2901
|
Kanther M, Tomkovich S, Xiaolun S, Grosser MR, Koo J, Flynn EJ, Jobin C, Rawls JF. Commensal microbiota stimulate systemic neutrophil migration through induction of serum amyloid A. Cell Microbiol 2014; 16:1053-67. [PMID: 24373309 DOI: 10.1111/cmi.12257] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 12/13/2013] [Accepted: 12/18/2013] [Indexed: 12/18/2022]
Abstract
Neutrophils serve critical roles in inflammatory responses to infection and injury, and mechanisms governing their activity represent attractive targets for controlling inflammation. The commensal microbiota is known to regulate the activity of neutrophils and other leucocytes in the intestine, but the systemic impact of the microbiota on neutrophils remains unknown. Here we utilized in vivo imaging in gnotobiotic zebrafish to reveal diverse effects of microbiota colonization on systemic neutrophil development and function. The presence of a microbiota resulted in increased neutrophil number and myeloperoxidase expression, and altered neutrophil localization and migratory behaviours. These effects of the microbiota on neutrophil homeostasis were accompanied by an increased recruitment of neutrophils to injury. Genetic analysis identified the microbiota-induced acute phase protein serum amyloid A (Saa) as a host factor mediating microbial stimulation of tissue-specific neutrophil migratory behaviours. In vitro studies revealed that zebrafish cells respond to Saa exposure by activating NF-κB, and that Saa-dependent neutrophil migration requires NF-κB-dependent gene expression. These results implicate the commensal microbiota as an important environmental factor regulating diverse aspects of systemic neutrophil development and function, and reveal a critical role for a Saa-NF-κB signalling axis in mediating neutrophil migratory responses.
Collapse
Affiliation(s)
- Michelle Kanther
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
2902
|
Werner C, Schubbert A, Schrödl W, Krüger M, Sundrum A. Effects of feeding different roughage components to sows in gestation on bacteriological and immunological parameters in colostrum and immune response of piglets. Arch Anim Nutr 2014; 68:29-41. [PMID: 24444334 DOI: 10.1080/1745039x.2013.876184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In piglet production, Escherichia coli (E. coli) and the presence of lipopolysaccharides (LPS) were considered as predominant causative agents in the aetiology of important diseases of sows and piglets. The aim of the study was to assess the effects of feeding different roughage sources to sows in gestation on the microbiota and endotoxin concentration in colostrum and on the specific immune response of their piglets. In two trial runs, a total of 144 sows were assigned to one of the six dietary treatments: the Control group receiving a concentrate diet and five groups with a restrictive supply of the control diet in combination with straw (S), hay (H), clover grass silage (GS), maize silage (MS) or Jerusalem artichoke (JA). During lactation no roughage was offered. Colostrum samples were analysed for bacteriological and immunological parameters. Blood samples were taken from two piglets per corresponding litter to examine the concentration of C-reactive protein (CRP) and of specific antibodies to LPS of E. coli (strain J5). Roughage feeding had an effect neither on the total bacterial count nor on the content of E. coli in colostrum of sows. The concentration of LPS in colostrum was reduced in Group MS. The concentration of CRP in the colostrum samples was low and independent of the feeding regime (max. 9.3 µg/ml). However, the administration of roughage components rich in crude fibre (Groups H and S) decreased the level of CRP in colostrum significantly. The analysis of the specific immunoglobulin to LPS of E. coli (strain J5) showed a higher concentration of IgG-anti-LPS in blood serum of piglets from sows of Group JA. The results suggest that the inclusion of selected roughage components may have the potential to affect the immunocompetence of sows and their corresponding piglets.
Collapse
Affiliation(s)
- Christina Werner
- a Department of Animal Nutrition and Animal Health, Faculty of Organic Agricultural Sciences , University of Kassel , Witzenhausen , Germany
| | | | | | | | | |
Collapse
|
2903
|
Xiao C, Dash S, Morgantini C, Lewis GF. New and emerging regulators of intestinal lipoprotein secretion. Atherosclerosis 2014; 233:608-615. [PMID: 24534456 DOI: 10.1016/j.atherosclerosis.2013.12.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/12/2013] [Accepted: 12/31/2013] [Indexed: 12/25/2022]
Abstract
Overproduction of hepatic apoB100-containing VLDL particles has been well documented in animal models and in humans with insulin resistance such as the metabolic syndrome and type 2 diabetes, and contributes to the typical dyslipidemia of these conditions. In addition, postprandial hyperlipidemia and elevated plasma concentrations of intestinal apoB48-containing chylomicron and chylomicron remnant particles have been demonstrated in insulin resistant states. Intestinal lipoprotein production is primarily determined by the amount of fat ingested and absorbed. Until approximately 10 years ago, however, relatively little attention was paid to the role of the intestine itself in regulating the production of triglyceride-rich lipoproteins (TRL) and its dysregulation in pathological states such as insulin resistance. We and others have shown that insulin resistant animal models and humans are characterized by overproduction of intestinal apoB48-containing lipoproteins. Whereas various factors are known to regulate hepatic lipoprotein particle production, less is known about factors that regulate the production of intestinal lipoprotein particles. Monosacharides, plasma free fatty acids (FFA), resveratrol, intestinal peptides (e.g. GLP-1 and GLP-2), and pancreatic hormones (e.g. insulin) have recently been shown to be important regulators of intestinal lipoprotein secretion. Available evidence in humans and animal models strongly supports the concept that the small intestine is not merely an absorptive organ but rather plays an active role in regulating the rate of production of chylomicrons in fed and fasting states. Metabolic signals in insulin resistance and type 2 diabetes and in some cases an aberrant intestinal response to these factors contribute to the enhanced formation and secretion of TRL. Understanding the regulation of intestinal lipoprotein production is imperative for the development of new therapeutic strategies for the prevention and treatment of dyslipidemia. Here we review recent developments in this field and present evidence that intestinal lipoprotein production is a process with metabolic plasticity and that modulation of intestinal lipoprotein secretion may be a feasible therapeutic strategy in the treatment of dyslipidemia and possibly prevention of atherosclerosis.
Collapse
Affiliation(s)
- Changting Xiao
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada
| | - Satya Dash
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada
| | - Cecilia Morgantini
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada
| | - Gary F Lewis
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
2904
|
Robinson CM, Jesudhasan PR, Pfeiffer JK. Bacterial lipopolysaccharide binding enhances virion stability and promotes environmental fitness of an enteric virus. Cell Host Microbe 2014; 15:36-46. [PMID: 24439896 PMCID: PMC3920179 DOI: 10.1016/j.chom.2013.12.004] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/02/2013] [Accepted: 12/11/2013] [Indexed: 02/08/2023]
Abstract
Enteric viruses, including poliovirus and reovirus, encounter a vast microbial community in the mammalian gastrointestinal tract, which has been shown to promote virus replication and pathogenesis. Investigating the underlying mechanisms, we find that poliovirus binds bacterial surface polysaccharides, which enhances virion stability and cell attachment by increasing binding to the viral receptor. Additionally, we identified a poliovirus mutant, VP1-T99K, with reduced lipopolysaccharide (LPS) binding. Although T99K and WT poliovirus cell attachment, replication, and pathogenesis in mice are equivalent, VP1-T99K poliovirus was unstable in feces following peroral inoculation of mice. Consequently, the ratio of mutant virus in feces is reduced following additional cycles of infection in mice. Thus, the mutant virus incurs a fitness cost when environmental stability is a factor. These data suggest that poliovirus binds bacterial surface polysaccharides, enhancing cell attachment and environmental stability, potentially promoting transmission to a new host.
Collapse
Affiliation(s)
- Christopher M Robinson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Palmy R Jesudhasan
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Julie K Pfeiffer
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
2905
|
Beebe K, Sampey B, Watkins SM, Milburn M, Eckhart AD. Understanding the apothecaries within: the necessity of a systematic approach for defining the chemical output of the human microbiome. Clin Transl Sci 2014; 7:74-81. [PMID: 24422665 DOI: 10.1111/cts.12131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The human microbiome harbors a massive diversity of microbes that effectively form an "organ" that strongly influences metabolism and immune function and hence, human health. Although the growing interest in the microbiome has chiefly arisen due to its impact on human physiology, the probable rules of operation are embedded in the roots of microbiology where chemical communication (i.e., with metabolites) is a dominant feature of coexistence. Indeed, recent examples in microbiome research offer the impression that the collective microbiome operates as an "apothecary," creating chemical concoctions that influence health and alter drug response. Although these principles are not unappreciated, the majority of emphasis is on metagenomics and research efforts often omit systematic efforts to interrogate the chemical component of the complex equation between microbial community and host phenotype. One of the reasons for this omission may be due to the inaccessibility to high-breadth, high-throughput, and scalable technologies. Since these technologies are now available, we propose that a more systematic effort to survey the host-microbiota chemical output be embedded into microbiome research as there is strong likelihood, and growing precedence, that this component may often be integral to developing our understanding of these ultimate apothecaries and how they impact human health.
