester J, Kell DB, Pretorius E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS Microbiol Rev 2015; 39:567-91. [PMID: 25940667 PMCID: PMC4487407 DOI: 10.1093/femsre/fuv013] [Citation(s) in RCA: 274] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 03/02/2015] [Indexed: 02/07/2023] Open
Abstract
Blood in healthy organisms is seen as a ‘sterile’ environment: it lacks proliferating microbes. Dormant or not-immediately-culturable forms are not absent, however, as intracellular dormancy is well established. We highlight here that a great many pathogens can survive in blood and inside erythrocytes. ‘Non-culturability’, reflected by discrepancies between plate counts and total counts, is commonplace in environmental microbiology. It is overcome by improved culturing methods, and we asked how common this would be in blood. A number of recent, sequence-based and ultramicroscopic studies have uncovered an authentic blood microbiome in a number of non-communicable diseases. The chief origin of these microbes is the gut microbiome (especially when it shifts composition to a pathogenic state, known as ‘dysbiosis’). Another source is microbes translocated from the oral cavity. ‘Dysbiosis’ is also used to describe translocation of cells into blood or other tissues. To avoid ambiguity, we here use the term ‘atopobiosis’ for microbes that appear in places other than their normal location. Atopobiosis may contribute to the dynamics of a variety of inflammatory diseases. Overall, it seems that many more chronic, non-communicable, inflammatory diseases may have a microbial component than are presently considered, and may be treatable using bactericidal antibiotics or vaccines. Atopobiosis of microbes (the term describing microbes that appear in places other than where they should be), as well as the products of their metabolism, seems to correlate with, and may contribute to, the dynamics of a variety of inflammatory diseases.
Collapse
Affiliation(s)
- Marnie Potgieter
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Janette Bester
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Douglas B Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, 131, Princess St, Manchester M1 7DN, Lancs, UK
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| |
Collapse
|
521
|
Lu Z, Li Y, Jin J, Zhang X, Hannun YA, Huang Y. GPR40/FFA1 and neutral sphingomyelinase are involved in palmitate-boosted inflammatory response of microvascular endothelial cells to LPS. Atherosclerosis 2015; 240:163-73. [PMID: 25795558 PMCID: PMC4397186 DOI: 10.1016/j.atherosclerosis.2015.03.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/27/2014] [Revised: 01/25/2015] [Accepted: 03/08/2015] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Increased levels of both saturated fatty acids (SFAs) and lipopolysaccharide (LPS) are associated with type 2 diabetes. However, it remains largely unknown how SFAs interact with LPS to regulate inflammatory responses in microvascular endothelial cells (MIC ECs) that are critically involved in atherosclerosis as a diabetic complication. In this study, we compared the effects of LPS, palmitic acid (PA), the most abundant saturated fatty acid, or the combination of LPS and PA on interleukin (IL)-6 expression by MIC ECs and explored the underlying mechanisms. METHODS Human cardiac MIC ECs were treated with LPS, PA and LPS plus PA and the regulatory pathways including receptors, signal transduction, transcription and post-transcription, and sphingolipid metabolism for IL-6 expression were investigated. RESULTS G protein-coupled receptor (GPR)40 or free fatty acid receptor 1 (FFA1), but not toll-like receptor 4, was involved in PA-stimulated IL-6 expression. PA not only stimulated IL-6 expression by itself, but also remarkably enhanced LPS-stimulated IL-6 expression via a cooperative stimulation on mitogen-activated protein kinase and nuclear factor kappa B signaling pathways, and both transcriptional and post-transcriptional activation. Furthermore, PA induced a robust neutral sphingomyelinase (nSMase)-mediated sphingomyelin hydrolysis that was involved in PA-augmented IL-6 upregulation. CONCLUSION PA boosted inflammatory response of microvascular endothelial cells to LPS via GPR40 and nSMase.
Collapse
Affiliation(s)
- Zhongyang Lu
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Junfei Jin
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, People's Republic of China
| | - Xiaoming Zhang
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yusuf A Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yan Huang
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA; Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
522
|
Osto M, Lutz TA. Translational value of animal models of obesity-Focus on dogs and cats. Eur J Pharmacol 2015; 759:240-52. [PMID: 25814247 DOI: 10.1016/j.ejphar.2015.03.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/24/2014] [Revised: 01/15/2015] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
A prolonged imbalance between a relative increase in energy intake over a decrease in energy expenditure results in the development of obesity; extended periods of a positive energy balance eventually lead to the accumulation of abnormally high amounts of fat in adipose tissue but also in other organs. Obesity is considered a clinical state of impaired general heath in which the excessive increase in adipose tissue mass may be associated with metabolic disorders such as type 2 diabetes mellitus, hyperlipidemia, hypertension and cardiovascular diseases. This review discusses briefly the use of animal models for the study of obesity and its comorbidities. Generally, most studies are performed with rodents, such as diet induced obesity and genetic models. Here, we focus specifically on two different species, namely dogs and cats. Obese dogs and cats show many features of human obesity. Interestingly, however, dogs and cats differ from each other in certain aspects because even though obese dogs may become insulin resistant, this does not result in the development of diabetes mellitus. In fact, diabetes in dogs is typically not associated with obesity because dogs present a type 1 diabetes-like syndrome. On the other hand, obese cats often develop diabetes mellitus which shares many features with human type 2 diabetes; feline and human diabetes are similar in respect to their pathophysiology, underlying risk factors and treatment strategies. Our review discusses genetic and endocrine factors in obesity, discusses obesity induced changes in lipid metabolism and includes some recent findings on the role of gut microbiota in obesity. Compared to research in rodent models, the array of available techniques and tools is unfortunately still rather limited in dogs and cats. Hence, even though physiological and pathophysiological phenomena are well described in dogs and cats, the underlying mechanisms are often not known and studies investigating causality specifically are scarce.
Collapse
Affiliation(s)
- Melania Osto
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH 8057 Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH 8057 Zurich, Switzerland; Institute of Laboratory Animal Science, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 190, CH 8057 Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zurich, Switzerland.
| |
Collapse
|
523
|
Pekkala S, Munukka E, Kong L, Pöllänen E, Autio R, Roos C, Wiklund P, Fischer-Posovszky P, Wabitsch M, Alen M, Huovinen P, Cheng S. Toll-like receptor 5 in obesity: the role of gut microbiota and adipose tissue inflammation. Obesity (Silver Spring) 2015; 23:581-90. [PMID: 25611816 DOI: 10.1002/oby.20993] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/26/2014] [Accepted: 11/03/2014] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study aimed at establishing bacterial flagellin-recognizing toll-like receptor 5 (TLR5) as a novel link between gut microbiota composition, adipose tissue inflammation, and obesity. METHODS An adipose tissue microarray database was used to compare women having the highest (n = 4, H-TLR) and lowest (n = 4, L-TLR) expression levels of TLR5-signaling pathway genes. Gut microbiota composition was profiled using flow cytometry and FISH. Standard laboratory techniques were used to determine anthropometric and clinical variables. In vivo results were verified using cultured human adipocytes. RESULTS The H-TLR group had higher flagellated Clostridium cluster XIV abundance and Firmicutes-to-Bacteroides ratio. H-TLR subjects had obese phenotype characterized by greater waist circumference, fat %, and blood pressure (P < 0.05 for all). They also had higher leptin and lower adiponectin levels (P < 0.05 for both). Six hundred and sixty-eight metabolism- and inflammation-related adipose tissue genes were differentially expressed between the groups. In vitro studies confirmed that flagellin activated TLR5 inflammatory pathways, decreased insulin signaling, and increased glycerol secretion. CONCLUSIONS The in vivo findings suggest that flagellated Clostridium cluster XIV bacteria contribute to the development of obesity through distorted adipose tissue metabolism and inflammation. The in vitro studies in adipocytes show that the underlying mechanisms of the human findings may be due to flagellin-activated TLR5 signaling.
