901
|
Kim KA, Jang SE, Jeong JJ, Yu DH, Han MJ, Kim DH. Doenjang, a Korean soybean paste, ameliorates TNBS-induced colitis in mice by suppressing gut microbial lipopolysaccharide production and NF-κB activation. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.09.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
902
|
Romani L, Zelante T, De Luca A, Iannitti RG, Moretti S, Bartoli A, Aversa F, Puccetti P. Microbiota control of a tryptophan-AhR pathway in disease tolerance to fungi. Eur J Immunol 2014; 44:3192-200. [PMID: 25256754 DOI: 10.1002/eji.201344406] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/30/2014] [Accepted: 09/23/2014] [Indexed: 01/09/2023]
Abstract
An increased understanding of the importance of microbiota in shaping the host's immune and metabolic activities has rendered fungal interactions with their hosts more complex than previously appreciated. The aryl hydrocarbon receptor (AhR) has a pivotal role in connecting tryptophan catabolism by microbial communities and the host's own pathway of tryptophan metabolite production with the orchestration of T-cell function. AhR activation by a Lactobacillus-derived AhR ligand leads to the production of IL-22 to the benefit of mucosal defense mechanisms, an activity upregulated in the absence of the host tryptophan catabolic enzyme, indoleamine 2,3-dioxygenase 1 (IDO1), which is required for protection from fungal diseases ("disease tolerance"). As AhR activation in turn leads to the activation-in a feedback fashion-of IDO1, the regulatory loop involving AhR and IDO1 may have driven the coevolution of commensal fungi with the mammalian immune system and the microbiota, to the benefit of host survival and fungal commensalism. This review will discuss the essential help the microbiota provides in controlling the balance between the dual nature of the fungal-host relationship, namely, commensalism vs. infection.
Collapse
Affiliation(s)
- Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
903
|
Corware K, Yardley V, Mack C, Schuster S, Al-Hassi H, Herath S, Bergin P, Modolell M, Munder M, Müller I, Kropf P. Protein energy malnutrition increases arginase activity in monocytes and macrophages. Nutr Metab (Lond) 2014; 11:51. [PMID: 25392710 PMCID: PMC4228191 DOI: 10.1186/1743-7075-11-51] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/24/2014] [Indexed: 11/24/2022] Open
Abstract
Background Protein energy malnutrition is commonly associated with immune dysfunctions and is a major factor in susceptibility to infectious diseases. Methods In this study, we evaluated the impact of protein energy malnutrition on the capacity of monocytes and macrophages to upregulate arginase, an enzyme associated with immunosuppression and increased pathogen replication. Results Our results show that monocytes and macrophages are significantly increased in the bone marrow and blood of mice fed on a protein low diet. No alteration in the capacity of bone marrow derived macrophages isolated from malnourished mice to phagocytose particles, to produce the microbicidal molecule nitric oxide and to kill intracellular Leishmania parasites was detected. However, macrophages and monocytes from malnourished mice express significantly more arginase both in vitro and in vivo. Using an experimental model of visceral leishmaniasis, we show that following protein energy malnutrition, the increased parasite burden measured in the spleen of these mice coincided with increased arginase activity and that macrophages provide a more permissive environment for parasite growth. Conclusions Taken together, these results identify a novel mechanism in protein energy malnutrition that might contributes to increased susceptibility to infectious diseases by upregulating arginase activity in myeloid cells. Electronic supplementary material The online version of this article (doi:10.1186/1743-7075-11-51) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karina Corware
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Vanessa Yardley
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, UK
| | - Christopher Mack
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Steffen Schuster
- Department of Biochemistry, WHO Immunology Research and Training Center, University of Lausanne, Lausanne, Switzerland
| | - Hafid Al-Hassi
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Shanthi Herath
- School of Biological Sciences, Royal Holloway, University of London, Egham, UK
| | - Philip Bergin
- International AIDS Vaccine Initiative Human Immunology Laboratory, Faculty of Medicine, Imperial College London, London, UK
| | - Manuel Modolell
- Department of Cellular Immunology, Max-Planck-Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center Mainz, Mainz, Germany
| | - Ingrid Müller
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Pascale Kropf
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| |
Collapse
|
904
|
Genton L, Cani PD, Schrenzel J. Alterations of gut barrier and gut microbiota in food restriction, food deprivation and protein-energy wasting. Clin Nutr 2014; 34:341-9. [PMID: 25459400 DOI: 10.1016/j.clnu.2014.10.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 07/01/2014] [Accepted: 10/06/2014] [Indexed: 12/21/2022]
Abstract
Increasing evidence shows that gut microbiota composition is related to changes of gut barrier function including gut permeability and immune function. Gut microbiota is different in obese compared to lean subjects, suggesting that gut microbes are also involved in energy metabolism and subsequent nutritional state. While research on gut microbiota and gut barrier has presently mostly focused on intestinal inflammatory bowel diseases and more recently on obesity and type 2 diabetes, this review aims at summarizing the present knowledge regarding the impact, in vivo, of depleted nutritional states on structure and function of the gut epithelium, the gut-associated lymphoid tissue (GALT), the gut microbiota and the enteric nervous system. It highlights the complex interactions between the components of gut barrier in depleted states due to food deprivation, food restriction and protein energy wasting and shows that these interactions are multidirectional, implying the existence of feedbacks.