Collapse
Affiliation(s)
- Kirk Beebe
- Metabolon Inc, Durham, North Carolina, USA
| | | | | | | | | |
Collapse
|
2906
|
Bekkering P, Jafri I, van Overveld FJ, Rijkers GT. The intricate association between gut microbiota and development of Type 1, Type 2 and Type 3 diabetes. Expert Rev Clin Immunol 2014; 9:1031-41. [DOI: 10.1586/1744666x.2013.848793] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
2907
|
Campbell C, Grapov D, Fiehn O, Chandler CJ, Burnett DJ, Souza EC, Casazza GA, Gustafson MB, Keim NL, Newman JW, Hunter GR, Fernandez JR, Garvey WT, Harper ME, Hoppel CL, Meissen JK, Take K, Adams SH. Improved metabolic health alters host metabolism in parallel with changes in systemic xeno-metabolites of gut origin. PLoS One 2014; 9:e84260. [PMID: 24416208 PMCID: PMC3885560 DOI: 10.1371/journal.pone.0084260] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/13/2013] [Indexed: 12/18/2022] Open
Abstract
Novel plasma metabolite patterns reflective of improved metabolic health (insulin sensitivity, fitness, reduced body weight) were identified before and after a 14–17 wk weight loss and exercise intervention in sedentary, obese insulin-resistant women. To control for potential confounding effects of diet- or microbiome-derived molecules on the systemic metabolome, sampling was during a tightly-controlled feeding test week paradigm. Pairwise and multivariate analysis revealed intervention- and insulin-sensitivity associated: (1) Changes in plasma xeno-metabolites (“non-self” metabolites of dietary or gut microbial origin) following an oral glucose tolerance test (e.g. higher post-OGTT propane-1,2,3-tricarboxylate [tricarballylic acid]) or in the overnight-fasted state (e.g., lower γ-tocopherol); (2) Increased indices of saturated very long chain fatty acid elongation capacity; (3) Increased post-OGTT α-ketoglutaric acid (α-KG), fasting α-KG inversely correlated with Matsuda index, and altered patterns of malate, pyruvate and glutamine hypothesized to stem from improved mitochondrial efficiency and more robust oxidation of glucose. The results support a working model in which improved metabolic health modifies host metabolism in parallel with altering systemic exposure to xeno-metabolites. This highlights that interpretations regarding the origins of peripheral blood or urinary “signatures” of insulin resistance and metabolic health must consider the potentially important contribution of gut-derived metabolites toward the host's metabolome.
Collapse
Affiliation(s)
- Caitlin Campbell
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Dmitry Grapov
- West Coast Metabolomics Center, University of California Davis, Davis, California, United States of America
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, California, United States of America
- Genome Center, University of California Davis, Davis, California, United States of America
- * E-mail: (SHA); (OF)
| | - Carol J. Chandler
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Dustin J. Burnett
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Elaine C. Souza
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Gretchen A. Casazza
- Sports Medicine Program, University of California, Davis School of Medicine, Sacramento, California, United States of America
| | - Mary B. Gustafson
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
| | - Nancy L. Keim
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
- Department of Nutrition, University of California Davis, Davis, California, United States of America
| | - John W. Newman
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
- Department of Nutrition, University of California Davis, Davis, California, United States of America
| | - Gary R. Hunter
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama, United States of America
- Human Studies Department, University of Alabama, Birmingham, Alabama, United States of America
| | - Jose R. Fernandez
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama, United States of America
| | - W. Timothy Garvey
- Department of Nutrition Sciences, University of Alabama, Birmingham, Alabama, United States of America
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Charles L. Hoppel
- Pharmacology Department, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - John K. Meissen
- Genome Center, University of California Davis, Davis, California, United States of America
| | - Kohei Take
- Genome Center, University of California Davis, Davis, California, United States of America
| | - Sean H. Adams
- USDA-ARS Western Human Nutrition Research Center, Davis, California, United States of America
- Department of Nutrition, University of California Davis, Davis, California, United States of America
- * E-mail: (SHA); (OF)
| |
Collapse
|
2908
|
Mikami Y, Mizuno S, Nakamoto N, Hayashi A, Sujino T, Sato T, Kamada N, Matsuoka K, Hisamatsu T, Ebinuma H, Hibi T, Yoshimura A, Kanai T. Macrophages and dendritic cells emerge in the liver during intestinal inflammation and predispose the liver to inflammation. PLoS One 2014; 9:e84619. [PMID: 24392145 PMCID: PMC3879334 DOI: 10.1371/journal.pone.0084619] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/25/2013] [Indexed: 12/15/2022] Open
Abstract
The liver is a physiological site of immune tolerance, the breakdown of which induces immunity. Liver antigen-presenting cells may be involved in both immune tolerance and activation. Although inflammatory diseases of the liver are frequently associated with inflammatory bowel diseases, the underlying immunological mechanisms remain to be elucidated. Here we report two murine models of inflammatory bowel disease: RAG-2−/− mice adoptively transferred with CD4+CD45RBhigh T cells; and IL-10−/− mice, accompanied by the infiltration of mononuclear cells in the liver. Notably, CD11b−CD11clowPDCA-1+ plasmacytoid dendritic cells (DCs) abundantly residing in the liver of normal wild-type mice disappeared in colitic CD4+CD45RBhigh T cell-transferred RAG-2−/− mice and IL-10−/− mice in parallel with the emergence of macrophages (Mφs) and conventional DCs (cDCs). Furthermore, liver Mφ/cDCs emerging during intestinal inflammation not only promote the proliferation of naïve CD4+ T cells, but also instruct them to differentiate into IFN-γ-producing Th1 cells in vitro. The emergence of pathological Mφ/cDCs in the liver also occurred in a model of acute dextran sulfate sodium (DSS)-induced colitis under specific pathogen-free conditions, but was canceled in germ-free conditions. Last, the Mφ/cDCs that emerged in acute DSS colitis significantly exacerbated Fas-mediated hepatitis. Collectively, intestinal inflammation skews the composition of antigen-presenting cells in the liver through signaling from commensal bacteria and predisposes the liver to inflammation.
Collapse
Affiliation(s)
- Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Shinta Mizuno
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Hayashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Research Laboratory, Miyarisan Pharmaceutical, Tokyo, Japan
| | - Tomohisa Sujino
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiko Kamada
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Katsuyoshi Matsuoka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tadakazu Hisamatsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirotoshi Ebinuma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshifumi Hibi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- * E-mail: (TK); (AY)
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- * E-mail: (TK); (AY)
| |
Collapse
|
2909
|
Effect of bacteria used in food industry on the proliferation and cytokine production of epithelial intestinal cellular lines. J Funct Foods 2014. [DOI: 10.1016/j.jff.2013.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
2910
|
Neuropeptides and the microbiota-gut-brain axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:195-219. [PMID: 24997035 DOI: 10.1007/978-1-4939-0897-4_9] [Citation(s) in RCA: 289] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropeptides are important mediators both within the nervous system and between neurons and other cell types. Neuropeptides such as substance P, calcitonin gene-related peptide and neuropeptide Y (NPY), vasoactive intestinal polypeptide, somatostatin and corticotropin-releasing factor are also likely to play a role in the bidirectional gut-brain communication. In this capacity they may influence the activity of the gastrointestinal microbiota and its interaction with the gut-brain axis. Current efforts in elucidating the implication of neuropeptides in the microbiota-gut-brain axis address four information carriers from the gut to the brain (vagal and spinal afferent neurons; immune mediators such as cytokines; gut hormones; gut microbiota-derived signalling molecules) and four information carriers from the central nervous system to the gut (sympathetic efferent neurons; parasympathetic efferent neurons; neuroendocrine factors involving the adrenal medulla; neuroendocrine factors involving the adrenal cortex). Apart from operating as neurotransmitters, many biologically active peptides also function as gut hormones. Given that neuropeptides and gut hormones target the same cell membrane receptors (typically G protein-coupled receptors), the two messenger roles often converge in the same or similar biological implications. This is exemplified by NPY and peptide YY (PYY), two members of the PP-fold peptide family. While PYY is almost exclusively expressed by enteroendocrine cells, NPY is found at all levels of the gut-brain and brain-gut axis. The function of PYY-releasing enteroendocrine cells is directly influenced by short chain fatty acids generated by the intestinal microbiota from indigestible fibre, while NPY may control the impact of the gut microbiota on inflammatory processes, pain, brain function and behaviour. Although the impact of neuropeptides on the interaction between the gut microbiota and brain awaits to be analysed, biologically active peptides are likely to emerge as neural and endocrine messengers in orchestrating the microbiota-gut-brain axis in health and disease.
Collapse
|
2911
|
Qin C, Xu L, Yang Y, He S, Dai Y, Zhao H, Zhou Z. Comparison of fecundity and offspring immunity in zebrafish fed Lactobacillus rhamnosus CICC 6141 and Lactobacillus casei BL23. Reproduction 2014; 147:53-64. [DOI: 10.1530/rep-13-0141] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To increase the knowledge of probiotic effects on zebrafish (Danio rerio), we compare the effects of two probiotic strains, Lactobacillus rhamnosus CICC 6141 (a highly adhesive strain) and Lactobacillus casei BL23 (a weakly adhesive strain), on zebrafish reproduction and their offsprings' innate level of immunity to water-borne pathogens. During probiotics treatments from 7 to 28 days, both the Lactobacillus strains, and especially L. casei BL23, significantly increased fecundity in zebrafish: higher rates of egg ovulation, fertilization, and hatching were observed. Increased densities of both small and large vitellogenic follicles, seen in specimens fed either Lactobacillus strain, demonstrated accelerated oocyte maturation. Feeding either strain of Lactobacillus upregulated gene expression of leptin, kiss2, gnrh3, fsh, lh, lhcgr, and paqr8, which were regarded to enhance fecundity and encourage oocyte maturation. Concomitantly, the gene expression of bmp15 and tgfb1 was inhibited, which code for local factors that prevent oocyte maturation. The beneficial effects of the Lactobacillus strains on fecundity diminished after feeding of the probiotics was discontinued, even for the highly adhesive gut Lactobacillus strain. Administering L. rhamnosus CICC 6141 for 28 days was found to affect the innate immunity of offspring derived from their parents, as evinced by a lower level of alkaline phosphatase activity in early larval stages. This study highlights the effects of probiotics both upon the reproductive process and upon the offsprings' immunity during early developmental stages.