Collapse
Affiliation(s)
- Satu Pekkala
- Department of Health Sciences, University of Jyväskylä, Jyväskylä, Finland; Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
524
|
Patel PN, Shah RY, Ferguson JF, Reilly MP. Human experimental endotoxemia in modeling the pathophysiology, genomics, and therapeutics of innate immunity in complex cardiometabolic diseases. Arterioscler Thromb Vasc Biol 2015; 35:525-34. [PMID: 25550206 PMCID: PMC4344396 DOI: 10.1161/atvbaha.114.304455] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2014] [Accepted: 12/18/2014] [Indexed: 01/16/2023]
Abstract
Inflammation is a fundamental feature of several complex cardiometabolic diseases. Indeed, obesity, insulin resistance, metabolic dyslipidemia, and atherosclerosis are all closely linked inflammatory states. Increasing evidence suggests that the infectious, biome-related, or endogenous activation of the innate immune system may contribute to the development of metabolic syndrome and cardiovascular disease. Here, we describe the human experimental endotoxemia model for the specific study of innate immunity in understanding further the pathogenesis of cardiometabolic disease. In a controlled, experimental setting, administration of an intravenous bolus of purified Escherichia coli endotoxin activates innate immunity in healthy human volunteers. During endotoxemia, changes emerge in glucose metabolism, lipoprotein composition, and lipoprotein functions that closely resemble those observed chronically in inflammatory cardiovascular disease risk states. In this review, we describe the transient systemic inflammation and specific metabolic consequences that develop during human endotoxemia. Such a model provides a controlled induction of systemic inflammation, eliminates confounding, undermines reverse causation, and possesses unique potential as a starting point for genomic screening and testing of novel therapeutics for treatment of the inflammatory underpinning of cardiometabolic disease.
Collapse
Affiliation(s)
- Parth N Patel
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Rhia Y Shah
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Jane F Ferguson
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Muredach P Reilly
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.).
| |
Collapse
|
525
|
Zijlmans MAC, Korpela K, Riksen-Walraven JM, de Vos WM, de Weerth C. Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology 2015; 53:233-45. [PMID: 25638481 DOI: 10.1016/j.psyneuen.2015.01.006] [Citation(s) in RCA: 313] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/25/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 02/08/2023]
Abstract
Maternal prenatal stress has been often associated with infant physical development and health, as well as psychological functioning and behavior. However, the mechanisms underlying these relations remain elusive. The goal of the present study was to prospectively investigate the development of the intestinal microbiota as a potential pathway linking maternal prenatal stress and infant health. The development of the infant intestinal microbiota was followed over the first 110 days after birth in a healthy cohort of 56 vaginally born Dutch infants. Additionally, the relation between infant intestinal microbiota and gastrointestinal and allergic symptoms was examined. Results showed that maternal prenatal stress, i.e., either reported stress or elevated basal maternal salivary cortisol concentrations or both, was strongly and persistently associated with the infants' microbiota composition as determined by a phylogenetic microarray. Infants of mothers with high cumulative stress (i.e., high reported stress and high cortisol concentrations) during pregnancy had significantly higher relative abundances of Proteobacterial groups known to contain pathogens (related to Escherichia, Serratia, and Enterobacter), and lower relative abundances of lactic acid bacteria (i.e., Lactobacillus, Lactoccus, Aerococcus) and Bifidobacteria, altogether characteristics of a potentially increased level of inflammation. Furthermore, this aberrant colonization pattern was related to more maternally reported infant gastrointestinal symptoms and allergic reactions. In conclusion, clear links were found between maternal prenatal stress and the infant intestinal microbiota and health. Although causality cannot be concluded, the results suggest a possible mechanism by which maternal prenatal stress influences the offspring development. These results suggest a potential for bacterial interventions to enhance offspring health and development in pregnant women with stress.
Collapse
Affiliation(s)
- Maartje A C Zijlmans
- Department of Developmental Psychology, Behavioural Science Institute, Radboud University Nijmegen, Postbus 9104, 6500 HE Nijmegen, The Netherlands.
| | - Katri Korpela
- Department of Bacteriology and Immunology, Immunobiology Research Program, University of Helsinki, 00014 Helsinki, Finland
| | - J Marianne Riksen-Walraven
- Department of Developmental Psychology, Behavioural Science Institute, Radboud University Nijmegen, Postbus 9104, 6500 HE Nijmegen, The Netherlands
| | - Willem M de Vos
- Department of Bacteriology and Immunology, Immunobiology Research Program, University of Helsinki, 00014 Helsinki, Finland; Laboratory of Microbiology, Wageningen UR, Building 316, Dreijenplein 10, 6703 HB Wageningen, The Netherlands
| | - Carolina de Weerth
- Department of Developmental Psychology, Behavioural Science Institute, Radboud University Nijmegen, Postbus 9104, 6500 HE Nijmegen, The Netherlands.