Collapse
Affiliation(s)
- L Genton
- Clinical Nutrition, University Hospital, Geneva, Switzerland.
| | - P D Cani
- Université catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life Sciences and BIOtechnology), Metabolism and Nutrition Research Group, Brussels, Belgium
| | - J Schrenzel
- Service of Infectious Diseases, University Hospital, Geneva, Switzerland
| |
Collapse
|
905
|
Thatcher SE, Zhang X, Howatt DA, Yiannikouris F, Gurley SB, Ennis T, Curci JA, Daugherty A, Cassis LA. Angiotensin-converting enzyme 2 decreases formation and severity of angiotensin II-induced abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol 2014; 34:2617-23. [PMID: 25301841 DOI: 10.1161/atvbaha.114.304613] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Angiotensin-converting enzyme 2 (ACE2) cleaves angiotensin II (AngII) to form angiotensin-(1-7) (Ang-(1-7)), which generally opposes effects of AngII. AngII infusion into hypercholesterolemic male mice induces formation of abdominal aortic aneurysms (AAAs). This study tests the hypothesis that deficiency of ACE2 promotes AngII-induced AAAs, whereas ACE2 activation suppresses aneurysm formation. APPROACH AND RESULTS ACE2 protein was detectable by immunostaining in mice and human AAAs. Whole-body deficiency of ACE2 significantly increased aortic lumen diameters and external diameters of suprarenal aortas from AngII-infused mice. Conversely, ACE2 deficiency in bone marrow-derived cells had no effect on AngII-induced AAAs. In contrast to AngII-induced AAAs, ACE2 deficiency had no significant effect on external aortic diameters of elastase-induced AAAs. Because ACE2 deficiency promoted AAA formation in AngII-infused mice, we determined whether ACE2 activation suppressed AAAs. ACE2 activation by administration of diminazene aceturate (30 mg/kg per day) to Ldlr(-/-) mice increased kidney ACE2 mRNA abundance and activity and elevated plasma Ang-(1-7) concentrations. Unexpectedly, administration of diminazene aceturate significantly reduced total sera cholesterol and very low-density lipoprotein-cholesterol concentrations. Notably, diminazene aceturate significantly decreased aortic lumen diameters and aortic external diameters of AngII-infused mice resulting in a marked reduction in AAA incidence (from 73% to 29%). None of these effects of diminazene aceturate were observed in the Ace2(-/y) mice. CONCLUSIONS These results demonstrate that ACE2 exerts a modulatory role in AngII-induced AAA formation, and that therapeutic stimulation of ACE2 could be a benefit to reduce AAA expansion and rupture in patients with an activated renin-angiotensin system.
Collapse
Affiliation(s)
- Sean E Thatcher
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - Xuan Zhang
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - Deborah A Howatt
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - Frederique Yiannikouris
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - Susan B Gurley
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - Terri Ennis
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - John A Curci
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - Alan Daugherty
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.)
| | - Lisa A Cassis
- From the Department of Pharmacology and Nutritional Sciences (S.E.T., F.Y., L.A.C.), Graduate Center for Toxicology (X.Z.), and Saha Cardiovascular Research Center, Department of Internal Medicine (D.A.H., A.D.), University of Kentucky, Lexington; Division of Nephrology, Department of Medicine, Duke University, Durham, NC (S.B.G.); and Department of Surgery, Section of Vascular Surgery, Washington University, St Louis, MO (T.E., J.A.C.).
| |
Collapse
|
906
|
Thorburn AN, Macia L, Mackay CR. Diet, metabolites, and "western-lifestyle" inflammatory diseases. Immunity 2014; 40:833-42. [PMID: 24950203 DOI: 10.1016/j.immuni.2014.05.014] [Citation(s) in RCA: 658] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Indexed: 02/08/2023]
Abstract
One explanation for the increased incidence of allergies, asthma, and even some autoimmune diseases has been the hygiene hypothesis. However, recent studies also highlight an important role for diet and bacterial metabolites in controlling various immune pathways, including gut and immune homeostasis, regulatory T cell biology, and inflammation. Dietary-related metabolites engage "metabolite-sensing" G-protein-coupled receptors, such as GPR43, GPR41, GPR109A, GPR120, and GPR35. These receptors are expressed on immune cells and some gut epithelial cells and generally mediate a direct anti-inflammatory effect. Insufficient intake of "healthy foodstuffs" adversely affects the production of bacterial metabolites. These metabolites and those derived directly from food drive beneficial downstream effects on immune pathways. We propose that insufficient exposure to dietary and bacterial metabolites might underlie the development of inflammatory disorders in Western countries. This review highlights what is currently known about diet, metabolites, and their associated immune pathways in relation to the development of inflammatory disease.
Collapse
Affiliation(s)
- Alison N Thorburn
- Department of Immunology, Monash University, Clayton, VIC 3800, Australia
| | - Laurence Macia
- Department of Immunology, Monash University, Clayton, VIC 3800, Australia
| | - Charles R Mackay
- Department of Immunology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
907
|
Valdez Y, Brown EM, Finlay BB. Influence of the microbiota on vaccine effectiveness. Trends Immunol 2014; 35:526-37. [PMID: 25113637 DOI: 10.1016/j.it.2014.07.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 07/01/2014] [Accepted: 07/10/2014] [Indexed: 12/22/2022]
Abstract
Studies of the relationship between the microbiome and the development and function of the immune system are demonstrating novel concepts that could significantly alter the way we treat disease and promote wellness. Several diseases, including inflammatory bowel disease, allergy/asthma, and diabetes, are associated with changes in composition of the microbiome. Recent findings suggest novel complex mechanisms by which the microbiome impacts immune cell development and differentiation. A major implication of these findings is that the composition of microbiome may ultimately affect vaccine efficacy. We explore here the potential role of the microbiome in vaccine responses in the context of our growing understanding of the relationship between the gastrointestinal microbiota, resident immune cell populations, and systemic immunity.