Collapse
|
2912
|
Ahmer BMM. In this issue of Gut Microbes. Gut Microbes 2014; 5:83-5. [PMID: 24468723 PMCID: PMC4049943 DOI: 10.4161/gmic.28007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
2913
|
Lee KA, Lee WJ. Drosophila as a model for intestinal dysbiosis and chronic inflammatory diseases. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 42:102-10. [PMID: 23685204 DOI: 10.1016/j.dci.2013.05.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 05/07/2013] [Accepted: 05/07/2013] [Indexed: 05/14/2023]
Abstract
The association between deregulated intestinal microbial consortia and host diseases has been recognized since the birth of microbiology over a century ago. Intestinal dysbiosis refers to a state where living metazoans harbor harmful intestinal microflora. However, there is still an issue of whether causality arises from the host or the microbe because it is unclear whether deregulation of the gut microbiota community is the consequence or cause of the host disease. Recent studies using Drosophila and its simple microbiota have provided a valuable model system for dissecting the molecular mechanisms of intestinal dysbiosis. In this review, we examine recent exciting observations in Drosophila gut-microbiota interactions, particularly the links among the host immune genotype, the microbial community structure, and the host inflammatory phenotype. Future genetic analyses using Drosophila model system will provide a valuable outcome for understanding the evolutionarily conserved mechanisms that underlie intestinal dysbiosis and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Kyung-Ah Lee
- School of Biological Science, Seoul National University and National Creative Research Initiative Center for Symbiosystem, Seoul 151-742, South Korea
| | | |
Collapse
|
2914
|
Ettinger G, MacDonald K, Reid G, Burton JP. The influence of the human microbiome and probiotics on cardiovascular health. Gut Microbes 2014; 5:719-28. [PMID: 25529048 PMCID: PMC4615746 DOI: 10.4161/19490976.2014.983775] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/13/2014] [Accepted: 10/29/2014] [Indexed: 02/03/2023] Open
Abstract
Cardiovascular disease (CVD) is a major cause of death worldwide. Of the many etiological factors, microorganisms constitute one. From the local impact of the gut microbiota on energy metabolism and obesity, to the distal association of periodontal disease with coronary heart disease, microbes have a significant impact on cardiovascular health. In terms of the ability to modulate or influence the microbes, probiotic applications have been considered. These are live microorganisms which when administered in adequate amounts confer a benefit on the host. While a number of reports have established the beneficial abilities of certain probiotic bacterial strains to reduce cholesterol and hypertension, recent research suggests that their use could be more widely applied. This review presents an up-to-date summary of the known associations of the microbiome with CVD, and potential applications of probiotic therapy.
Collapse
Key Words
- ACE, Angiotensin converting enzyme
- ASD, Autism Spectrum Disorder
- BSH, Bile salt hydrolase
- CLA, Conjugate linoleic acid
- CRP, C-reactive protein
- CVD, Cardiovascular disease
- HSP, Heat shock protein
- I/R, Ischemia/reperfusion
- LDL-C Low density lipoprotein cholesterol
- PD, Periodontal disease
- TLR, Toll-like receptor
- TMA, Trimethylamine
- TMAO, Trimethylamine-N-oxide
- cardioprotection
- cardiovascular disease
- dysbiosis
- microbiome
- periodontal disease
- probiotics
Collapse
Affiliation(s)
- Grace Ettinger
- Canadian Center for Human Microbiome and Probiotic Research; Lawson Health Research Institute; London, Ontario, Canada
- Department of Microbiology and Immunology; Western University; London, Ontario, Canada
| | - Kyle MacDonald
- Canadian Center for Human Microbiome and Probiotic Research; Lawson Health Research Institute; London, Ontario, Canada
- Department of Microbiology and Immunology; Western University; London, Ontario, Canada
| | - Gregor Reid
- Canadian Center for Human Microbiome and Probiotic Research; Lawson Health Research Institute; London, Ontario, Canada
- Department of Microbiology and Immunology; Western University; London, Ontario, Canada
- Department of Surgery; Western University; London, Ontario, Canada
| | - Jeremy P Burton
- Canadian Center for Human Microbiome and Probiotic Research; Lawson Health Research Institute; London, Ontario, Canada
- Department of Microbiology and Immunology; Western University; London, Ontario, Canada
- Division of Urology, Department of Surgery; Western University; London, Ontario, Canada
| |
Collapse
|
2915
|
Palacios T, Coulson S, Butt H, Vitetta L. The gastrointestinal microbiota and multi-strain probiotic therapy: In children and adolescent obesity. ADVANCES IN INTEGRATIVE MEDICINE 2014. [DOI: 10.1016/j.aimed.2013.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
2916
|
Abstract
The colon serves as the habitat for trillions of microbes, which it must maintain, regulate, and sequester. This is managed by what is termed the mucosal barrier. The mucosal barrier separates the gut flora from the host tissues; regulates the absorption of water, electrolytes, minerals, and vitamins; and facilitates host-flora interactions. Colonic homeostasis depends on a complex interaction between the microflora and the mucosal epithelium, immune system, vasculature, stroma, and nervous system. Disruptions in the colonic microenvironment such as changes in microbial composition, epithelial cell function/proliferation/differentiation, mucus production/makeup, immune function, diet, motility, or blood flow may have substantial local and systemic consequences. Understanding the complex activities of the colon in health and disease is important in drug development, as xenobiotics can impact all segments of the colon. Direct and indirect effects of pharmaceuticals on intestinal function can produce adverse findings in laboratory animals and humans and can negatively impact drug development. This review will discuss normal colon homeostasis with examples, where applicable, of xenobiotics that disrupt normal function.
Collapse
Affiliation(s)
- Rani S Sellers
- 1Albert Einstein College of Medicine, Bronx, New York, USA
| | | |
Collapse
|
2917
|
Dowds CM, Kornell SC, Blumberg RS, Zeissig S. Lipid antigens in immunity. Biol Chem 2014; 395:61-81. [PMID: 23999493 PMCID: PMC4128234 DOI: 10.1515/hsz-2013-0220] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 08/27/2013] [Indexed: 02/07/2023]
Abstract
Lipids are not only a central part of human metabolism but also play diverse and critical roles in the immune system. As such, they can act as ligands of lipid-activated nuclear receptors, control inflammatory signaling through bioactive lipids such as prostaglandins, leukotrienes, lipoxins, resolvins, and protectins, and modulate immunity as intracellular phospholipid- or sphingolipid-derived signaling mediators. In addition, lipids can serve as antigens and regulate immunity through the activation of lipid-reactive T cells, which is the topic of this review. We will provide an overview of the mechanisms of lipid antigen presentation, the biology of lipid-reactive T cells, and their contribution to immunity.
Collapse
Affiliation(s)
- C. Marie Dowds
- Department of Internal Medicine I, University Medical Center
Schleswig-Holstein, Schittenhelmstraße 12, D-24105 Kiel,
Germany
| | - Sabin-Christin Kornell
- Department of Internal Medicine I, University Medical Center
Schleswig-Holstein, Schittenhelmstraße 12, D-24105 Kiel,
Germany
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham
and Women’s Hospital, Harvard Medical School, 75 Francis Street,
Boston, MA 02115, USA
| | - Sebastian Zeissig
- Department of Internal Medicine I, University Medical Center
Schleswig-Holstein, Schittenhelmstraße 12, D-24105 Kiel,
Germany
| |
Collapse
|
2918
|
Kozuka C, Yabiku K, Takayama C, Matsushita M, Shimabukuro M. Natural food science based novel approach toward prevention and treatment of obesity and type 2 diabetes: recent studies on brown rice and γ-oryzanol. Obes Res Clin Pract 2013; 7:e165-72. [PMID: 23697584 DOI: 10.1016/j.orcp.2013.02.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/14/2013] [Accepted: 02/25/2013] [Indexed: 02/07/2023]
Abstract
The prevalences of obesity and type 2 diabetes mellitus are dramatically increasing, and there is a strong need for more effective and safer therapies. However, some of drugs show limited efficacy and considerable adverse effects. Furthermore, artificial energy-dense foods and non-caloric foods may promote overeating and weight gain. In this context, a natural food-based approach may represent a valuable means of tackling the obesity-diabetes syndrome. Although recent studies have shown that brown rice improves glucose intolerance and prevents obesity and type 2 diabetes in humans, the underlying molecular mechanisms remain unclear. We found that one of the major components of brown rice, γ-oryzanol (Orz), plays an important role in the metabolically beneficial effects of brown rice. Orz acts as a chemical chaperone and decreases high fat diet (HFD)-induced endoplasmic reticulum (ER) stress in the hypothalamus, thereby leading to a significant shift in preference from fatty to healthy foods. Orz also decreases HFD-induced ER stress in pancreatic β-cells and improves β-cell function. Notably, Orz directly acts on pancreatic islets and enhances glucose-stimulated insulin secretion (GSIS). This evidence highlights food preference as a promising therapeutic target in obesity-diabetes syndrome and suggests that brown rice and Orz may have potential for the treatment of obesity and type 2 diabetes in humans.
Collapse
Affiliation(s)
- Chisayo Kozuka
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | | | | | | | | |
Collapse
|
2919
|
Pinel A, Morio-Liondore B, Capel F. n−3 polyunsaturated fatty acids modulate metabolism of insulin-sensitive tissues: implication for the prevention of type 2 diabetes. J Physiol Biochem 2013; 70:647-58. [DOI: 10.1007/s13105-013-0303-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 12/02/2013] [Indexed: 12/21/2022]
|
2920
|
Candela M, Biagi E, Brigidi P, O'Toole PW, De Vos WM. Maintenance of a healthy trajectory of the intestinal microbiome during aging: a dietary approach. Mech Ageing Dev 2013; 136-137:70-5. [PMID: 24373997 DOI: 10.1016/j.mad.2013.12.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 12/05/2013] [Accepted: 12/16/2013] [Indexed: 12/21/2022]
Abstract
Sharing an intense transgenomic metabolism with the host, the intestinal microbiota is an essential factor for several aspects of the human physiology. However, several age-related factors, such as changes diet, lifestyle, inflammation and frailty, force the deterioration of this intestinal microbiota-host mutualistic interaction, compromising the possibility to reach longevity. In this scenario, the NU-AGE project involves the development of dietary interventions specifically tailored to the maintenance of a healthy trajectory of the intestinal microbiome, counteracting all processes connected to the pathophysiology of the human aging.