| |
Collapse
|
526
|
Fagherazzi G, Gusto G, Clavel-Chapelon F, Balkau B, Bonnet F. ABO and Rhesus blood groups and risk of type 2 diabetes: evidence from the large E3N cohort study. Diabetologia 2015; 58:519-22. [PMID: 25533388 DOI: 10.1007/s00125-014-3472-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/27/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS The objective of this study was to evaluate the relationship of ABO blood type (A, B, AB and O), Rhesus factor (positive or negative) and a combination of the two (ABO × Rhesus) with type 2 diabetes mellitus risk. METHODS In total, 82,104 women from the large prospective E3N cohort were followed between 1990 and 2008. Multivariate Cox regression models were used to estimate HRs and 95% CIs. RESULTS Those with either the A (HR 1.10 [95% CI 1.02, 1.18]) or B (HR 1.21 [95% CI 1.07, 1.36]) group were at increased risk of type 2 diabetes mellitus compared with those with the O group. The association with the AB group did not reach statistical significance (HR 1.17 [95% CI 0.99, 1.39]). There was no difference in type 2 diabetes mellitus risk between Rhesus positive and negative groups (HR 0.96 [95% CI 0.88, 1.05]). When the universal donors (O(-)) were taken as the reference category, we observed an increased risk for both A(+) (HR 1.17 [95% CI 1.02, 1.35]) and A(-) (HR 1.22 [95% CI 1.03, 1.45]) individuals. The greatest increase in risk was seen for those with the B(+) blood group (HR 1.35 [95% CI 1.13, 1.60]). We also observed a greater type 2 diabetes mellitus risk for those with the AB(+) group (HR 1.26 [95% CI 1.02, 1.57]). Adjustment for fasting plasma glucose and lipid concentrations in a case-control subsample did not alter the associations. CONCLUSIONS/INTERPRETATION This study suggests that people with the O blood type have a lower risk of developing type 2 diabetes mellitus. Therefore, blood group should be investigated in future clinical and epidemiological studies on diabetes, and further pathophysiological research is needed to determine why individuals with blood type O have a lower risk of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Guy Fagherazzi
- Team 9, Nutrition, Hormones and Women's Health, Inserm U1018, Centre for Research in Epidemiology and Population Health, Gustave Roussy Institute, 114 rue Edouard Vaillant, 94805, Villejuif Cedex, France,
| | | | | | | | | |
Collapse
|
527
|
Park SH, Kim KA, Ahn YT, Jeong JJ, Huh CS, Kim DH. Comparative analysis of gut microbiota in elderly people of urbanized towns and longevity villages. BMC Microbiol 2015; 15:49. [PMID: 25887483 PMCID: PMC4345030 DOI: 10.1186/s12866-015-0386-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/04/2014] [Accepted: 02/12/2015] [Indexed: 12/12/2022] Open
Abstract
Background To understand differences in the gut microbiota between elderly people of urbanized town communities (UTC) and longevity village communities (LVC), we analyzed fecal microbiota collected from individuals living in 2 UTC (Seoul and Chuncheon) and 3 LVC (Gurye, Damyang, and Soonchang) selected on the basis of indices for superlongevity (the ratio of centenarians to the total population) and longevity (the ratio of those aged 85 years or greater to those aged 65 years or greater) in South Korea by 454 pyrosequencing. Results Taxonomy-based analysis showed that The relative abundance of Firmicutes, Tenericutes, and Actinobacteria was significantly lower in LVC than in UTC. Due to an increase of Firmicutes and a reduction of Bacteroidetes, the ratio of Firmicutes to Bacteroidetes in the gut microbiota was greater in UTC adults than in UTC children or LVC adults. The population levels of Bacteroides, Prevotella, and Lachnospira were significantly higher in LVC than in UTC, but the levels of Dialister, Subdoligranulum, Megamonas, EF401882_g, and AM275436_g were lower in LVC than in UTC. Although most of the species detected in LVC were detected in UTC, some Bacteroides spp. and Faecalibacterium spp. were detected only in LVC. Among Bacteroides spp., ACWH_s, EF403317_s, and EF403722_s were detected in children and LVC samples only but FJ363527_s, 4P000677_s, and 4P000015_s were detected in UTC samples. EF402172_s and EF404388_s, members of Faecalibacterium spp., which are known to have anti-inflammatory properties, were detected in LVC and children only (>3.9% of total sequence). In addition, the fecal lipopolysaccharides (LPS) content was significantly higher in UTC than in LVC. Conclusions These findings suggest that maintaining gut microbiota, including Faecalibacterium spp. EF402172_s and EF404388_s, as well as low LPS levels may play an important role in preserving residents’ health in LVC. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0386-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Se-Hoon Park
- R &B D Center, Korea Yakult Co., Ltd., Yongin-si, Kyunggi-do, 446-901, Korea.
| | - Kyung-Ah Kim
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, 1, Hoegi, Dongdaemun-ku, Seoul, 130-701, Korea.
| | - Young-Tae Ahn
- R &B D Center, Korea Yakult Co., Ltd., Yongin-si, Kyunggi-do, 446-901, Korea.
| | - Jin-Ju Jeong
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, 1, Hoegi, Dongdaemun-ku, Seoul, 130-701, Korea.
| | - Chul-Sung Huh
- Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, 151-742, Korea.
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, 1, Hoegi, Dongdaemun-ku, Seoul, 130-701, Korea.
| |
Collapse
|
528
|
Marimoutou M, Le Sage F, Smadja J, Lefebvre d'Hellencourt C, Gonthier MP, Robert-Da Silva C. Antioxidant polyphenol-rich extracts from the medicinal plants Antirhea borbonica, Doratoxylon apetalum and Gouania mauritiana protect 3T3-L1 preadipocytes against H2O2, TNFα and LPS inflammatory mediators by regulating the expression of superoxide dismutase and NF-κB genes. JOURNAL OF INFLAMMATION-LONDON 2015; 12:10. [PMID: 25685071 PMCID: PMC4329220 DOI: 10.1186/s12950-015-0055-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/15/2014] [Accepted: 01/20/2015] [Indexed: 12/22/2022]
Abstract
Background Adipose cells responsible for fat storage are the targets of reactive oxygen species (ROS) like H2O2 and pro-inflammatory agents including TNFα and LPS. Such mediators contribute to oxidative stress and alter inflammatory processes in adipose tissue, leading to insulin resistance during obesity. Thus, the identification of natural compounds such as plant polyphenols able to increase the antioxidant and anti-inflammatory capacity of the body is of high interest. We aimed to evaluate the biological properties of polyphenol-rich extracts from the medicinal plants A. borbonica, D. apetalum and G. mauritiana on preadipocytes exposed to H2O2, TNFα or LPS mediators. Methods Medicinal plant extracts were analysed for their polyphenol contents by Folin-Ciocalteu and UPLC-ESI-MS methods as well as for their free radical-scavenging activities by DPPH and ORAC assays. To assess the ability of polyphenol-rich extracts to protect 3T3-L1 preadipocytes against H2O2, TNFα or LPS mediators, several parameters including cell viability (MTT and LDH assays), ROS production (DCFH-DA test), IL-6 and MCP-1 secretion (ELISA) were evaluated. Moreover, the expression of superoxide dismutase, catalase and NF-κB genes was explored (RT-QPCR). Results All medicinal plants exhibited high levels of polyphenols with free radical-scavenging capacities. Flavonoids such as quercetin, kaempferol, epicatechin and procyanidins, and phenolic acids derived from caffeic acid including chlorogenic acid, were detected. Polyphenol-rich plant extracts did not exert a cytotoxic effect on preadipocytes but protected them against H2O2 anti-proliferative action. Importantly, they down-regulated ROS production and the secretion of IL-6 and MCP-1 pro-inflammatory markers induced by H2O2, TNFα and LPS mediators. Such a protective action was associated with an increase in superoxide dismutase antioxidant enzyme gene expression and a decrease in mRNA levels of NF-κB pro-inflammatory transcription factor. Conclusion This study highlights that antioxidant strategies based on polyphenols derived from medicinal plants tested could contribute to regulate adipose tissue redox status and immune process, and thus participate to the improvement of obesity-related oxidative stress and inflammation.