Collapse
Affiliation(s)
- Yanet Valdez
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Eric M Brown
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
908
|
Cao X, Yang FY, Xin Z, Xie RR, Yang JK. The ACE2/Ang-(1-7)/Mas axis can inhibit hepatic insulin resistance. Mol Cell Endocrinol 2014; 393:30-8. [PMID: 24911884 DOI: 10.1016/j.mce.2014.05.024] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 12/24/2022]
Abstract
Blocking the renin-angiotensin system (RAS) can reduce the risk of diabetes. Meanwhile, the angiotensin (Ang)-converting enzyme-2 (ACE2)/Ang-(1-7)/Mas axis has recently been proposed to function as a negative regulator of the RAS. In previous studies, we first demonstrated that ACE2 knockout (ACE2(-/)(y)) mice exhibit impaired glucose tolerance or diabetes. However the precise roles of ACE2 on glucose metabolism are unknown. Here we show that the ACE2/Ang-(1-7)/Mas axis can ameliorate insulin resistance in the liver. Activation of the ACE2/Ang-(1-7)/Mas axis increases glucose uptake and decreases glycogen synthesis in the liver accompanied by increased expression of glucose transporters, insulin receptor substrates and decreased expression of enzymes for glycogen synthesis. ACE2 knockout mice displayed elevated levels of oxidative stress and exposure to Ang-(1-7) reduced the stress in hepatic cells. As a consequence of anti-oxidative stress, activation of the ACE2/Ang-(1-7)/Mas axis led to improved hepatic insulin resistance through the Akt/PI3K/IRS-1/JNK insulin signaling pathway. This is the first time documented that the ACE2/Ang-(1-7)/Mas axis can ameliorate insulin resistance in the liver. As insulin resistance in the liver is considered to be the primary cause of the development of type 2 diabetes, this axis may serve as a new diabetes target.
Collapse
Affiliation(s)
- Xi Cao
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 10730, China; Beijng Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Fang-Yuan Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 10730, China; Beijng Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Zhong Xin
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 10730, China; Beijng Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Rong-Rong Xie
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 10730, China; Beijng Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Jin-Kui Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 10730, China; Beijng Key Laboratory of Diabetes Research and Care, Beijing 100730, China.
| |
Collapse
|
909
|
Shapiro H, Thaiss CA, Levy M, Elinav E. The cross talk between microbiota and the immune system: metabolites take center stage. Curr Opin Immunol 2014; 30:54-62. [PMID: 25064714 DOI: 10.1016/j.coi.2014.07.003] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/28/2014] [Accepted: 07/06/2014] [Indexed: 02/07/2023]
Abstract
The human meta-organism consists of more than 90% of microbial cells. The gastrointestinal tract harbors trillions of commensal microorganisms that influence the development and homeostasis of the host. Alterations in composition and function of the microbiota, termed dysbiosis, have been implicated in a multitude of metabolic and inflammatory diseases in humans. Thus, understanding the molecular underpinnings the cross talk between commensal bacteria and their host during homeostasis and dysbiosis may hold the key to understanding many idiopathic diseases. While most attention has focused on the innate recognition of immune-stimulatory bacterial molecules, such as cell wall components and nucleic acids, we emphasize here the impact of diet-dependent microbial metabolites on the development and function of the immune system.
Collapse
Affiliation(s)
- Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maayan Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
910
|
|
911
|
Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol 2014; 11:413-26. [PMID: 24776703 PMCID: PMC7097196 DOI: 10.1038/nrcardio.2014.59] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) 2 and its product angiotensin 1–7 are thought to have effects that counteract the adverse actions of other, better-known renin–angiotensin system (RAS) components Numerous experimental studies have suggested that ACE2 and angiotensin 1–7 have notable protective effects in the heart and blood vessels ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the functional importance and signalling mechanisms of angiotensin-1–7-induced actions remain unclear New pharmacological interventions targeting ACE2 are expected to be useful in clinical treatment of cardiovascular disease, especially those associated with overactivation of the conventional RAS More studies, especially randomized controlled clinical trials, are needed to clearly delineate the benefits of therapies targeting angiotensin 1–7 actions
Angiotensin-converting enzyme 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of the better-known members of the renin–angiotensin system and might, therefore, be useful therapeutic targets in patients with cardiovascular disease. Professor Jiang and colleagues review the evidence for the potential roles of these proteins in various cardiovascular conditions, including hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes. The renin–angiotensin system (RAS) has pivotal roles in the regulation of normal physiology and the pathogenesis of cardiovascular disease. Angiotensin-converting enzyme (ACE) 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of other, better known and understood, members of the RAS. The physiological and pathological importance of ACE2 and angiotensin 1–7 in the cardiovascular system are not completely understood, but numerous experimental studies have indicated that these components have protective effects in the heart and blood vessels. Here, we provide an overview on the basic properties of ACE2 and angiotensin 1–7 and a summary of the evidence from experimental and clinical studies of various pathological conditions, such as hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes mellitus. ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the relevant functions and signalling mechanisms of actions induced by angiotensin 1–7 have not been conclusively determined. The ACE2–angiotensin 1–7 pathway, however, might provide a useful therapeutic target for the treatment of cardiovascular disease, especially in patients with overactive RAS.