Collapse
Affiliation(s)
- Marco Candela
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| | - Elena Biagi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Paul W O'Toole
- Department of Microbiology & Alimentary Pharmabiotic Centre, University College Cork, Ireland
| | - Willem M De Vos
- Laboratory of Microbiology, Wageningen University, The Netherlands, and Departments of Veterinary Biosciences and Bacteriology & Immunology, Helsinki University, Finland
| |
Collapse
|
2921
|
House dust exposure mediates gut microbiome Lactobacillus enrichment and airway immune defense against allergens and virus infection. Proc Natl Acad Sci U S A 2013; 111:805-10. [PMID: 24344318 DOI: 10.1073/pnas.1310750111] [Citation(s) in RCA: 315] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exposure to dogs in early infancy has been shown to reduce the risk of childhood allergic disease development, and dog ownership is associated with a distinct house dust microbial exposure. Here, we demonstrate, using murine models, that exposure of mice to dog-associated house dust protects against ovalbumin or cockroach allergen-mediated airway pathology. Protected animals exhibited significant reduction in the total number of airway T cells, down-regulation of Th2-related airway responses, as well as mucin secretion. Following dog-associated dust exposure, the cecal microbiome of protected animals was extensively restructured with significant enrichment of, amongst others, Lactobacillus johnsonii. Supplementation of wild-type animals with L. johnsonii protected them against both airway allergen challenge or infection with respiratory syncytial virus. L. johnsonii-mediated protection was associated with significant reductions in the total number and proportion of activated CD11c(+)/CD11b(+) and CD11c(+)/CD8(+) cells, as well as significantly reduced airway Th2 cytokine expression. Our results reveal that exposure to dog-associated household dust results in protection against airway allergen challenge and a distinct gastrointestinal microbiome composition. Moreover, the study identifies L. johnsonii as a pivotal species within the gastrointestinal tract capable of influencing adaptive immunity at remote mucosal surfaces in a manner that is protective against a variety of respiratory insults.
Collapse
|
2922
|
The intestinal microbiota interferes with the microRNA response upon oral Listeria infection. mBio 2013; 4:e00707-13. [PMID: 24327339 PMCID: PMC3870255 DOI: 10.1128/mbio.00707-13] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The intestinal tract is the largest reservoir of microbes in the human body. The intestinal microbiota is thought to be able to modulate alterations of the gut induced by enteropathogens, thereby maintaining homeostasis. Listeria monocytogenes is the agent of listeriosis, an infection transmitted to humans upon ingestion of contaminated food. Crossing of the intestinal barrier is a critical step of the infection before dissemination into deeper organs. Here, we investigated the role of the intestinal microbiota in the regulation of host protein-coding genes and microRNA (miRNA or miR) expression during Listeria infection. We first established the intestinal miRNA signatures corresponding to the 10 most highly expressed miRNAs in the murine ileum of conventional and germfree mice, noninfected and infected with Listeria. Next, we identified 6 miRNAs whose expression decreased upon Listeria infection in conventional mice. Strikingly, five of these miRNA expression variations (in miR-143, miR-148a, miR-200b, miR-200c, and miR-378) were dependent on the presence of the microbiota. In addition, as is already known, protein-coding genes were highly affected by infection in both conventional and germfree mice. By crossing bioinformatically the predicted targets of the miRNAs to our whole-genome transcriptomic data, we revealed an miRNA-mRNA network that suggested miRNA-mediated global regulation during intestinal infection. Other recent studies have revealed an miRNA response to either bacterial pathogens or commensal bacteria. In contrast, our work provides an unprecedented insight into the impact of the intestinal microbiota on host transcriptional reprogramming during infection by a human pathogen. While the crucial role of miRNAs in regulating the host response to bacterial infection is increasingly recognized, the involvement of the intestinal microbiota in the regulation of miRNA expression has not been explored in detail. Here, we investigated the impact of the intestinal microbiota on the regulation of protein-coding genes and miRNA expression in a host infected by L. monocytogenes, a food-borne pathogen. We show that the microbiota interferes with the microRNA response upon oral Listeria infection and identify several protein-coding target genes whose expression correlates inversely with that of the miRNA. Further investigations of the regulatory networks involving miR-143, miR-148a, miR-200b, miR-200c, and miR-378 will provide new insights into the impact of the intestinal microbiota on the host upon bacterial infection.
Collapse
|
2923
|
Remely M, Aumueller E, Merold C, Dworzak S, Hippe B, Zanner J, Pointner A, Brath H, Haslberger AG. Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity. Gene 2013; 537:85-92. [PMID: 24325907 DOI: 10.1016/j.gene.2013.11.081] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/09/2013] [Accepted: 11/30/2013] [Indexed: 12/25/2022]
Abstract
The human gut microbiota and microbial influences on lipid and glucose metabolism, satiety, and chronic low-grade inflammation are known to be involved in metabolic syndrome. Fermentation end products, especially short chain fatty acids, are believed to engage the epigenetic regulation of inflammatory reactions via FFARs (free fatty acid receptor) and other short chain fatty acid receptors. We studied a potential interaction of the microbiota with epigenetic regulation in obese and type 2 diabetes patients compared to a lean control group over a four month intervention period. Intervention comprised a GLP-1 agonist (glucagon-like peptide 1) for type 2 diabetics and nutritional counseling for both intervention groups. Microbiota was analyzed for abundance, butyryl-CoA:acetate CoA-transferase gene and for diversity by polymerase chain reaction and 454 high-throughput sequencing. Epigenetic methylation of the promoter region of FFAR3 and LINE1 (long interspersed nuclear element 1) was analyzed using bisulfite conversion and pyrosequencing. The diversity of the microbiota as well as the abundance of Faecalibacterium prausnitzii were significantly lower in obese and type 2 diabetic patients compared to lean individuals. Results from Clostridium cluster IV and Clostridium cluster XIVa showed a decreasing trend in type 2 diabetics in comparison to the butyryl-CoA:acetate CoA-transferase gene and according to melt curve analysis. During intervention no significant changes were observed in either intervention group. The analysis of five CpGs in the promoter region of FFAR3 showed a significant lower methylation in obese and type 2 diabetics with an increase in obese patients over the intervention period. These results disclosed a significant correlation between a higher body mass index and lower methylation of FFAR3. LINE-1, a marker of global methylation, indicated no significant differences between the three groups or the time points, although methylation of type 2 diabetics tended to increase over time. Our results provide evidence that a different composition of gut microbiota in obesity and type 2 diabetes affect the epigenetic regulation of genes. Interactions between the microbiota and epigenetic regulation may involve not only short chain fatty acids binding to FFARs. Therefore dietary interventions influencing microbial composition may be considered as an option in the engagement against metabolic syndrome.
Collapse
Affiliation(s)
- Marlene Remely
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - Eva Aumueller
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - Christine Merold
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - Simone Dworzak
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - Berit Hippe
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - Julia Zanner
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - Angelika Pointner
- Department of Nutritional Sciences, University Vienna, Vienna, Austria
| | - Helmut Brath
- Diabetes Outpatient Clinic, Health Center South, Vienna, Austria
| | | |
Collapse
|
2924
|
Gu Y, Zhao A, Huang F, Zhang Y, Liu J, Wang C, Jia W, Xie G, Jia W. Very low carbohydrate diet significantly alters the serum metabolic profiles in obese subjects. J Proteome Res 2013; 12:5801-5811. [PMID: 24224694 PMCID: PMC6088239 DOI: 10.1021/pr4008199] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Emerging evidence has consistently shown that a very low carbohydrate diet (VLCD) can protect against the development of obesity, but the underlying mechanisms are not fully understood. Here we applied a comprehensive metabonomics approach using ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry and gas chromatography-time-of-flight mass spectrometry to study the effects of an 8-week dietary intervention with VLCD on serum metabolic profiles in obese subjects. The VLCD intervention resulted in a weight loss and significantly decreased homeostasis model assessment-insulin resistance. The metabonomics analysis identified a number of differential serum metabolites (p < 0.05) primarily attributable to fatty acids, amino acids including branched chain amino acids, amines, lipids, carboxylic acids, and carbohydrates in obese subjects compared to healthy controls. The correlation analysis among time, VLCD intervention, and clinical parameters revealed that the changes of metabolites correlated with the changes of clinical parameters and showed differences in males and females. Fatty acids, amino acids, and carboxylic acids were increased in obese subjects compared with their normal healthy counterparts. Such increased levels of serum metabolites were attenuated after VLCD intake, suggesting that the health beneficial effects of VLCD are associated with attenuation of impaired fatty acid and amino acid metabolism. It also appears that VLCD induced significant metabolic alterations independent of the obesity-related metabolic changes. The altered metabolites in obese subjects post-VLCD intervention include arachidonate, cis-11,14-eicosadienoate, cis-11,14,17-eicosatrienoate, 2-aminobutyrate, acetyl-carnitine, and threonate, all of which are involved in inflammation and oxidation processes. The results revealed favorable shifts in fatty acids and amino acids after VLCD intake in obese subjects, which should be considered biomarkers for evaluating health beneficial effects of VLCD and similar dietary interventions.
Collapse
Affiliation(s)
- Yunjuan Gu
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Aihua Zhao
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Fengjie Huang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yinan Zhang
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Jiajian Liu
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Congrong Wang
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Wei Jia
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Guoxiang Xie
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Weiping Jia
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
2925
|
Microbial enterotypes, inferred by the prevotella-to-bacteroides ratio, remained stable during a 6-month randomized controlled diet intervention with the new nordic diet. Appl Environ Microbiol 2013; 80:1142-9. [PMID: 24296500 DOI: 10.1128/aem.03549-13] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
It has been suggested that the human gut microbiota can be divided into enterotypes based on the abundance of specific bacterial groups; however, the biological significance and stability of these enterotypes remain unresolved. Here, we demonstrated that subjects (n = 62) 18 to 65 years old with central obesity and components of metabolic syndrome could be grouped into two discrete groups simply by their relative abundance of Prevotella spp. divided by Bacteroides spp. (P/B ratio) obtained by quantitative PCR analysis. Furthermore, we showed that these groups remained stable during a 6-month, controlled dietary intervention, where the effect of consuming a diet in accord with the new Nordic diet (NND) recommendations as opposed to consuming the average Danish diet (ADD) on the gut microbiota was investigated. In this study, subjects (with and without stratification according to P/B ratio) did not reveal significant changes in 35 selected bacterial taxa quantified by quantitative PCR (ADD compared to NND) resulting from the dietary interventions. However, we found higher total plasma cholesterol within the high-P/B group than in the low-P/B group after the intervention. We propose that stratification of humans based simply on their P/B ratio could allow better assessment of possible effects of interventions on the gut microbiota and physiological biomarkers.