Collapse
Affiliation(s)
- Méry Marimoutou
- UMR Diabète athérothrombose Thérapies Réunion Océan Indien, Inserm U1188 - Université de La Réunion, Plateforme CYROI, 2 rue Maxime Rivière, 97490 Saint-Denis, La Réunion France
| | - Fanny Le Sage
- UMR Diabète athérothrombose Thérapies Réunion Océan Indien, Inserm U1188 - Université de La Réunion, Plateforme CYROI, 2 rue Maxime Rivière, 97490 Saint-Denis, La Réunion France
| | - Jacqueline Smadja
- Laboratoire de Chimie des Substances Naturelles et des Sciences des Aliments, EA 2212, Université de La Réunion, 15 avenue René Cassin, CS 92003, 97744 Saint-Denis, La Réunion France
| | - Christian Lefebvre d'Hellencourt
- UMR Diabète athérothrombose Thérapies Réunion Océan Indien, Inserm U1188 - Université de La Réunion, Plateforme CYROI, 2 rue Maxime Rivière, 97490 Saint-Denis, La Réunion France
| | - Marie-Paule Gonthier
- UMR Diabète athérothrombose Thérapies Réunion Océan Indien, Inserm U1188 - Université de La Réunion, Plateforme CYROI, 2 rue Maxime Rivière, 97490 Saint-Denis, La Réunion France
| | - Christine Robert-Da Silva
- UMR Diabète athérothrombose Thérapies Réunion Océan Indien, Inserm U1188 - Université de La Réunion, Plateforme CYROI, 2 rue Maxime Rivière, 97490 Saint-Denis, La Réunion France
| |
Collapse
|
529
|
Jiang W, Wu N, Wang X, Chi Y, Zhang Y, Qiu X, Hu Y, Li J, Liu Y. Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci Rep 2015; 5:8096. [PMID: 25644696 PMCID: PMC4314632 DOI: 10.1038/srep08096] [Citation(s) in RCA: 431] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2014] [Accepted: 01/06/2015] [Indexed: 12/18/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has recently been considered to be under the influence of the gut microbiota, which might exert toxic effects on the human host after intestinal absorption and delivery to the liver via the portal vein. In this study, the composition of the gut microbiota in NAFLD patients and healthy subjects was determined via 16S ribosomal RNA Illumina next-generation sequencing. Among those taxa displaying greater than 0.1% average abundance in all samples, five genera, including Alistipes and Prevotella, were significantly more abundant in the gut microbiota of healthy subjects compared to NAFLD patients. Alternatively, Escherichia, Anaerobacter, Lactobacillus and Streptococcus were increased in the gut microbiota of NAFLD patients compared to healthy subjects. In addition, decreased numbers of CD4+ and CD8+ T lymphocytes and increased levels of TNF-α, IL-6 and IFN-γ were detected in the NAFLD group compared to the healthy group. Furthermore, irregularly arranged microvilli and widened tight junctions were observed in the gut mucosa of the NAFLD patients via transmission electron microscopy. We postulate that aside from dysbiosis of the gut microbiota, gut microbiota-mediated inflammation of the intestinal mucosa and the related impairment in mucosal immune function play an important role in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Weiwei Jiang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Na Wu
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People's Hospital, Beijing, China
| | - Xuemei Wang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Yujing Chi
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People's Hospital, Beijing, China
| | - Yuanyuan Zhang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Xinyun Qiu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Ying Hu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Jing Li
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
530
|
Mira-Pascual L, Cabrera-Rubio R, Ocon S, Costales P, Parra A, Suarez A, Moris F, Rodrigo L, Mira A, Collado MC. Microbial mucosal colonic shifts associated with the development of colorectal cancer reveal the presence of different bacterial and archaeal biomarkers. J Gastroenterol 2015; 50:167-79. [PMID: 24811328 DOI: 10.1007/s00535-014-0963-x] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/28/2014] [Accepted: 04/11/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND Epidemiological studies demonstrate a link between gastrointestinal cancers and environmental factors such as diet. It has been suggested that environmental cancer risk is determined by the interaction between diet and microbes. Thus, the purpose of this study was to examine the hypothesis that microbiota composition during colorectal cancer (CRC) progression might differ depending on the stage of the disease. METHODS A total of 28 age-matched and sex-matched subjects, seven with CRC adenocarcinoma, 11 with tubular adenomas and ten healthy subjects with intact colon, were included into the study. Microbiomes from mucosal and fecal samples were analyzed with 16S ribosomal RNA gene pyrosequencing, together with quantitative PCR of specific bacteria and archaea. RESULTS The principal coordinates analysis clearly separated healthy tissue samples from polyps and tumors, supporting the presence of specific bacterial consortia that are associated with affected sites and that can serve as potential biomarkers of CRC progression. A higher presence of Fusobacterium nucleatum and Enterobacteriaceae was found by qPCR in samples from CRC compared to healthy controls. We observed a correlation between CRC process development and levels of Methanobacteriales (R = 0.537, P = 0.007) and Methanobrevibacterium (R = 0.574, P = 0.03) in fecal samples. CONCLUSION Differences in microbial and archaeal composition between mucosal samples from healthy and disease tissues were observed in tubular adenoma and adenocarcinoma. In addition, microbiota from mucosal samples represented the underlying dysbiosis, whereas fecal samples seem not to be appropriate to detect shifts in microbial composition. CRC risk is influenced by microbial composition, showing differences according to disease progression step and tumor severity.
Collapse
Affiliation(s)
- L Mira-Pascual
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Av. Agustin Escardino 7, 49860, Paterna, Valencia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
531
|
Abstract
The incidence of obesity in the western world has increased dramatically during recent decades. Epidemiological data suggest that obesity is associated with an increased risk of several but not all types of cancers, with clear sex-specific differences. The underlying mechanisms are still a matter of debate. This review focuses on the potential factors linking obesity to cancer. Current experimental evidence suggests that insulin resistance and a chronic, subclinical inflammation in the visceral fat are the major metabolic events causing alterations in the levels of insulin, glucose, free fatty acids, insulin-like growth factor 1 (IGF-1) and 2, adipose tissue-derived proinflammatory cytokines and other bioactive molecules, such as adipokines (e.g. leptin and adiponectin), vascular endothelial growth factor (VEGF), sex hormones, gut microbiota and secondary bile acids. All these factors may act directly or indirectly on the tumor microenvironment to drive tumor progression via stimulation of cell survival/antiapoptosis, cell proliferation, angiogenesis and invasion/metastasis of the cancer cells. Therapeutic strategies that target dysfunctional or inflamed fat and have been shown to benefit patients include bariatric surgery, while other cell or hormone-directed interventions, such as conversion of visceral fat macrophages to an anti-inflammatory M2 phenotype or the pharmacological modulation of serum adipokine levels are still theoretical and need to be clinically evaluated for their ability to successfully treat or prevent obesity-related cancers.
Collapse
Affiliation(s)
- H Ungefroren
- Abteilung Hämatologie/Onkologie, Medizinische Klinik I -ZK,UKSH Campus Lübeck, Universitätsklinikum Schleswig-Holstein, Ratzeburger Allee 160, 23538, Lübeck, Deutschland,
| | | | | |
Collapse
|
532
|
The food born mycotoxin deoxynivalenol induces low-grade inflammation in mice in the absence of observed-adverse effects. Toxicol Lett 2015; 232:601-11. [DOI: 10.1016/j.toxlet.2014.12.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/09/2014] [Revised: 12/20/2014] [Accepted: 12/22/2014] [Indexed: 11/24/2022]
|
533
|
Shaikh M, Rajan K, Forsyth CB, Voigt RM, Keshavarzian A. Simultaneous gas-chromatographic urinary measurement of sugar probes to assess intestinal permeability: use of time course analysis to optimize its use to assess regional gut permeability. Clin Chim Acta 2015; 442:24-32. [PMID: 25591964 DOI: 10.1016/j.cca.2014.12.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/28/2014] [Revised: 12/26/2014] [Accepted: 12/29/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Measurement of intestinal permeability is important in several diseases but currently several methods are employed. We sought to: (1) develop a new GC based method to measure urinary mannitol, lactulose and sucralose to assess regional and total gut permeability; (2) analyze the kinetics of these sugars in the urine to determine which ratio is useful to represent intestinal permeability; and (3) determine whether age, gender, race and BMI impact these values. METHODS Subjects drank a cocktail of sucrose, lactulose, mannitol and sucralose and these sugars were measured in the urine at 5, 12 and 24h with gas chromatography. RESULTS Urinary mannitol exhibited significantly different kinetics than lactulose and sucralose which were similar to each other and varied little over the 24h. No permeability differences were observed for renal function, age, race, sex, or BMI. CONCLUSIONS Our data do not support the use of the widely used L/M ratio as an accurate estimate of intestinal permeability. Our data support the use of: the sucralose/lactulose (S/M) ratio to measure: small intestine permeability (first 5h); small and large intestine (first 12h), and total gut permeability (24h). This was also found to be true in a Parkinson's disease model.