Collapse
Affiliation(s)
- Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Jianmin Yang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Yongtao Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Mei Dong
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Fang Fang Liu
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Kai Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| |
Collapse
|
912
|
Bettenworth D, Nowacki TM, Ross M, Kyme P, Schwammbach D, Kerstiens L, Thoennissen GB, Bokemeyer C, Hengst K, Berdel WE, Heidemann J, Thoennissen NH. Nicotinamide treatment ameliorates the course of experimental colitis mediated by enhanced neutrophil‐specific antibacterial clearance. Mol Nutr Food Res 2014; 58:1474-90. [DOI: 10.1002/mnfr.201300818] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 12/19/2022]
Affiliation(s)
| | | | - Matthias Ross
- Department of Medicine BUniversity of Münster Münster Germany
| | - Pierre Kyme
- Department of Biomedical SciencesCedars‐Sinai Medical Center Los Angeles CA USA
| | - Daniela Schwammbach
- Department of Medicine A, HematologyOncology and PneumologyUniversity of Münster Münster Germany
| | - Linda Kerstiens
- Department of Medicine A, HematologyOncology and PneumologyUniversity of Münster Münster Germany
| | - Gabriela B. Thoennissen
- Department of Medicine A, HematologyOncology and PneumologyUniversity of Münster Münster Germany
| | - Carsten Bokemeyer
- Department of Oncology and HematologyBMT with Section of PneumologyHubertus Wald TumorzentrumUniversity Cancer Center HamburgUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Karin Hengst
- Department of Medicine BUniversity of Münster Münster Germany
| | - Wolfgang E. Berdel
- Department of Medicine A, HematologyOncology and PneumologyUniversity of Münster Münster Germany
| | - Jan Heidemann
- Department of Medicine BUniversity of Münster Münster Germany
| | - Nils H. Thoennissen
- Department of Medicine A, HematologyOncology and PneumologyUniversity of Münster Münster Germany
- Department of Oncology and HematologyBMT with Section of PneumologyHubertus Wald TumorzentrumUniversity Cancer Center HamburgUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| |
Collapse
|
913
|
Abstract
PURPOSE OF REVIEW Epithelial neutral amino acid transporters have been identified at the molecular level in recent years. Mouse models have now established the crucial role of these transporters for systemic amino acid homeostasis. This review summarizes recent progress in this field. RECENT FINDINGS Epithelial neutral amino acid transporters play an important role in the homeostasis of neutral amino acid levels in the body. They are important for the maintenance of body weight and muscle mass and serve as fuels. They also serve a role in providing nutrients to epithelial cells. Changes of plasma amino acid levels are not necessarily correlated to the amino acids appearing in the urine; changes in organ amino acid metabolism need to be taken into account. SUMMARY Genetic deletion of neutral amino acid transporters provides insight into their role in protein nutrition and homeostasis.
Collapse
|
914
|
Epigenetic regulation of angiotensin-converting enzyme 2 (ACE2) by SIRT1 under conditions of cell energy stress. Clin Sci (Lond) 2014; 126:507-16. [PMID: 24147777 DOI: 10.1042/cs20130291] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ACE2 (angiotensin-converting enzyme 2) counterbalances the actions of ACE (angiotensin-converting enzyme) by metabolizing its catalytic product, the vasoactive and fibrogenic peptide AngII (angiotensin II), into Ang-(1-7) [angiotensin-(1-7)]. Enhanced ACE2 expression may be protective in diabetes, cardiovascular disease and cancer. However, relatively little is known about the specific physiological factors regulating ACE2 expression. In the present paper, we show, by Western blotting and qPCR (quantitative real-time PCR), that ACE2 expression is increased under conditions of cell stress, including hypoxic conditions, IL (interleukin)-1β treatment and treatment with the AMP mimic AICAR (5-amino-4-imidazolecarboxamide riboside). The NAD+-dependent deacetylase SIRT1 (silent information regulator T1) was found to be up-regulated after AICAR treatment but, conversely, was down-regulated after IL-1β treatment. ChIP analysis demonstrated that SIRT1 bound to the ACE2 promoter and that binding was increased after AICAR treatment, but decreased after IL-1β treatment. Inhibition of SIRT1 activity ablated the AICAR-induced increase in ACE2. In conclusion, we have established that the expression of the ACE2 transcript is controlled by the activity of SIRT1 under conditions of energy stress.
Collapse
|
915
|
Garg M, Burrell LM, Velkoska E, Griggs K, Angus PW, Gibson PR, Lubel JS. Upregulation of circulating components of the alternative renin-angiotensin system in inflammatory bowel disease: A pilot study. J Renin Angiotensin Aldosterone Syst 2014; 16:559-69. [PMID: 24505094 DOI: 10.1177/1470320314521086] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 12/15/2013] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The relationship between intestinal inflammation and circulating components of the renin-angiotensin system (RAS) is poorly understood. MATERIALS AND METHODS Demographic and clinical data were obtained from healthy controls and patients with inflammatory bowel disease (IBD). Plasma concentrations of the classical RAS components (angiotensin-converting enzyme (ACE) and angiotensin II (Ang II)) and alternative RAS components (ACE2 and angiotensin (1-7) (Ang (1-7))) were analysed by radioimmuno- and enzymatic assays. Systemic inflammation was assessed using serum C-reactive protein (CRP), white cell count, platelet count and albumin, and intestinal inflammation by faecal calprotectin. RESULTS Nineteen healthy controls (11 female; mean age 38 years, range 23-68), 19 patients with Crohn's disease (11 female; aged 45 years, range 23-76) and 15 patients with ulcerative colitis (6 female; aged 42 years, 26-64) were studied. Circulating classical RAS component levels were similar across the three groups, whereas ACE2 activity and Ang (1-7) concentrations were higher in patients with IBD compared to controls (ACE2: 21.5 vs 13.3 pmol/ml/min, p<0.05; Ang (1-7): 22.8 vs 14.1 pg/ml, p<0.001). Ang (1-7) correlated weakly with platelet and white cell counts, but not calprotectin or CRP, in patients with IBD. CONCLUSIONS Circulating components of the alternative RAS are increased in patients with IBD.