Collapse
|
2926
|
Ramakrishna BS. Role of the gut microbiota in human nutrition and metabolism. J Gastroenterol Hepatol 2013; 28 Suppl 4:9-17. [PMID: 24251697 DOI: 10.1111/jgh.12294] [Citation(s) in RCA: 320] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2013] [Indexed: 02/06/2023]
Abstract
The human gastrointestinal tract harbors trillions of bacteria, most of which are commensal and have adapted over time to the milieu of the human colon. Their many metabolic interactions with each other, and with the human host, influence human nutrition and metabolism in diverse ways. Our understanding of these influences has come through breakthroughs in the molecular profiling of the phylogeny and the metabolic capacities of the microbiota. The gut microbiota produce a variety of nutrients including short-chain fatty acids, B vitamins, and vitamin K. Because of their ability to interact with receptors on epithelial cells and subepithelial cells, the microbiota also release a number of cellular factors that influence human metabolism. Thus, they have potential roles in the pathogenesis of metabolic syndrome, diabetes, non-alcoholic fatty liver disease, and cognition, which extend well beyond their traditional contribution to nutrition. This review explores the roles of the gut microbiota in human nutrition and metabolism, and the putative mechanisms underlying these effects.
Collapse
Affiliation(s)
- Balakrishnan S Ramakrishna
- Department of Gastroenterology, SRM Institutes of Medical Science, Chennai, India; Flinders University of South Australia, Bedford Park, South Australia, Australia
| |
Collapse
|
2927
|
Abstract
Bacterial colonisation of the gut plays a major role in postnatal development and maturation of key systems that have the capacity to influence central nervous system (CNS) programming and signaling, including the immune and endocrine systems. Individually, these systems have been implicated in the neuropathology of many CNS disorders and collectively they form an important bidirectional pathway of communication between the microbiota and the brain in health and disease. Regulation of the microbiome-brain-gut axis is essential for maintaining homeostasis, including that of the CNS. Moreover, there is now expanding evidence for the view that commensal organisms within the gut play a role in early programming and later responsivity of the stress system. Research has focused on how the microbiota communicates with the CNS and thereby influences brain function. The routes of this communication are not fully elucidated but include neural, humoral, immune and metabolic pathways. This view is underpinned by studies in germ-free animals and in animals exposed to pathogenic bacterial infections, probiotic agents or antibiotics which indicate a role for the gut microbiota in the regulation of mood, cognition, pain and obesity. Thus, the concept of a microbiome-brain-gut axis is emerging which suggests that modulation of the gut microflora may be a tractable strategy for developing novel therapeutics for complex stress-related CNS disorders where there is a huge unmet medical need.
Collapse
|
2928
|
Close DW, Ferrara F, Dichosa AEK, Kumar S, Daughton AR, Daligault HE, Reitenga KG, Velappan N, Sanchez TC, Iyer S, Kiss C, Han CS, Bradbury ARM. Using phage display selected antibodies to dissect microbiomes for complete de novo genome sequencing of low abundance microbes. BMC Microbiol 2013; 13:270. [PMID: 24279426 PMCID: PMC3907030 DOI: 10.1186/1471-2180-13-270] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/21/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Single cell genomics has revolutionized microbial sequencing, but complete coverage of genomes in complex microbiomes is imperfect due to enormous variation in organismal abundance and amplification bias. Empirical methods that complement rapidly improving bioinformatic tools will improve characterization of microbiomes and facilitate better genome coverage for low abundance microbes. METHODS We describe a new approach to sequencing individual species from microbiomes that combines antibody phage display against intact bacteria with fluorescence activated cell sorting (FACS). Single chain (scFv) antibodies are selected using phage display against a bacteria or microbial community, resulting in species-specific antibodies that can be used in FACS for relative quantification of an organism in a community, as well as enrichment or depletion prior to genome sequencing. RESULTS We selected antibodies against Lactobacillus acidophilus and demonstrate a FACS-based approach for identification and enrichment of the organism from both laboratory-cultured and commercially derived bacterial mixtures. The ability to selectively enrich for L. acidophilus when it is present at a very low abundance (<0.2%) leads to complete (>99.8%) de novo genome coverage whereas the standard single-cell sequencing approach is incomplete (<68%). We show that specific antibodies can be selected against L. acidophilus when the monoculture is used as antigen as well as when a community of 10 closely related species is used demonstrating that in principal antibodies can be generated against individual organisms within microbial communities. CONCLUSIONS The approach presented here demonstrates that phage-selected antibodies against bacteria enable identification, enrichment of rare species, and depletion of abundant organisms making it tractable to virtually any microbe or microbial community. Combining antibody specificity with FACS provides a new approach for characterizing and manipulating microbial communities prior to genome sequencing.
Collapse
Affiliation(s)
- Devin W Close
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2929
|
Commensal bacteria-dependent indole production enhances epithelial barrier function in the colon. PLoS One 2013; 8:e80604. [PMID: 24278294 PMCID: PMC3835565 DOI: 10.1371/journal.pone.0080604] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 10/04/2013] [Indexed: 12/14/2022] Open
Abstract
Microbiota have been shown to have a great influence on functions of intestinal epithelial cells (ECs). The role of indole as a quorum-sensing (QS) molecule mediating intercellular signals in bacteria has been well appreciated. However, it remains unknown whether indole has beneficial effects on maintaining intestinal barriers in vivo. In this study, we analyzed the effect of indole on ECs using a germ free (GF) mouse model. GF mice showed decreased expression of junctional complex molecules in colonic ECs. The feces of specific pathogen-free (SPF) mice contained a high amount of indole; however the amount was significantly decreased in the feces of GF mice by 27-fold. Oral administration of indole-containing capsules resulted in increased expression of both tight junction (TJ)- and adherens junction (AJ)-associated molecules in colonic ECs in GF mice. In accordance with the increased expression of these junctional complex molecules, GF mice given indole-containing capsules showed higher resistance to dextran sodium sulfate (DSS)-induced colitis. A similar protective effect of indole on DSS-induced epithelial damage was also observed in mice bred in SPF conditions. These findings highlight the beneficial role of indole in establishing an epithelial barrier in vivo.
Collapse
|
2930
|
Mattijssen F, Alex S, Swarts HJ, Groen AK, van Schothorst EM, Kersten S. Angptl4 serves as an endogenous inhibitor of intestinal lipid digestion. Mol Metab 2013; 3:135-44. [PMID: 24634819 DOI: 10.1016/j.molmet.2013.11.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 11/08/2013] [Accepted: 11/13/2013] [Indexed: 02/07/2023] Open
Abstract
Dietary triglycerides are hydrolyzed in the small intestine principally by pancreatic lipase. Following uptake by enterocytes and secretion as chylomicrons, dietary lipids are cleared from the bloodstream via lipoprotein lipase. Whereas lipoprotein lipase is inhibited by several proteins including Angiopoietin-like 4 (Angptl4), no endogenous regulator of pancreatic lipase has yet been identified. Here we present evidence that Angptl4 is an endogenous inhibitor of dietary lipid digestion. Angptl4-/- mice were heavier compared to their wild-type counterparts without any difference in food intake, energy expenditure or locomotor activity. However, Angptl4-/- mice showed decreased lipid content in the stools and increased accumulation of dietary triglycerides in the small intestine, which coincided with elevated luminal lipase activity in Angptl4-/- mice. Furthermore, recombinant Angptl4 reduced the activity of pancreatic lipase as well as the lipase activity in human ileostomy output. In conclusion, our data suggest that Angptl4 is an endogenous inhibitor of intestinal lipase activity.
Collapse
Affiliation(s)
- Frits Mattijssen
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands
| | - Sheril Alex
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands
| | - Hans J Swarts
- Human and Animal Physiology, Department of Animal Sciences, Wageningen University, 6700 EV Wageningen, The Netherlands
| | - Albert K Groen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Evert M van Schothorst
- Human and Animal Physiology, Department of Animal Sciences, Wageningen University, 6700 EV Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands
| |
Collapse
|
2931
|
Curcuma longa extract associated with white pepper lessens high fat diet-induced inflammation in subcutaneous adipose tissue. PLoS One 2013; 8:e81252. [PMID: 24260564 PMCID: PMC3834320 DOI: 10.1371/journal.pone.0081252] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/21/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Supra-nutritional doses of curcumin, derived from the spice Curcuma longa, have been proposed as a potential treatment of inflammation and metabolic disorders related to obesity. The aim of the present study was to test whether Curcuma longa extract rich in curcumin and associated with white pepper (Curcuma-P®), at doses compatible with human use, could modulate systemic inflammation in diet-induced obese mice. We questioned the potential relevance of changes in adiposity and gut microbiota in the effect of Curcuma-P® in obesity. METHODOLOGY/PRINCIPAL FINDINGS Mice were fed either a control diet (CT), a high fat (HF) diet or a HF diet containing Curcuma longa extract (0.1 % of curcumin in the HF diet) associated with white pepper (0.01 %) for four weeks. Curcumin has been usually combined with white pepper, which contain piperine, in order to improve its bioavailability. This combination did not significantly modify body weight gain, glycemia, insulinemia, serum lipids and intestinal inflammatory markers. Tetrahydrocurcumin, but not curcumin accumulated in the subcutaneous adipose tissue. Importantly, the co-supplementation in curcuma extract and white pepper decreased HF-induced pro-inflammatory cytokines expression in the subcutaneous adipose tissue, an effect independent of adiposity, immune cells recruitment, angiogenesis, or modulation of gut bacteria controlling inflammation. CONCLUSIONS/SIGNIFICANCE These findings support that nutritional doses of Curcuma longa, associated with white pepper, is able to decrease inflammatory cytokines expression in the adipose tissue and this effect could be rather linked to a direct effect of bioactive metabolites reaching the adipose tissue, than from changes in the gut microbiota composition.