Collapse
Affiliation(s)
- Maliha Shaikh
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL, United States.
| | - Kumar Rajan
- Rush Institute for Healthy Aging, Chicago, IL, United States.
| | - Christopher B Forsyth
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL, United States; Department of Biochemistry, Rush University, Chicago, IL, United States.
| | - Robin M Voigt
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL, United States.
| | - Ali Keshavarzian
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL, United States; Department of Pharmacology, Rush University, Chicago, IL, United States; Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL, United States; F.C. Donders Chair, Faculty of Science, University of Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
534
|
|
535
|
Conlon MA, Bird AR. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014; 7:17-44. [PMID: 25545101 PMCID: PMC4303825 DOI: 10.3390/nu7010017] [Citation(s) in RCA: 940] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/17/2014] [Accepted: 12/09/2014] [Indexed: 02/07/2023] Open
Abstract
There is growing recognition of the role of diet and other environmental factors in modulating the composition and metabolic activity of the human gut microbiota, which in turn can impact health. This narrative review explores the relevant contemporary scientific literature to provide a general perspective of this broad area. Molecular technologies have greatly advanced our understanding of the complexity and diversity of the gut microbial communities within and between individuals. Diet, particularly macronutrients, has a major role in shaping the composition and activity of these complex populations. Despite the body of knowledge that exists on the effects of carbohydrates there are still many unanswered questions. The impacts of dietary fats and protein on the gut microbiota are less well defined. Both short- and long-term dietary change can influence the microbial profiles, and infant nutrition may have life-long consequences through microbial modulation of the immune system. The impact of environmental factors, including aspects of lifestyle, on the microbiota is particularly poorly understood but some of these factors are described. We also discuss the use and potential benefits of prebiotics and probiotics to modify microbial populations. A description of some areas that should be addressed in future research is also presented.
Collapse
Affiliation(s)
- Michael A Conlon
- CSIRO Food and Nutrition Flagship, Kintore Ave, Adelaide, SA 5000, Australia.
| | - Anthony R Bird
- CSIRO Food and Nutrition Flagship, Kintore Ave, Adelaide, SA 5000, Australia.
| |
Collapse
|
536
|
Han JL, Lin HL. Intestinal microbiota and type 2 diabetes: From mechanism insights to therapeutic perspective. World J Gastroenterol 2014; 20:17737-17745. [PMID: 25548472 PMCID: PMC4273124 DOI: 10.3748/wjg.v20.i47.17737] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/01/2014] [Revised: 09/26/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
The incidence of type 2 diabetes (T2DM) is rapidly increasing worldwide. However, the pathogenesis of T2DM has not yet been well explained. Recent evidence suggests that the intestinal microbiota composition is associated with obesity and T2DM. In this review, we provide an overview about the mechanisms underlying the role of intestinal microbiota in the pathogenesis of T2DM. There is clear evidence that the intestinal microbiota influences the host through its effect on body weight, bile acid metabolism, proinflammatory activity and insulin resistance, and modulation of gut hormones. Modulating gut microbiota with the use of probiotics, prebiotics, antibiotics, and fecal microbiota transplantation may have benefits for improvement in glucose metabolism and insulin resistance in the host. Further studies are required to increase our understanding of the complex interplay between intestinal microbiota and the host with T2DM. Further studies may be able to boost the development of new effective therapeutic approaches for T2DM.
Collapse
|
537
|
Akiyama T, Kimura K, Hatano H. Diverse galactooligosaccharides consumption by bifidobacteria: implications of β-galactosidase--LacS operon. Biosci Biotechnol Biochem 2014; 79:664-72. [PMID: 25483279 DOI: 10.1080/09168451.2014.987204] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/24/2022]
Abstract
Galactooligosaccharides (GOS) possess prebiotic properties that specifically increase the number of bifidobacteria in the human intestine, thus giving health benefits to the host. Although the bifidogenic effect of GOS has been demonstrated in numerous studies, the utilization of GOS by specific bifidobacteria remains unclear. The goal of our study was to elucidate GOS consumption by specific bifidobacteria and gain insights into the mechanism. First, we examined GOS consumption by 14 bifidobacterial strains belonging to seven different species by comparing growth rate, carbohydrate consumption, and acid production. We then performed a transcription analysis in the case of one strong GOS consumer, Bifidobacterium adolescentis YIT 4011(T), to predict the operon contributing to GOS use. The study indicated the contribution of an operon consisted of LacS symporter and β-galactosidase to bifidobacterial GOS consumption.
Collapse
Affiliation(s)
- Takuya Akiyama
- a Yakult Honsha European Research Center for Microbiology , Gent-Zwijnaarde , Belgium
| | | | | |
Collapse
|
538
|
Do genetic susceptibility, Toll-like receptors, and pathogen-associated molecular patterns modulate the effects of wear? Clin Orthop Relat Res 2014; 472:3709-17. [PMID: 25034980 PMCID: PMC4397765 DOI: 10.1007/s11999-014-3786-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/31/2023]
Abstract
Overwhelming evidence supports the concept that wear particles are the primary initiator of aseptic loosening of orthopaedic implants. It is likely, however, that other factors modulate the biologic response to wear particles. This review focuses on three potential other factors: genetic susceptibility, Toll-like receptors (TLRs), and bacterial pathogen-associated molecular patterns (PAMPs). WHERE ARE WE NOW?: Considerable evidence is emerging that both genetic susceptibility and TLR activation are important factors that modulate the biologic response to wear particles, but it remains controversial whether bacterial PAMPs also do so. WHERE DO WE NEED TO GO?: Detailed understanding of the roles of these other factors may lead to identification of novel therapeutic targets for patients with aseptic loosening. HOW DO WE GET THERE?: Highest priority should be given to polymorphism replication studies with large numbers of patients and studies to replicate the reported correlation between bacterial biofilms and the severity of aseptic loosening.
Collapse
|
539
|
Mathieu P, Boulanger MC, Després JP. Ectopic visceral fat: a clinical and molecular perspective on the cardiometabolic risk. Rev Endocr Metab Disord 2014; 15:289-98. [PMID: 25326657 DOI: 10.1007/s11154-014-9299-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
Abstract
Worldwide, cardiovascular diseases (CVDs) are a leading cause of mortality. While in many westernized societies there has been a decrease prevalence of smoking and that a special emphasis has been put on the urgency to control the, so called, classical risk factors, it is more and more recognized that there remains a residual risk, which contributes to the development of CVDs. Imaging studies conducted over two decades have highlighted that the accumulation of ectopic visceral fat is associated with a plethora of metabolic dysfunctions, which have complex and intertwined interactions and participate to the development/progression/events of many cardiovascular disorders. The contribution of visceral ectopic fat to the development of coronary artery disease (CAD) is now well established, while in the last several years emerging evidence has pointed out that accumulation of harmful ectopic fat is associated with other cardiovascular disorders such as calcific aortic valve disease (CAVD), atrial fibrillation and left ventricular dysfunction. We review herein the key molecular processes linking the accumulation of ectopic fat to the development of CVDs. We have attempted, whenever possible, to use a translational approach whereby the pathobiology processes are linked to clinical observations.