Collapse
Affiliation(s)
- Mayur Garg
- Department of Gastroenterology and Hepatology, Eastern Health, Australia Eastern Health Clinical School, Monash University, Australia
| | | | - Elena Velkoska
- Department of Medicine, The University of Melbourne, Australia
| | - Karen Griggs
- Department of Medicine, The University of Melbourne, Australia
| | - Peter W Angus
- Department of Medicine, The University of Melbourne, Australia Gastroenterology and Liver Transplant Unit, Austin Hospital, Australia
| | - Peter R Gibson
- Eastern Health Clinical School, Monash University, Australia Department of Gastroenterology, Department of Gastroenterology, The Alfred Hospital and Monash University, Australia
| | - John S Lubel
- Department of Gastroenterology and Hepatology, Eastern Health, Australia Eastern Health Clinical School, Monash University, Australia
| |
Collapse
|
916
|
Liu R, Qi H, Wang J, Wang Y, Cui L, Wen Y, Yin C. Angiotensin-converting enzyme (ACE and ACE2) imbalance correlates with the severity of cerulein-induced acute pancreatitis in mice. Exp Physiol 2014; 99:651-63. [PMID: 24414175 DOI: 10.1113/expphysiol.2013.074815] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Angiotensin-converting enzyme (ACE) and its effector peptide angiotensin II (Ang II) have been implicated in the pathogenesis of pancreatitis. Angiotensin-converting enzyme 2 (ACE2) degrades Ang II to angiotensin-(1-7) [Ang-(1-7)] and has recently been described to have an antagonistic effect on ACE signalling. However, the specific underlying role of ACE2 in the pathogenesis of severe acute pancreatitis (SAP) is unclear. In the present study, the local imbalance of ACE and ACE2, as well as Ang II and Ang-(1-7) expression, was compared in wild-type (WT) and ACE2 knock-out (KO) or ACE2 transgenic (TG) mice subjected to cerulein-induced SAP. Serum amylase, tumour necrosis factor-α, interleukin (IL)-1β, IL-6 and IL-10 levels and histological morphometry were used to determine the severity of pancreatitis. In WT mice, pancreatic ACE and Ang II and serum Ang II expression increased (P < 0.05), while pancreatic ACE2 and Ang-(1-7) and serum Ang-(1-7) levels were also significantly elevated (P < 0.05) from 2 to 72 h after the onset of SAP. However, the ratio of pancreatic ACE2 to ACE expression was significantly reduced (from 1.46 ± 0.09 to 0.27 ± 0.05, P < 0.001) and paralleled the severity of pancreatitis. The Ace2 KO mice exhibited increased levels of tumour necrosis factor-α, IL-1β, IL-6, multifocal coagulative necrosis and inflammatory infiltrate, and lower levels of serum IL-10 and pancreatic Ang-(1-7) (4.70 ± 2.13 versus 10.87 ± 2.51, P < 0.001) compared with cerulein-treated WT mice at the same time point. Conversely, Ace2 TG mice with normal ACE expression were more resistant to SAP challenge as evidenced by a decreased inflammatory response, attenuated pathological changes and increased survival rates. These data suggest that the ACE2-ACE imbalance plays an important role in the pathogenesis of SAP and that pancreatic ACE2 is an important factor in determining the severity of SAP.
Collapse
Affiliation(s)
- Ruixia Liu
- * Department of Infection, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-An Road, Xicheng District, Beijing 100050, PR China.
| | | | | | | | | | | | | |
Collapse
|
917
|
Grammes F, Reveco FE, Romarheim OH, Landsverk T, Mydland LT, Øverland M. Candida utilis and Chlorella vulgaris counteract intestinal inflammation in Atlantic salmon (Salmo salar L.). PLoS One 2013; 8:e83213. [PMID: 24386162 PMCID: PMC3873917 DOI: 10.1371/journal.pone.0083213] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/01/2013] [Indexed: 12/18/2022] Open
Abstract
Intestinal inflammation, caused by impaired intestinal homeostasis, is a serious condition in both animals and humans. The use of conventional extracted soybean meal (SBM) in diets for Atlantic salmon and several other fish species is known to induce enteropathy in the distal intestine, a condition often referred to as SBM induced enteropathy (SBMIE). In the present study, we investigated the potential of different microbial ingredients to alleviate SBMIE in Atlantic salmon, as a model of feed-induced inflammation. The dietary treatments consisted of a negative control based on fish meal (FM), a positive control based on 20% SBM, and four experimental diets combining 20% SBM with either one of the three yeasts Candida utilis (CU), Kluyveromyces marxianus (KM), Saccharomyces cerevisiae (SC) or the microalgae Chlorella vulgaris (CV). Histopathological examination of the distal intestine showed that all fish fed the SC or SBM diets developed characteristic signs of SBMIE, while those fed the FM, CV or CU diets showed a healthy intestine. Fish fed the KM diet showed intermediate signs of SBMIE. Corroborating results were obtained when measuring the relative length of PCNA positive cells in the crypts of the distal intestine. Gene set enrichment analysis revealed decreased expression of amino acid, fat and drug metabolism pathways as well as increased expression of the pathways for NOD-like receptor signalling and chemokine signalling in both the SC and SBM groups while CV and CU were similar to FM and KM was intermediate. Gene expression of antimicrobial peptides was reduced in the groups showing SBMIE. The characterisation of microbial communities using PCR-DGGE showed a relative increased abundance of Firmicutes bacteria in fish fed the SC or SBM diets. Overall, our results show that both CU and CV were highly effective to counteract SBMIE, while KM had less effect and SC had no functional effects.
Collapse
Affiliation(s)
- Fabian Grammes
- Aquaculture Protein Centre, CoE, Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Felipe Eduardo Reveco
- Aquaculture Protein Centre, CoE, Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Odd Helge Romarheim
- Aquaculture Protein Centre, CoE, Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Thor Landsverk
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, Oslo, Norway
| | - Liv Torunn Mydland
- Aquaculture Protein Centre, CoE, Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Margareth Øverland
- Aquaculture Protein Centre, CoE, Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
- * E-mail:
| |
Collapse
|
918
|
Kuraishi T, Hori A, Kurata S. Host-microbe interactions in the gut of Drosophila melanogaster. Front Physiol 2013; 4:375. [PMID: 24381562 PMCID: PMC3865371 DOI: 10.3389/fphys.2013.00375] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 12/02/2013] [Indexed: 12/02/2022] Open
Abstract
Many insect species subsist on decaying and contaminated matter and are thus exposed to large quantities of microorganisms. To control beneficial commensals and combat infectious pathogens, insects must be armed with efficient systems for microbial recognition, signaling pathways, and effector molecules. The molecular mechanisms regulating these host-microbe interactions in insects have been largely clarified in Drosophila melanogaster with its powerful genetic and genomic tools. Here we review recent advances in this field, focusing mainly on the relationships between microbes and epithelial cells in the intestinal tract where the host exposure to the external environment is most frequent.