Collapse
|
2932
|
Lu K, Cable PH, Abo RP, Ru H, Graffam ME, Schlieper KA, Parry NMA, Levine S, Bodnar WM, Wishnok JS, Styblo M, Swenberg JA, Fox JG, Tannenbaum SR. Gut microbiome perturbations induced by bacterial infection affect arsenic biotransformation. Chem Res Toxicol 2013; 26:1893-903. [PMID: 24134150 DOI: 10.1021/tx4002868] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Exposure to arsenic affects large human populations worldwide and has been associated with a long list of human diseases, including skin, bladder, lung, and liver cancers, diabetes, and cardiovascular disorders. In addition, there are large individual differences in susceptibility to arsenic-induced diseases, which are frequently associated with different patterns of arsenic metabolism. Several underlying mechanisms, such as genetic polymorphisms and epigenetics, have been proposed, as these factors closely impact the individuals' capacity to metabolize arsenic. In this context, the role of the gut microbiome in directly metabolizing arsenic and triggering systemic responses in diverse organs raises the possibility that perturbations of the gut microbial communities affect the spectrum of metabolized arsenic species and subsequent toxicological effects. In this study, we used an animal model with an altered gut microbiome induced by bacterial infection, 16S rRNA gene sequencing, and inductively coupled plasma mass spectrometry-based arsenic speciation to examine the effect of gut microbiome perturbations on the biotransformation of arsenic. Metagenomics sequencing revealed that bacterial infection significantly perturbed the gut microbiome composition in C57BL/6 mice, which in turn resulted in altered spectra of arsenic metabolites in urine, with inorganic arsenic species and methylated and thiolated arsenic being perturbed. These data clearly illustrated that gut microbiome phenotypes significantly affected arsenic metabolic reactions, including reduction, methylation, and thiolation. These findings improve our understanding of how infectious diseases and environmental exposure interact and may also provide novel insight regarding the gut microbiome composition as a new risk factor of individual susceptibility to environmental chemicals.
Collapse
Affiliation(s)
- Kun Lu
- Department of Biological Engineering, ‡Department of Biology, §Division of Comparative Medicine, and ∥Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2933
|
Couch RD, Navarro K, Sikaroodi M, Gillevet P, Forsyth CB, Mutlu E, Engen PA, Keshavarzian A. The approach to sample acquisition and its impact on the derived human fecal microbiome and VOC metabolome. PLoS One 2013; 8:e81163. [PMID: 24260553 PMCID: PMC3832442 DOI: 10.1371/journal.pone.0081163] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 10/09/2013] [Indexed: 01/28/2023] Open
Abstract
Recent studies have illustrated the importance of the microbiota in maintaining a healthy state, as well as promoting disease states. The intestinal microbiota exerts its effects primarily through its metabolites, and metabolomics investigations have begun to evaluate the diagnostic and health implications of volatile organic compounds (VOCs) isolated from human feces, enabled by specialized sampling methods such as headspace solid-phase microextraction (hSPME). The approach to stool sample collection is an important consideration that could potentially introduce bias and affect the outcome of a fecal metagenomic and metabolomic investigation. To address this concern, a comparison of endoscopically collected (in vivo) and home collected (ex vivo) fecal samples was performed, revealing slight variability in the derived microbiomes. In contrast, the VOC metabolomes differ widely between the home collected and endoscopy collected samples. Additionally, as the VOC extraction profile is hyperbolic, with short extraction durations more vulnerable to variation than extractions continued to equilibrium, a second goal of our investigation was to ascertain if hSPME-based fecal metabolomics studies might be biased by the extraction duration employed. As anticipated, prolonged extraction (18 hours) results in the identification of considerably more metabolites than short (20 minute) extractions. A comparison of the metabolomes reveals several analytes deemed unique to a cohort with the 20 minute extraction, but found common to both cohorts when the VOC extraction was performed for 18 hours. Moreover, numerous analytes perceived to have significant fold change with a 20 minute extraction were found insignificant in fold change with the prolonged extraction, underscoring the potential for bias associated with a 20 minute hSPME.
Collapse
Affiliation(s)
- Robin D. Couch
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Karl Navarro
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Masoumeh Sikaroodi
- Department of Environmental Science and Policy, George Mason University, Manassas, Virginia, United States of America
- The Microbiome Analysis Center, George Mason University, Manassas, Virginia, United States of America
| | - Pat Gillevet
- Department of Environmental Science and Policy, George Mason University, Manassas, Virginia, United States of America
- The Microbiome Analysis Center, George Mason University, Manassas, Virginia, United States of America
| | - Christopher B. Forsyth
- Division of Digestive Diseases and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Ece Mutlu
- Division of Digestive Diseases and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Phillip A. Engen
- Division of Digestive Diseases and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Ali Keshavarzian
- Division of Digestive Diseases and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, Illinois, United States of America
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2934
|
Vitetta L, Coulson S, Linnane AW, Butt H. The gastrointestinal microbiome and musculoskeletal diseases: a beneficial role for probiotics and prebiotics. Pathogens 2013; 2:606-26. [PMID: 25437335 PMCID: PMC4235701 DOI: 10.3390/pathogens2040606] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/04/2013] [Accepted: 11/07/2013] [Indexed: 12/11/2022] Open
Abstract
Natural medicines are an attractive option for patients diagnosed with common and debilitating musculoskeletal diseases such as Osteoarthritis (OA) or Rheumatoid Arthritis (RA). The high rate of self-medication with natural products is due to (1) lack of an available cure and (2) serious adverse events associated with chronic use of pharmaceutical medications in particular non-steroidal anti-inflammatory drugs (NSAIDs) and high dose paracetamol. Pharmaceuticals to treat pain may disrupt gastrointestinal (GIT) barrier integrity inducing GIT inflammation and a state of and hyper-permeability. Probiotics and prebiotics may comprise plausible therapeutic options that can restore GIT barrier functionality and down regulate pro-inflammatory mediators by modulating the activity of, for example, Clostridia species known to induce pro-inflammatory mediators. The effect may comprise the rescue of gut barrier physiological function. A postulated requirement has been the abrogation of free radical formation by numerous natural antioxidant molecules in order to improve musculoskeletal health outcomes, this notion in our view, is in error. The production of reactive oxygen species (ROS) in different anatomical environments including the GIT by the epithelial lining and the commensal microbe cohort is a regulated process, leading to the formation of hydrogen peroxide which is now well recognized as an essential second messenger required for normal cellular homeostasis and physiological function. The GIT commensal profile that tolerates the host does so by regulating pro-inflammatory and anti-inflammatory GIT mucosal actions through the activity of ROS signaling thereby controlling the activity of pathogenic bacterial species.
Collapse
Affiliation(s)
| | - Samantha Coulson
- School of Medicine, The University of Queensland, Brisbane 4102, Australia.
| | | | - Henry Butt
- Bioscreen, Bio21, The University of Melbourne, Melbourne 3010, Australia.
| |
Collapse
|
2935
|
Ramezani A, Raj DS. The gut microbiome, kidney disease, and targeted interventions. J Am Soc Nephrol 2013; 25:657-70. [PMID: 24231662 DOI: 10.1681/asn.2013080905] [Citation(s) in RCA: 516] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The human gut harbors >100 trillion microbial cells, which influence the nutrition, metabolism, physiology, and immune function of the host. Here, we review the quantitative and qualitative changes in gut microbiota of patients with CKD that lead to disturbance of this symbiotic relationship, how this may contribute to the progression of CKD, and targeted interventions to re-establish symbiosis. Endotoxin derived from gut bacteria incites a powerful inflammatory response in the host organism. Furthermore, protein fermentation by gut microbiota generates myriad toxic metabolites, including p-cresol and indoxyl sulfate. Disruption of gut barrier function in CKD allows translocation of endotoxin and bacterial metabolites to the systemic circulation, which contributes to uremic toxicity, inflammation, progression of CKD, and associated cardiovascular disease. Several targeted interventions that aim to re-establish intestinal symbiosis, neutralize bacterial endotoxins, or adsorb gut-derived uremic toxins have been developed. Indeed, animal and human studies suggest that prebiotics and probiotics may have therapeutic roles in maintaining a metabolically-balanced gut microbiota and reducing progression of CKD and uremia-associated complications. We propose that further research should focus on using this highly efficient metabolic machinery to alleviate uremic symptoms.
Collapse
Affiliation(s)
- Ali Ramezani
- Division of Renal Diseases and Hypertension, The George Washington University, Washington DC
| | | |
Collapse
|
2936
|
Brestoff JR, Artis D. Commensal bacteria at the interface of host metabolism and the immune system. Nat Immunol 2013; 14:676-84. [PMID: 23778795 DOI: 10.1038/ni.2640] [Citation(s) in RCA: 660] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 05/10/2013] [Indexed: 02/07/2023]
Abstract
The mammalian gastrointestinal tract, the site of digestion and nutrient absorption, harbors trillions of beneficial commensal microbes from all three domains of life. Commensal bacteria, in particular, are key participants in the digestion of food, and are responsible for the extraction and synthesis of nutrients and other metabolites that are essential for the maintenance of mammalian health. Many of these nutrients and metabolites derived from commensal bacteria have been implicated in the development, homeostasis and function of the immune system, suggesting that commensal bacteria may influence host immunity via nutrient- and metabolite-dependent mechanisms. Here we review the current knowledge of how commensal bacteria regulate the production and bioavailability of immunomodulatory, diet-dependent nutrients and metabolites and discuss how these commensal bacteria-derived products may regulate the development and function of the mammalian immune system.