Collapse
Affiliation(s)
- Patrick Mathieu
- Institut de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada,
| | | | | |
Collapse
|
540
|
LeBlanc ADMD, LeBlanc JG. Effect of probiotic administration on the intestinal microbiota, current knowledge and potential applications. World J Gastroenterol 2014; 20:16518-16528. [PMID: 25469019 PMCID: PMC4248194 DOI: 10.3748/wjg.v20.i44.16518] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/07/2014] [Revised: 04/22/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
Although it is now known that the human body is colonized by a wide variety of microbial populations in different parts (such as the mouth, pharynx and respiratory system, the skin, the gastro- and urogenital tracts), many effects of the complex interactions between the human host and microbial symbionts are still not completely understood. The dysbiosis of the gastrointestinal tract microbiota is considered to be one of the most important contributing factors in the development of many gastrointestinal diseases such as inflammatory bowel disease, irritable bowel syndrome and colorectal cancer, as well as systemic diseases like obesity, diabetes, atherosclerosis and non-alcoholic fatty liver disease. Fecal microbial transplantations appear to be promising therapies for dysbiosis-associated diseases; however, probiotic microorganisms have been growing in popularity due to increasing numbers of studies proving that certain strains present health promoting properties, among them the beneficial balance of the intestinal microbiota. Inflammatory bowel diseases and obesity are the pathologies in which there are more studies showing this beneficial association using animal models and even in human clinical trials. In this review, the association of the human gut microbiota and human health will be discussed along with the benefits that probiotics can confer on this symbiotic activity and on the prevention or treatment of associated diseases.
Collapse
|
541
|
Huang CJ, Stewart JK, Shibata Y, Slusher AL, Acevedo EO. Lipopolysaccharide-binding protein and leptin are associated with stress-induced interleukin-6 cytokine expression ex vivo in obesity. Psychophysiology 2014; 52:687-94. [PMID: 25424507 DOI: 10.1111/psyp.12387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/20/2014] [Accepted: 10/25/2014] [Indexed: 12/25/2022]
Abstract
Obesity is associated with enhanced inflammation and mental stress, but limited information has addressed the potential additive effect of psychological stress on obesity-associated inflammation. This study examined whether obese subjects would elicit a greater host immune response (IL-6 mRNA and cytokine) to lipopolysaccharide (LPS) in response to mental stress. Blood samples for LPS-stimulated IL-6 mRNA and cytokine were collected prior to and following mental stress. Results showed that obese subjects elicited a greater LPS-induced IL-6 along with its mRNA expression following mental stress compared to normal-weight subjects. Stress-induced IL-6 cytokine response to LPS was correlated with the baseline levels of plasma LPS binding protein (LBP) and leptin. These findings are consistent with the idea that endogenous inflammatory agents (e.g., LBP and leptin), often elevated with obesity, enhance inflammatory responses to psychological stress.
Collapse
Affiliation(s)
- Chun-Jung Huang
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, Florida, USA
| | | | | | | | | |
Collapse
|
542
|
Geurts L, Neyrinck AM, Delzenne NM, Knauf C, Cani PD. Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and interventions using prebiotics. Benef Microbes 2014; 5:3-17. [PMID: 23886976 DOI: 10.3920/bm2012.0065] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023]
Abstract
Crosstalk between organs is crucial for controlling numerous homeostatic systems (e.g. energy balance, glucose metabolism and immunity). Several pathological conditions, such as obesity and type 2 diabetes, are characterised by a loss of or excessive inter-organ communication that contributes to the development of disease. Recently, we and others have identified several mechanisms linking the gut microbiota with the development of obesity and associated disorders (e.g. insulin resistance, type 2 diabetes, hepatic steatosis). Among these, we described the concept of metabolic endotoxaemia (increase in plasma lipopolysaccharide levels) as one of the triggering factors leading to the development of metabolic inflammation and insulin resistance. Growing evidence suggests that gut microbes contribute to the onset of low-grade inflammation characterising these metabolic disorders via mechanisms associated with gut barrier dysfunctions. We have demonstrated that enteroendocrine cells (producing glucagon-like peptide-1, peptide YY and glucagon-like peptide-2) and the endocannabinoid system control gut permeability and metabolic endotoxaemia. Recently, we hypothesised that specific metabolic dysregulations occurring at the level of numerous organs (e.g. gut, adipose tissue, muscles, liver and brain) rely from gut microbiota modifications. In this review, we discuss the mechanisms linking gut permeability, adipose tissue metabolism, and glucose homeostasis, and recent findings that show interactions between the gut microbiota, the endocannabinoid system and the apelinergic system. These specific systems are discussed in the context of the gut-to-peripheral organ axis (intestine, adipose tissue and brain) and impacts on metabolic regulation. In the present review, we also briefly describe the impact of a variety of non-digestible nutrients (i.e. inulin-type fructans, arabinoxylans, chitin glucans and polyphenols). Their effects on the composition of the gut microbiota and activity are discussed in the context of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- L Geurts
- WELBIO, Walloon Excellence in Life Sciences and BIOtechnology Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université Catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, 1200 Brussels, Belgium
| | - A M Neyrinck
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université Catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, 1200 Brussels, Belgium
| | - N M Delzenne
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université Catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, 1200 Brussels, Belgium
| | - C Knauf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Team 3, 31432 Toulouse, France Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier, UPS, CHU Rangueil, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | - P D Cani
- WELBIO, Walloon Excellence in Life Sciences and BIOtechnology Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université Catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, 1200 Brussels, Belgium
| |
Collapse
|
543
|
Pepin E, Higa A, Schuster-Klein C, Bernard C, Sulpice T, Guardiola B, Chevet E, Alquier T. Deletion of apoptosis signal-regulating kinase 1 (ASK1) protects pancreatic beta-cells from stress-induced death but not from glucose homeostasis alterations under pro-inflammatory conditions. PLoS One 2014; 9:e112714. [PMID: 25383781 PMCID: PMC4226582 DOI: 10.1371/journal.pone.0112714] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/22/2014] [Accepted: 10/14/2014] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Type 2 diabetes is characterized by pancreatic beta-cell dysfunction and is associated with low-grade inflammation. Recent observations suggest that apoptosis signal-regulating kinase 1 (ASK1) is involved in beta-cell death in response to different stressors. In this study, we tested whether ASK1 deficiency protects beta-cells from glucolipotoxic conditions and cytokines treatment or from glucose homeostasis alteration induced by endotoxemia. METHODOLOGY/PRINCIPAL FINDINGS Insulin secretion was neither affected upon shRNA-mediated downregulation of ASK1 in MIN6 cells nor in islets from ASK1-deficient mice. ASK1 silencing in MIN6 cells and deletion in islets did not prevent the deleterious effect of glucolipotoxic conditions or cytokines on insulin secretion. However, it protected MIN6 cells from death induced by ER stress or palmitate and islets from short term caspase activation in response to cytokines. Moreover, endotoxemia induced by LPS infusion increased insulin secretion during hyperglycemic clamps but the response was similar in wild-type and ASK1-deficient mice. Finally, insulin sensitivity in the presence of LPS was not affected by ASK1-deficiency. CONCLUSIONS/SIGNIFICANCE Our study demonstrates that ASK1 is not involved in beta-cell function and dysfunction but controls stress-induced beta-cell death.