Collapse
Affiliation(s)
- Takayuki Kuraishi
- Department of Molrcular Biopharmacy and Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University Sendai, Japan ; PRESTO, Japan Science and Technology Agency Tokyo, Japan
| | - Aki Hori
- Department of Molrcular Biopharmacy and Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University Sendai, Japan
| | - Shoichiro Kurata
- Department of Molrcular Biopharmacy and Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University Sendai, Japan
| |
Collapse
|
919
|
Characteristic changes in microbial community composition and expression of innate immune genes in acute appendicitis. Innate Immun 2013; 21:30-41. [DOI: 10.1177/1753425913515033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Appendicitis represents a common and severe gastrointestinal illness in younger individuals worldwide. The disease is characterized by an excessive inflammatory response and it is believed that bacterial overgrowth due to blockage of the appendix lumen might be involved. Despite the high incidence, only limited data on the pathophysiological changes exist; in particular, the innate immune responses involved are largely unknown. Real-time PCR analysis of tissue samples from inflamed and normal appendices demonstrated differentially regulated expression patterns of epithelial-derived antimicrobial peptides (AMP). The α-defensins human neutrophil peptides 1–3, HD5 and HD6, as well as the two β-defensins, human β-defensins (hBD)-2 and hBD-3, were up-regulated, whereas hBD-1 was down-regulated in acute appendicitis. Expression of upstream regulators of AMP expression, NOD-2 and TLRs 1, 2, 4, 5, 7, 8 and 10 was significantly increased as detected by real-time PCR. Finally, we confirmed the involvement of the pro-inflammatory cytokines IL-1β and IL-8, and detected characteristic changes in microbial community composition in appendicitis tissue specimens by 16S rDNA based detection techniques. In this study, we demonstrate a differential regulation of the innate immune system along with an altered bacterial diversity in acute appendicitis.
Collapse
|
920
|
Foley SL, Johnson TJ, Ricke SC, Nayak R, Danzeisen J. Salmonella pathogenicity and host adaptation in chicken-associated serovars. Microbiol Mol Biol Rev 2013; 77:582-607. [PMID: 24296573 PMCID: PMC3973385 DOI: 10.1128/mmbr.00015-13] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enteric pathogens such as Salmonella enterica cause significant morbidity and mortality. S. enterica serovars are a diverse group of pathogens that have evolved to survive in a wide range of environments and across multiple hosts. S. enterica serovars such as S. Typhi, S. Dublin, and S. Gallinarum have a restricted host range, in which they are typically associated with one or a few host species, while S. Enteritidis and S. Typhimurium have broad host ranges. This review examines how S. enterica has evolved through adaptation to different host environments, especially as related to the chicken host, and continues to be an important human pathogen. Several factors impact host range, and these include the acquisition of genes via horizontal gene transfer with plasmids, transposons, and phages, which can potentially expand host range, and the loss of genes or their function, which would reduce the range of hosts that the organism can infect. S. Gallinarum, with a limited host range, has a large number of pseudogenes in its genome compared to broader-host-range serovars. S. enterica serovars such as S. Kentucky and S. Heidelberg also often have plasmids that may help them colonize poultry more efficiently. The ability to colonize different hosts also involves interactions with the host's immune system and commensal organisms that are present. Thus, the factors that impact the ability of Salmonella to colonize a particular host species, such as chickens, are complex and multifactorial, involving the host, the pathogen, and extrinsic pressures. It is the interplay of these factors which leads to the differences in host ranges that we observe today.
Collapse
|
921
|
Thaiss CA, Levy M, Suez J, Elinav E. The interplay between the innate immune system and the microbiota. Curr Opin Immunol 2013; 26:41-8. [PMID: 24556399 DOI: 10.1016/j.coi.2013.10.016] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/21/2013] [Accepted: 10/28/2013] [Indexed: 01/03/2023]
Abstract
The human gastrointestinal tract harbors one of the highest densities of microorganisms on earth, called the microbiota. In fact, the number of microbial cells in the intestine outnumbers the amount of human cells of the entire organism by a factor of 10. As such, a human being is more and more perceived as a super-organism consisting of a eukaryotic and a prokaryotic part. The compartment mediating the communication between both parts is the innate immune system and its various microbe-sensing pattern-recognition receptors. Co-evolution of the microbiota with the innate immune system has resulted in elaborate interdependency and feedback mechanisms by which both systems control mutual homeostasis. Here, we review the most important innate immune-microbiota interdependencies known to date. While microbial sensing by pattern-recognition receptors is required for stable microbial composition, the presence of the microbiota, in turn, is necessary for proper development and function of the immune system.