Collapse
Affiliation(s)
- Jonathan R Brestoff
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
2937
|
Vinjé S, Stroes E, Nieuwdorp M, Hazen SL. The gut microbiome as novel cardio-metabolic target: the time has come! Eur Heart J 2013; 35:883-7. [PMID: 24216389 DOI: 10.1093/eurheartj/eht467] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent studies reveal a potential contribution of intestinal microbes in the expression of certain human cardio-metabolic diseases. The mechanisms through which intestinal microbiota and/or their metabolic products alter systemic homoeostasis and cardio-metabolic disease risks are just beginning to be dissected. Intervention studies in humans aiming to either selectively alter the composition of the intestinal microbiota or to pharmacologically manipulate the microbiota to influence production of their metabolites are crucial next steps. The intestinal microbiome represents a new potential therapeutic target for the treatment of cardio-metabolic diseases.
Collapse
Affiliation(s)
- Sarah Vinjé
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, AMC-UvA, Meibergdreef 9, room F4-159.2, 1105 AZ Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
2938
|
Amirian ES, Petrosino JF, Ajami NJ, Liu Y, Mims MP, Scheurer ME. Potential role of gastrointestinal microbiota composition in prostate cancer risk. Infect Agent Cancer 2013; 8:42. [PMID: 24180596 PMCID: PMC3826836 DOI: 10.1186/1750-9378-8-42] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/12/2013] [Indexed: 12/12/2022] Open
Abstract
Background Among men in the U.S., prostate cancer is the most common cancer and the second leading cause of cancer death. Despite its prevalence, there are few established risk factors for prostate cancer. Some studies have found that intake of certain foods/nutrients may be associated with prostate cancer risk, but few have accounted for how intake and metabolic factors may interact to influence bioavailable nutrient levels and subsequent disease risk. Presentation of the hypothesis The composition of the gastrointestinal (GI) microbiome may influence metabolism of dietary compounds and nutrients (e.g., plant phenols, calcium, choline) that may be relevant to prostate cancer risk. We, therefore, propose the hypothesis that GI microbiota may have a markedly different composition among individuals with higher prostate cancer risk. These individuals could have microbial profiles that are conducive to intestinal inflammation and/or are less favorable for the metabolism and uptake of chemopreventive agents. Testing the hypothesis Because very little preliminary data exist on this potential association, a case–control study may provide valuable information on this topic. Such a study could evaluate whether the GI microbial profile is markedly different between three groups of individuals: healthy men, those with latent prostate cancer, and those with invasive prostate cancer. Any findings could then be validated in a larger study, designed to collect a series of specimens over time. Implications of the hypothesis Given the plethora of information emerging from the Human Microbiome Project, this is an opportune time to explore associations between the microbiome and complex human diseases. Identification of profiles that alter the host’s risk for disease may clarify inconsistencies in the literature on dietary factors and cancer risk, and could provide valuable targets for novel cancer prevention strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael E Scheurer
- Dan L Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza MS:BCM305, 77030 Houston, TX, USA.
| |
Collapse
|
2939
|
Smits LP, Bouter KEC, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology 2013; 145:946-53. [PMID: 24018052 DOI: 10.1053/j.gastro.2013.08.058] [Citation(s) in RCA: 464] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 08/23/2013] [Accepted: 08/26/2013] [Indexed: 02/08/2023]
Abstract
There has been growing interest in the use of fecal microbiota for the treatment of patients with chronic gastrointestinal infections and inflammatory bowel diseases. Lately, there has also been interest in its therapeutic potential for cardiometabolic, autoimmune, and other extraintestinal conditions that were not previously considered to be associated with the intestinal microbiota. Although it is not clear if changes in the microbiota cause these conditions, we review the most current and best methods for performing fecal microbiota transplantation and summarize clinical observations that have implicated the intestinal microbiota in various diseases. We also discuss case reports of fecal microbiota transplantations for different disorders, including Clostridium difficile infection, irritable bowel syndrome, inflammatory bowel diseases, insulin resistance, multiple sclerosis, and idiopathic thrombocytopenic purpura. There has been increasing focus on the interaction between the intestinal microbiome, obesity, and cardiometabolic diseases, and we explore these relationships and the potential roles of different microbial strains. We might someday be able to mine for intestinal bacterial strains that can be used in the diagnosis or treatment of these diseases.
Collapse
Affiliation(s)
- Loek P Smits
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
2940
|
Musculoskeletal system in the old age and the demand for healthy ageing biomarkers. Mech Ageing Dev 2013; 134:541-7. [DOI: 10.1016/j.mad.2013.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 10/24/2013] [Accepted: 11/11/2013] [Indexed: 12/19/2022]
|
2941
|
Abstract
The development of obesity and NAFLD is known to be determined by host genetics, diet and lack of exercise. In addition, the gut microbiota has been identified to influence the development of both obesity and NAFLD. Evidence for the role of the gut microbiota has been shown by preclinical studies of transfer of gut microbiota from lean and obese individuals, with the recipient developing the metabolic features of the donor. Many bidirectional interactions of the gut microbiota, including with food, bile and the intestinal epithelium, have been identified. These interactions might contribute to the distinct steps in the progression from lean to obese states, and to steatosis, steatohepatitis and eventually fibrosis. The predominant steps are efficient caloric extraction from the diet, intestinal epithelial damage and greater entry of bacterial components into the portal circulation. These steps result in activation of the innate immune system, liver inflammation and fibrosis. Fortunately, therapeutic interventions might not require a full understanding of these complex interactions. Although antibiotics are too unselective in their action, probiotics have shown efficacy in reversing obesity and NASH in experimental systems, and are under investigation in humans.
Collapse
Affiliation(s)
- Wajahat Z Mehal
- Section of Digestive Diseases, Yale University, 300 Cedar Street, TAC S241, PO Box 208019, New Haven, CT 06520-8019, USA.
| |
Collapse
|
2942
|
Neef A, Sanz Y. Future for probiotic science in functional food and dietary supplement development. Curr Opin Clin Nutr Metab Care 2013; 16:679-87. [PMID: 24071779 DOI: 10.1097/mco.0b013e328365c258] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW The purpose of this study is to provide an update of probiotic science evolving from classical approaches to the development of next-generation probiotics, parallel to advances in the understanding of the complexity of the gut microbiome and its role in human health. RECENT FINDINGS The probiotic concept is based on the notion that the gut ecosystem contributes to human physiology and, consequently, its modulation may help to maintain health and reduce disease risk. The understanding of the complexity of the gut microbiota and the specific components associated with progression from health to disease is rapidly increasing, thanks to the use of high-throughput and next-generation sequencing techniques in progressively better controlled epidemiological studies. Evidence on microbiome-mediated effects by intervention with classical probiotics on humans is, however, limited. The new information is helping to set a rationale for selection of a next generation of probiotics. Candidates include Clostridia clusters IV, XIVa and XVIII, Faecalibacterium prausnitzii, Akkermansia muciniphila and Bacteroides uniformis, the effects of which have been evaluated in preclinical trials with promising results for inflammatory and diet-related disorders. Yet, the extent to which new probiotic formulations consisting of nonconventional indigenous gut bacteria will be effective on humans at a population level or in personalized nutrition strategies remains to be explored. SUMMARY Understanding the role that indigenous intestinal bacteria and their ecological interactions play in human health and disease based on epidemiological, intervention and mechanistic studies will provide a robust rationale for selection of probiotic strains and facilitate the optimization of integrated dietary strategies to efficiently modulate the human gut microbiome, leading to improvements in nutrition and clinical practice.
Collapse
Affiliation(s)
- Alexander Neef
- Microbial Ecology, Nutrition & Health Research Group, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | | |
Collapse
|
2943
|
Trøseid M, Nestvold TK, Rudi K, Thoresen H, Nielsen EW, Lappegård KT. Plasma lipopolysaccharide is closely associated with glycemic control and abdominal obesity: evidence from bariatric surgery. Diabetes Care 2013; 36:3627-32. [PMID: 23835694 PMCID: PMC3816876 DOI: 10.2337/dc13-0451] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE It is of vital importance to elucidate the triggering factors of obesity and type 2 diabetes to improve patient care. Bariatric surgery has been shown to prevent and even cure diabetes, but the mechanism is unknown. Elevated levels of lipopolysaccharide (LPS) predict incident diabetes, but the sources of LPS are not clarified. The objective of the current study was to evaluate the potential impact of plasma LPS on abdominal obesity and glycemic control in subjects undergoing bariatric surgery. RESEARCH DESIGN AND METHODS This was a prospective observational study involving a consecutive sample of 49 obese subjects undergoing bariatric surgery and 17 controls. Main assessments were plasma LPS, HbA1c, adipose tissue volumes (computed tomography), and quantified bacterial DNA in adipose tissue compartments. RESULTS Plasma levels of LPS were elevated in obese individuals compared with controls (P < 0.001) and were reduced after bariatric surgery (P = 0.010). LPS levels were closely correlated with HbA1c (r = 0.56; P = 0.001) and intra-abdominal fat volumes (r = 0.61; P < 0.001), but only moderately correlated with subcutaneous fat volumes (r = 0.33; P = 0.038). Moreover, there was a decreasing gradient (twofold) in bacterial DNA levels going from mesenteric via omental to subcutaneous adipose tissue compartments (P = 0.041). Finally, reduced LPS levels after bariatric surgery were directly correlated with a reduction in HbA1c (r = 0.85; P < 0.001). CONCLUSIONS Our findings support a hypothesis of translocated gut bacteria as a potential trigger of obesity and diabetes, and suggest that the antidiabetic effects of bariatric surgery might be mechanistically linked to, and even the result of, a reduction in plasma levels of LPS.
Collapse
|
2944
|
Moschen AR, Kaser S, Tilg H. Non-alcoholic steatohepatitis: a microbiota-driven disease. Trends Endocrinol Metab 2013; 24:537-45. [PMID: 23827477 DOI: 10.1016/j.tem.2013.05.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 05/24/2013] [Accepted: 05/29/2013] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a major health problem worldwide. Whereas overnutrition and obesity are crucially involved in the development of a simple fatty liver, it remains unclear why approximately 10% of all affected individuals develop the 'inflammatory' phenotype so-called non-alcoholic steatohepatitis (NASH). A link between the intestinal microbiota and the development of obesity and its metabolic consequences including NAFLD is becoming clearer. First clinical, but especially experimental, studies are suggesting that microbiotal factors are driving forces of hepatic steatosis and inflammation that involve Toll-like receptors and proinflammatory cytokines such as tumor necrosis factor-α (TNFα). Future studies focused on deciphering how manipulation of the gut microbiota might prove beneficial for patients with NAFLD are warranted.