Collapse
Affiliation(s)
| | - Arisa Higa
- Inserm U1053, Team Endoplasmic Reticulum Stress and Cancer, Université de Bordeaux, Bordeaux, France, 33076
| | | | | | - Thierry Sulpice
- Betagenex Inc, Laval, QC, Canada, H7V5B7
- Physiogenex SAS, Labège, France, 31670
| | | | - Eric Chevet
- Inserm U1053, Team Endoplasmic Reticulum Stress and Cancer, Université de Bordeaux, Bordeaux, France, 33076
| | - Thierry Alquier
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), and Departments of Medicine, Biochemistry and Cell Biology and Pathology, Université de Montréal, Montréal, QC, Canada, H3T 1J4
- * E-mail:
| |
Collapse
|
544
|
Mitchell P, Marette A. Statin-induced insulin resistance through inflammasome activation: sailing between Scylla and Charybdis. Diabetes 2014; 63:3569-71. [PMID: 25342725 DOI: 10.2337/db14-1059] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022]
Affiliation(s)
- Patricia Mitchell
- Department of Medicine, Faculty of Medicine, Quebec Heart and Lung Institute, Laval University, Quebec, Quebec, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Quebec Heart and Lung Institute, Laval University, Quebec, Quebec, Canada
| |
Collapse
|
545
|
Zhang D, Zhang L, Zheng Y, Yue F, Russell RD, Zeng Y. Circulating zonulin levels in newly diagnosed Chinese type 2 diabetes patients. Diabetes Res Clin Pract 2014; 106:312-8. [PMID: 25238913 DOI: 10.1016/j.diabres.2014.08.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/04/2014] [Revised: 07/30/2014] [Accepted: 08/22/2014] [Indexed: 01/20/2023]
Abstract
AIMS Studies suggest that type 2 diabetes mellitus is associated with increased gut permeability. Human zonulin is the only physiological mediator discovered to date that is known to regulate gut permeability reversibly by disassembling intestinal tight junctions. However, the relationship between zonulin and type 2 diabetes remains to be defined, and no Chinese population-based data were reported. The aim of this study was to investigate the association between serum zonulin levels and type 2 diabetes in a Chinese Han population. METHODS 143 newly diagnosed type 2 diabetes patients, 124 patients with impaired glucose tolerance and 121 subjects with normal glucose tolerance were enrolled in this study. Serum zonulin was measured by ELISA. RESULTS Patients with type 2 diabetes had higher serum zonulin levels than impaired or normal glucose tolerant subjects. Serum zonulin correlated with body mass index, waist-to-hip ratio, triglyceride, total cholesterol, HDL-C, fasting plasma glucose, 2h plasma glucose, HbA1c, tumor necrosis factor α, interleukin 6, HOMA-IR and QUICK index using correlation analysis (p < 0.05 for all). Multivariate stepwise regression analysis showed that zonulin levels were independently associated with insulin resistance (β = 0.024, p = 0.005). In logistic regression analysis, zonulin levels were an independent predictor of type 2 diabetes (OR = 1.080, p = 0.037). CONCLUSIONS Serum zonulin levels are significantly elevated in newly diagnosed Chinese Type 2 diabetes patients, and are associated with dyslipidemia, inflammation and insulin resistance, indicating a potential role of zonulin in the pathophysiology of type 2 diabetes in Chinese.
Collapse
Affiliation(s)
- D Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China.
| | - L Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - Y Zheng
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - F Yue
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - R D Russell
- School of Nursing, University of California Los Angeles, Los Angeles, CA, USA
| | - Y Zeng
- Department of Endocrinology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| |
Collapse
|
546
|
Intestinal alkaline phosphatase deficiency leads to lipopolysaccharide desensitization and faster weight gain. Infect Immun 2014; 83:247-58. [PMID: 25348635 DOI: 10.1128/iai.02520-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
Abstract
Animals develop in the presence of complex microbial communities, and early host responses to these microbes can influence key aspects of development, such as maturation of the immune system, in ways that impact adult physiology. We previously showed that the zebrafish intestinal alkaline phosphatase (ALPI) gene alpi.1 was induced by Gram-negative bacterium-derived lipopolysaccharide (LPS), a process dependent on myeloid differentiation primary response gene 88 (MYD88), and functioned to detoxify LPS and prevent excessive host inflammatory responses to commensal microbiota in the newly colonized intestine. In the present study, we examined whether the regulation and function of ALPI were conserved in mammals. We found that among the mouse ALPI genes, Akp3 was specifically upregulated by the microbiota, but through a mechanism independent of LPS or MYD88. We showed that disruption of Akp3 did not significantly affect intestinal inflammatory responses to commensal microbiota or animal susceptibility to Yersinia pseudotuberculosis infection. However, we found that Akp3(-/-) mice acquired LPS tolerance during postweaning development, suggesting that Akp3 plays an important role in immune education. Finally, we demonstrated that inhibiting LPS sensing with a mutation in CD14 abrogated the accelerated weight gain in Akp3(-/-) mice receiving a high-fat diet, suggesting that the weight gain is caused by excessive LPS in Akp3(-/-) mice.
Collapse
|
547
|
Abstract
A large body of clinical evidence suggests that the consumption of 3 g or more per d of β-glucan from oats or barley, as part of a diet low in saturated fat and cholesterol, may reduce the risk of CHD. The unique chemical and physical properties of oats and physiological responses to oat consumption contribute to their demonstrated health benefits; other health attributes are still under evaluation. Many of these benefits, such as those associated with a reduced risk of CVD, are codified in health claims by several regulatory agencies, such as the Food and Drug Administration in the USA and the European Food Safety Authority in Europe. Despite these oat–health relationships, an apparent decline in agricultural production, the presence of an array of plant pathogens, and dynamics of climatic conditions may preclude the availability and subsequent consumption of this commodity worldwide. Therefore, it is incumbent on scientists from multiple disciplines to advance research in a spectrum of arenas, including physico-chemical properties of oats, the impact of oats on an array of non-communicable diseases and human microbiome, agricultural practices and environments, and processing technologies that contribute to global food policies.
Collapse
|
548
|
Abstract
The intestines of obese humans and mice are enriched with Erysipelotrichi, a class within the Firmicutes. Clostridium ramosum, a member of the Erysipelotrichi, is associated with symptoms of the metabolic syndrome in humans. To clarify the possible obesogenic potential of this bacterial species and to unravel the underlying mechanism, we investigated the role of C. ramosum in obesity development in gnotobiotic mice. Mice were associated with a simplified human intestinal (SIHUMI) microbiota of eight bacterial species, including C. ramosum, with the SIHUMI microbiota except C. ramosum (SIHUMIw/oCra), or with C. ramosum only (Cra) and fed a high-fat diet (HFD) or a low-fat diet (LFD). Parameters related to the development of obesity and metabolic diseases were compared. After 4 weeks of HFD feeding, the mouse groups did not differ in energy intake, diet digestibility, gut permeability, and parameters of low-grade inflammation. However, SIHUMI and Cra mice fed the HFD gained significantly more body weight and body fat and displayed higher food efficiency than SIHUMIw/oCra mice fed the HFD. Gene expression of glucose transporter 2 (Glut2) in jejunal mucosa and of fatty acid translocase (CD36) in ileal mucosa was significantly increased in the obese SIHUMI and Cra mice compared with the less obese SIHUMIw/oCra mice. The data demonstrate that the presence of C. ramosum in SIHUMI and Cra mice enhanced diet-induced obesity. Upregulation of small intestinal glucose and fat transporters in these animals may contribute to their increased body fat deposition. Obesity is a growing health problem worldwide. Changes in the proportions of Bacteroidetes and Firmicutes, the two dominant phyla in the human and the murine intestinal tract, link the intestinal microbiota to obesity. Erysipelotrichi, a class within the Firmicutes, increase in response to high-fat feeding in mice. Clostridium ramosum, a member of the Erysipelotrichi, has been linked to symptoms of the metabolic syndrome. We hypothesized that C. ramosum promotes obesity development and related pathologies. Our experiments in gnotobiotic mice show that C. ramosum promoted diet-induced obesity, probably by enhancing nutrient absorption. Identification of obesogenic bacteria and understanding their mode of action enable the development of novel strategies for the treatment of this epidemic disease. Pharmaceuticals that target obesogenic bacteria or their metabolism could help to prevent and treat obesity and related disorders in the future.