Collapse
Affiliation(s)
- Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maayan Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jotham Suez
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
922
|
Abstract
To prevent bacterial overgrowth, colonization of the epithelium and subsequent translocation, the gastrointestinal tract maintains an effective mucosal barrier. Besides mucus the most important components of this protective system are epithelial antimicrobial peptides such as defensins, the cathelicidin LL-37, lysozyme, phospholipase A, and proteins with additional antimicrobial properties such as ubiquicidin, ribosomal proteins or histones. Commensal species may tolerate intestinal antimicrobial peptides, for example Bacteroides ssp. or Parabacteroides ssp. as major species in the human colon were highly resistant to the constitutive defensin HBD-1 and only susceptible to the inducible defensin HBD-3. Reduction of disulfide bonds is an important mechanism activating HBD-1. As several studies show, alterations in the expression of antimicrobial peptides directly influence the composition of the intestinal flora. Correspondingly, an increased production of defensins or inhibition of the processing of mouse defensins to their active form led to a quantitative shift of luminal and mucosal bacterial species. On the other hand, microorganisms also modulate the synthesis of host defensins by induction or inhibition of specific peptides. Lactobacilli, the probiotic strain Escherichia coli Nissle and Salmonella enteritica stimulate HBD-2 expression, whereas Shigella flexneri downregulates the synthesis of HBD-1, HBD-3 and LL-37. Thus, the proper balance between the luminal flora and the mucosa is a permanently dynamic, sensitive and host-specific relationship.
Collapse
Affiliation(s)
- S Nuding
- University of Tübingen, Tübingen, Germany
| | | | | |
Collapse
|
923
|
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: 657] [Impact Index Per Article: 54.8] [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
|
924
|
Said HM. Recent advances in transport of water-soluble vitamins in organs of the digestive system: a focus on the colon and the pancreas. Am J Physiol Gastrointest Liver Physiol 2013; 305:G601-10. [PMID: 23989008 PMCID: PMC3840235 DOI: 10.1152/ajpgi.00231.2013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This review focuses on recent advances in our understanding of the mechanisms and regulation of water-soluble vitamin (WSV) transport in the large intestine and pancreas, two important organs of the digestive system that have only recently received their fair share of attention. WSV, a group of structurally unrelated compounds, are essential for normal cell function and development and, thus, for overall health and survival of the organism. Humans cannot synthesize WSV endogenously; rather, WSV are obtained from exogenous sources via intestinal absorption. The intestine is exposed to two sources of WSV: a dietary source and a bacterial source (i.e., WSV generated by the large intestinal microbiota). Contribution of the latter source to human nutrition/health has been a subject of debate and doubt, mostly based on the absence of specialized systems for efficient uptake of WSV in the large intestine. However, recent studies utilizing a variety of human and animal colon preparations clearly demonstrate that such systems do exist in the large intestine. This has provided strong support for the idea that the microbiota-generated WSV are of nutritional value to the host, and especially to the nutritional needs of the local colonocytes and their health. In the pancreas, WSV are essential for normal metabolic activities of all its cell types and for its exocrine and endocrine functions. Significant progress has also been made in understanding the mechanisms involved in the uptake of WSV and the effect of chronic alcohol exposure on the uptake processes.
Collapse
Affiliation(s)
- Hamid M. Said
- Departments of Medicine and Physiology/Biophysics, University of California, Irvine, California; and Department of Veterans Affairs Medical Center, Long Beach, California
| |
Collapse
|
925
|
Abstract
The intestine and its immune system have evolved to meet the extraordinary task of maintaining tolerance to the largest, most complex and diverse microbial commensal habitat, while meticulously attacking and containing even minute numbers of occasionally incoming pathogens. While our understanding is still far from complete, recent studies have provided exciting novel insights into the complex interplay of the many distinct intestinal immune cell types as well as the discovery of entirely new cell subsets. These studies have also revealed how proper development and function of the intestinal immune system is dependent on its specific microbiota, which appears to have evolutionarily co-evolved. Here we review key immune cells that maintain intestinal homeostasis and, conversely, describe how altered function and imbalances may lead to inflammatory bowel disease (IBD). We highlight the latest developments within this field, covering the major players in IBD including intestinal epithelial cells, macrophages, dendritic cells, adaptive immune cells, and the newly discovered innate lymphoid cells, which appear of characteristic importance for immune function at mucosal surfaces. We set these mucosal immune pathways in the functional context of IBD risk genes where such insight is available. Moreover, we frame our discussion of fundamental biological pathways that have been elucidated in model systems in the context of results from clinical trials in IBD that targeted key mediators secreted by these cells, as an attempt of 'functional' appraisal of these pathways in human disease.
Collapse
Affiliation(s)
- M Zaeem Cader
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Cambridge, Addenbrooke's Hospital, , Cambridge, UK
| | | |
Collapse
|
926
|
Sato T, Suzuki T, Watanabe H, Kadowaki A, Fukamizu A, Liu PP, Kimura A, Ito H, Penninger JM, Imai Y, Kuba K. Apelin is a positive regulator of ACE2 in failing hearts. J Clin Invest 2013; 123:5203-11. [PMID: 24177423 DOI: 10.1172/jci69608] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 08/29/2013] [Indexed: 11/17/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is a negative regulator of the renin-angiotensin system (RAS), catalyzing the conversion of Angiotensin II to Angiotensin 1-7. Apelin is a second catalytic substrate for ACE2 and functions as an inotropic and cardioprotective peptide. While an antagonistic relationship between the RAS and apelin has been proposed, such functional interplay remains elusive. Here we found that ACE2 was downregulated in apelin-deficient mice. Pharmacological or genetic inhibition of angiotensin II type 1 receptor (AT1R) rescued the impaired contractility and hypertrophy of apelin mutant mice, which was accompanied by restored ACE2 levels. Importantly, treatment with angiotensin 1-7 rescued hypertrophy and heart dysfunctions of apelin-knockout mice. Moreover, apelin, via activation of its receptor, APJ, increased ACE2 promoter activity in vitro and upregulated ACE2 expression in failing hearts in vivo. Apelin treatment also increased cardiac contractility and ACE2 levels in AT1R-deficient mice. These data demonstrate that ACE2 couples the RAS to the apelin system, adding a conceptual framework for the apelin-ACE2-angiotensin 1-7 axis as a therapeutic target for cardiovascular diseases.