Collapse
Affiliation(s)
- Alexander R Moschen
- Department of Internal Medicine I, Endocrinology, Gastroenterology, and Metabolism, Medical University Innsbruck, Austria
| | | | | |
Collapse
|
2945
|
Shen W, Gaskins HR, McIntosh MK. Influence of dietary fat on intestinal microbes, inflammation, barrier function and metabolic outcomes. J Nutr Biochem 2013; 25:270-80. [PMID: 24355793 DOI: 10.1016/j.jnutbio.2013.09.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/09/2013] [Accepted: 09/16/2013] [Indexed: 02/07/2023]
Abstract
Recent studies using germ-free, gnotobiotic microbial transplantation/conventionalization or antibiotic treatment in rodent models have highlighted the critical role of intestinal microbes on gut health and metabolic functions of the host. Genetic and environmental factors influence the abundance and type of mutualistic vs. pathogenic bacteria, each of which has preferred substrates for growth and unique products of fermentation. Whereas some fermentation products or metabolites promote gut function and health, others impair gut function, leading to compromised nutrient digestion and barrier function that adversely impact the host. Such products may also influence food intake, energy harvest and expenditure, and insulin action, thereby influencing adiposity and related metabolic outcomes. Diet composition influences gut microbiota and subsequent fermentation products that impact the host, as demonstrated by prebiotic studies using oligosaccharides or other types of indigestible fiber. Recent studies also show that dietary lipids affect specific populations of gut microbes and their metabolic end products. This review will focus on studies examining the influence of dietary fat amount and type on the gut microbiome, intestinal health and positive and negative metabolic consequences. The protective role of omega-3-rich fatty acids on intestinal inflammation will also be examined.
Collapse
Affiliation(s)
- Wan Shen
- Department of Nutrition, UNC-Greensboro, Greensboro, NC 27410, USA
| | - H Rex Gaskins
- Department of Animal Sciences, Department of Pathobiology, Division of Nutritional Sciences, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 8-8-13, USA
| | | |
Collapse
|
2946
|
Cheng XY, Tian XL, Wang YS, Lin RM, Mao ZC, Chen N, Xie BY. Metagenomic analysis of the pinewood nematode microbiome reveals a symbiotic relationship critical for xenobiotics degradation. Sci Rep 2013; 3:1869. [PMID: 23694939 PMCID: PMC3660777 DOI: 10.1038/srep01869] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 04/30/2013] [Indexed: 11/09/2022] Open
Abstract
Our recent research revealed that pinewood nematode (PWN) possesses few genes encoding enzymes for degrading α-pinene, which is the main compound in pine resin. In this study, we examined the role of PWN microbiome in xenobiotics detoxification by metagenomic and bacteria culture analyses. Functional annotation of metagenomes illustrated that benzoate degradation and its related metabolisms may provide the main metabolic pathways for xenobiotics detoxification in the microbiome, which is obviously different from that in PWN that uses cytochrome P450 metabolism as the main pathway for detoxification. The metabolic pathway of degrading α-pinene is complete in microbiome, but incomplete in PWN genome. Experimental analysis demonstrated that most of tested cultivable bacteria can not only survive the stress of 0.4% α-pinene, but also utilize α-pinene as carbon source for their growth. Our results indicate that PWN and its microbiome have established a potentially mutualistic symbiotic relationship with complementary pathways in detoxification metabolism.
Collapse
Affiliation(s)
- Xin-Yue Cheng
- College of Life Sciences, Beijing Normal University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
2947
|
Hobden MR, Martin-Morales A, Guérin-Deremaux L, Wils D, Costabile A, Walton GE, Rowland I, Kennedy OB, Gibson GR. In vitro fermentation of NUTRIOSE(®) FB06, a wheat dextrin soluble fibre, in a continuous culture human colonic model system. PLoS One 2013; 8:e77128. [PMID: 24204753 PMCID: PMC3811981 DOI: 10.1371/journal.pone.0077128] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/05/2013] [Indexed: 01/01/2023] Open
Abstract
Wheat dextrin soluble fibre may have metabolic and health benefits, potentially acting via mechanisms governed by the selective modulation of the human gut microbiota. Our aim was to examine the impact of wheat dextrin on the composition and metabolic activity of the gut microbiota. We used a validated in vitro three-stage continuous culture human colonic model (gut model) system comprised of vessels simulating anatomical regions of the human colon. To mimic human ingestion, 7 g of wheat dextrin (NUTRIOSE® FB06) was administered to three gut models, twice daily at 10.00 and 15.00, for a total of 18 days. Samples were collected and analysed for microbial composition and organic acid concentrations by 16S rRNA-based fluorescence in situ hybridisation and gas chromatography approaches, respectively. Wheat dextrin mediated a significant increase in total bacteria in vessels simulating the transverse and distal colon, and a significant increase in key butyrate-producing bacteria Clostridium cluster XIVa and Roseburia genus in all vessels of the gut model. The production of principal short-chain fatty acids, acetate, propionate and butyrate, which have been purported to have protective, trophic and metabolic host benefits, were increased. Specifically, wheat dextrin fermentation had a significant butyrogenic effect in all vessels of the gut model and significantly increased production of acetate (vessels 2 and 3) and propionate (vessel 3), simulating the transverse and distal regions of the human colon, respectively. In conclusion, wheat dextrin NUTRIOSE® FB06 is selectively fermented in vitro by Clostridium cluster XIVa and Roseburia genus and beneficially alters the metabolic profile of the human gut microbiota.
Collapse
Affiliation(s)
- Mark R. Hobden
- Department of Food and Nutritional Sciences, The University of Reading, Reading, United Kingdom
- * E-mail:
| | - Agustin Martin-Morales
- Department of Food and Nutritional Sciences, The University of Reading, Reading, United Kingdom
| | | | | | - Adele Costabile
- Department of Food and Nutritional Sciences, The University of Reading, Reading, United Kingdom
| | - Gemma E. Walton
- Department of Food and Nutritional Sciences, The University of Reading, Reading, United Kingdom
| | - Ian Rowland
- Department of Food and Nutritional Sciences, The University of Reading, Reading, United Kingdom
| | - Orla B. Kennedy
- Department of Food and Nutritional Sciences, The University of Reading, Reading, United Kingdom
| | - Glenn R. Gibson
- Department of Food and Nutritional Sciences, The University of Reading, Reading, United Kingdom
| |
Collapse
|
2948
|
Ussher JR, Lopaschuk GD, Arduini A. Gut microbiota metabolism of L-carnitine and cardiovascular risk. Atherosclerosis 2013; 231:456-61. [PMID: 24267266 DOI: 10.1016/j.atherosclerosis.2013.10.013] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/12/2013] [Accepted: 10/14/2013] [Indexed: 02/07/2023]
Abstract
In recent years, a number of studies have alluded to the importance of the intestinal microflora in controlling whole-body metabolic homeostasis and organ physiology. In particular, it has been suggested that the hepatic production of trimethylamine-N-oxide (TMAO) from gut microbiota-derived trimethylamine (TMA) may enhance cardiovascular risk via promoting atherosclerotic lesion development. The source of TMA production via the gut microbiota appears to originate from 2 principle sources, either phosphatidylcholine/choline and/or L-carnitine. Therefore, it has been postulated that consumption of these dietary sources, which are often found in large quantities in red meats, may be critical factors promoting cardiovascular risk. In contrast, a number of studies demonstrate beneficial properties for l-carnitine consumption against metabolic diseases including skeletal muscle insulin resistance and ischemic heart disease. Furthermore, fish are a significant source of TMAO, but dietary fish consumption and fish oil supplementation may exhibit positive effects on cardiovascular health. In this mini-review we will discuss the discrepancies regarding L-carnitine supplementation and its possible negative effects on cardiovascular risk through potential increases in TMAO production, as well as its positive effects on metabolic health via increasing glucose metabolism in the muscle and heart.
Collapse
Affiliation(s)
- John R Ussher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, 5-1001-DD, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada.
| | | | | |
Collapse
|
2949
|
Martínez I, Brown AW, Walter J. Does host cholesterol metabolism impact the gut microbiota and why does it matter? Future Microbiol 2013; 8:571-3. [PMID: 23642112 DOI: 10.2217/fmb.13.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
2950
|
Greer RL, Morgun A, Shulzhenko N. Bridging immunity and lipid metabolism by gut microbiota. J Allergy Clin Immunol 2013; 132:253-62; quiz 263. [PMID: 23905915 DOI: 10.1016/j.jaci.2013.06.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 06/13/2013] [Accepted: 06/24/2013] [Indexed: 12/13/2022]
Abstract
The human gut is a unique organ in which hundreds of different microbial species find their habitat and in which different host physiologic functions, such as digestion, nutrition, and immunity, coexist. Although all these players were studied separately for decades, recently, there has been an explosion of studies demonstrating the essential role for interactions between these components in gut function. Furthermore, new systems biology methods provide essential tools to study this complex system as a whole and to identify key elements that define the crosstalk between the gut microbiota, immunity, and metabolism. This review is devoted to several human diseases resulting from the disruption in this crosstalk, including immunodeficiency-associated and environmental enteropathies, celiac disease, inflammatory bowel disease, and obesity. We describe findings in experimental models of these diseases and in germ-free animals that help us understand the mechanisms and test new therapeutic strategies. We also discuss current challenges that the field is facing and propose that a new generation of antibiotics, prebiotics, and probiotics coupled with novel, systems biology-driven diagnostics will provide the basis for future personalized therapy.
Collapse
Affiliation(s)
- Renee L Greer
- College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | | | | |
Collapse
|