Collapse
|
549
|
Sato J, Kanazawa A, Ikeda F, Yoshihara T, Goto H, Abe H, Komiya K, Kawaguchi M, Shimizu T, Ogihara T, Tamura Y, Sakurai Y, Yamamoto R, Mita T, Fujitani Y, Fukuda H, Nomoto K, Takahashi T, Asahara T, Hirose T, Nagata S, Yamashiro Y, Watada H. Gut dysbiosis and detection of "live gut bacteria" in blood of Japanese patients with type 2 diabetes. Diabetes Care 2014; 37:2343-50. [PMID: 24824547 DOI: 10.2337/dc13-2817] [Citation(s) in RCA: 290] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Mounting evidence indicates that the gut microbiota are an important modifier of obesity and diabetes. However, so far there is no information on gut microbiota and "live gut bacteria" in the systemic circulation of Japanese patients with type 2 diabetes. RESEARCH DESIGN AND METHODS Using a sensitive reverse transcription-quantitative PCR (RT-qPCR) method, we determined the composition of fecal gut microbiota in 50 Japanese patients with type 2 diabetes and 50 control subjects, and its association with various clinical parameters, including inflammatory markers. We also analyzed the presence of gut bacteria in blood samples. RESULTS The counts of the Clostridium coccoides group, Atopobium cluster, and Prevotella (obligate anaerobes) were significantly lower (P < 0.05), while the counts of total Lactobacillus (facultative anaerobes) were significantly higher (P < 0.05) in fecal samples of diabetic patients than in those of control subjects. Especially, the counts of Lactobacillus reuteri and Lactobacillus plantarum subgroups were significantly higher (P < 0.05). Gut bacteria were detected in blood at a significantly higher rate in diabetic patients than in control subjects (28% vs. 4%, P < 0.01), and most of these bacteria were Gram-positive. CONCLUSIONS This is the first report of gut dysbiosis in Japanese patients with type 2 diabetes as assessed by RT-qPCR. The high rate of gut bacteria in the circulation suggests translocation of bacteria from the gut to the bloodstream.
Collapse
Affiliation(s)
- Junko Sato
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akio Kanazawa
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, JapanCenter for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fuki Ikeda
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoaki Yoshihara
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiromasa Goto
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroko Abe
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koji Komiya
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Minako Kawaguchi
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoaki Shimizu
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takeshi Ogihara
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshifumi Tamura
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, JapanSportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuko Sakurai
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Risako Yamamoto
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoya Mita
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshio Fujitani
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, JapanCenter for Beta Cell Biology and Regeneration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Fukuda
- Department of General Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koji Nomoto
- Yakult Central Institute for Microbiological Research, Tokyo, Japan
| | - Takuya Takahashi
- Yakult Central Institute for Microbiological Research, Tokyo, Japan
| | - Takashi Asahara
- Yakult Central Institute for Microbiological Research, Tokyo, Japan
| | - Takahisa Hirose
- Department of Metabolism & Endocrinology, Toho University School of Medicine, Tokyo, Japan
| | - Satoru Nagata
- Probiotics Research Laboratory, Juntendo University Graduate School of Medicine, Tokyo, JapanDepartment of Pediatrics, Tokyo Women's Medical University, Tokyo, Japan
| | - Yuichiro Yamashiro
- Probiotics Research Laboratory, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, JapanCenter for Beta Cell Biology and Regeneration, Juntendo University Graduate School of Medicine, Tokyo, JapanCenter for Molecular Diabetology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
550
|
Napolitano A, Miller S, Nicholls AW, Baker D, Van Horn S, Thomas E, Rajpal D, Spivak A, Brown JR, Nunez DJ. Novel gut-based pharmacology of metformin in patients with type 2 diabetes mellitus. PLoS One 2014; 9:e100778. [PMID: 24988476 PMCID: PMC4079657 DOI: 10.1371/journal.pone.0100778] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2014] [Accepted: 05/23/2014] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Metformin, a biguanide derivate, has pleiotropic effects beyond glucose reduction, including improvement of lipid profiles and lowering microvascular and macrovascular complications associated with type 2 diabetes mellitus (T2DM). These effects have been ascribed to adenosine monophosphate-activated protein kinase (AMPK) activation in the liver and skeletal muscle. However, metformin effects are not attenuated when AMPK is knocked out and intravenous metformin is less effective than oral medication, raising the possibility of important gut pharmacology. We hypothesized that the pharmacology of metformin includes alteration of bile acid recirculation and gut microbiota resulting in enhanced enteroendocrine hormone secretion. In this study we evaluated T2DM subjects on and off metformin monotherapy to characterize the gut-based mechanisms of metformin. Subjects were studied at 4 time points: (i) at baseline on metformin, (ii) 7 days after stopping metformin, (iii) when fasting blood glucose (FBG) had risen by 25% after stopping metformin, and (iv) when FBG returned to baseline levels after restarting the metformin. At these timepoints we profiled glucose, insulin, gut hormones (glucagon-like peptide-1 (GLP-1), peptide tyrosine-tyrosine (PYY) and glucose-dependent insulinotropic peptide (GIP) and bile acids in blood, as well as duodenal and faecal bile acids and gut microbiota. We found that metformin withdrawal was associated with a reduction of active and total GLP-1 and elevation of serum bile acids, especially cholic acid and its conjugates. These effects reversed when metformin was restarted. Effects on circulating PYY were more modest, while GIP changes were negligible. Microbiota abundance of the phylum Firmicutes was positively correlated with changes in cholic acid and conjugates, while Bacteroidetes abundance was negatively correlated. Firmicutes and Bacteroidetes representation were also correlated with levels of serum PYY. Our study suggests that metformin has complex effects due to gut-based pharmacology which might provide insights into novel therapeutic approaches to treat T2DM and associated metabolic diseases. TRIAL REGISTRATION www.ClinicalTrials.gov NCT01357876.
Collapse
Affiliation(s)
| | - Sam Miller
- Quantitative Sciences, GSK R&D, Stevenage, Herts, United Kingdom
| | | | - David Baker
- Safety Assessment, GSK R&D, Ware, Herts, United Kingdom
| | - Stephanie Van Horn
- Target and Pathways Validation, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Elizabeth Thomas
- Target and Pathways Validation, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Deepak Rajpal
- Computational Biology, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Aaron Spivak
- Computational Biology, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - James R. Brown
- Computational Biology, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Derek J. Nunez
- Enteroendocrine Discovery Unit, GlaxoSmithKline R&D, GSK R&D, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|