Collapse
|
927
|
Sueyoshi R, Woods Ignatoski KM, Daignault S, Okawada M, Teitelbaum DH. Angiotensin converting enzyme-inhibitor reduces colitis severity in an IL-10 knockout model. Dig Dis Sci 2013; 58:3165-77. [PMID: 23949641 PMCID: PMC3859685 DOI: 10.1007/s10620-013-2825-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 07/23/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND We previously demonstrated angiotensin converting enzymes (ACE) over-expression in a dextran-sodium sulfate colitis model; ACE inhibitor (ACE-I) treatment reduced colitis severity in this model. However, ACE-I has not been tested in more immunologically relevant colitis models. AIM We hypothesized that ACE-I would decrease disease severity in an IL-10 knockout (-/-) colitis model. METHODS Colitis was induced by giving 10-week old IL-10-/- mice piroxicam (P.O.) for 14 days. The ACE-I enalaprilat was given transanally at a dose of 6.25 mg/kg for 21 days. Prednisolone (PSL) with or without enalaprilat were used as therapeutic, comparative groups. All groups were compared to a placebo treated group. Outcome measures were clinical course, histology, abundance of pro-inflammatory cytokines/chemokines, and epithelial barrier function. RESULTS Enalaprilat exhibited better survival (91 %) versus other treatment groups (PSL: 85.7 %, PSL + ACE-I: 71.4 %, placebo: 66.6 %). The ACE-I and PSL + ACE-I groups showed significantly better histological scores versus placebo mice. ACE-I and the PSL groups significantly reduced several pro-inflammatory cytokines versus placebo mice. FITC-dextran permeability was reduced in the ACE-I and PSL + ACE-I groups. Blood pressure was not affected in ACE-I treated mice compared to placebo mice. CONCLUSIONS ACE-I was effective in reducing severity of colitis in an IL-10-/- model. The addition of prednisolone minimally augmented this effect. The findings suggest that appropriately dosed ACE-I with or without steroids may be a new therapeutic agent for colitis.
Collapse
Affiliation(s)
- Ryo Sueyoshi
- Section of Pediatric Surgery, Department of Surgery, Mott Children’s Hospital, University of Michigan, 1540 E. Hospital Dr., SPC 4211, Ann Arbor, MI 48109-4211, USA
| | - Kathleen M. Woods Ignatoski
- Section of Pediatric Surgery, Department of Surgery, Mott Children’s Hospital, University of Michigan, 1540 E. Hospital Dr., SPC 4211, Ann Arbor, MI 48109-4211, USA
| | - Stephanie Daignault
- Biostatistics, Comprehensive Cancer Center, University of Michigan Health System, Ann Arbor, MI, USA
| | - Manabu Okawada
- Section of Pediatric Surgery, Department of Surgery, Mott Children’s Hospital, University of Michigan, 1540 E. Hospital Dr., SPC 4211, Ann Arbor, MI 48109-4211, USA
| | - Daniel H. Teitelbaum
- Section of Pediatric Surgery, Department of Surgery, Mott Children’s Hospital, University of Michigan, 1540 E. Hospital Dr., SPC 4211, Ann Arbor, MI 48109-4211, USA
| |
Collapse
|
928
|
Abstract
PURPOSE OF REVIEW To present and evaluate the recent findings that contribute to our understanding of the functional impact of diet on the enteric microbiome and outcomes of disease. RECENT FINDINGS Nutrients in excess and in deficiency have significant impact on gut microbial communities in both rodents and humans, acting directly on the microbiota or indirectly via altering host physiology. Furthermore, the effects of diet on the microbiome in determining health or disease can differ substantially depending on the age and environment of the individual. SUMMARY Dietary compounds can have profound short-term and long-term effects on the assemblage of the gut microbiome, which in turn affects the host-microbe interactions critically important for intestinal, metabolic, and immune homeostasis. Until recently, the mechanisms underlying these effects were poorly understood. However, new insights have now been gained, made possible through the application of advanced technologies and bioinformatics, novel experimental models, and human research. As a result, our conceptual framework for understanding the impact of diet on the gut microbiome, health, and disease has advanced considerably, bringing the promise of better tools of risk assessment, diagnostics, and therapeutic intervention in an age of personalized medicine.
Collapse
|
929
|
Schnupf P, Gaboriau-Routhiau V, Cerf-Bensussan N. Host interactions with Segmented Filamentous Bacteria: an unusual trade-off that drives the post-natal maturation of the gut immune system. Semin Immunol 2013; 25:342-51. [PMID: 24184014 DOI: 10.1016/j.smim.2013.09.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Segmented Filamentous Bacteria (SFB) are present in the gut microbiota of a large number of vertebrate species where they are found intimately attached to the intestinal epithelium. SFB has recently attracted considerable attention due to its outstanding capacity to stimulate innate and adaptive host immune responses without causing pathology. Recent genomic analysis placed SFB between obligate and facultative symbionts, unraveled its highly auxotrophic needs, and provided a rationale for the complex SFB life-style in close contact with the epithelium. Herein, we examine how the SFB life-style may underlie its potent immunostimulatory properties and discuss how the trade-off set up between SFB and its hosts can simultaneously help to establish and maintain the ecological niche of SFB in the intestine and drive the post-natal maturation of the host gut immune barrier.
Collapse
Affiliation(s)
- Pamela Schnupf
- INSERM, U989, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, and Institut IMAGINE, 75015 Paris, France; Institut Pasteur, Unité de Pathogénie Microbienne Moleculaire, 25-28 rue du Dr. Roux, 75015 Paris, France
| | | | | |
Collapse
|
930
|
Mani S, Boelsterli UA, Redinbo MR. Understanding and modulating mammalian-microbial communication for improved human health. Annu Rev Pharmacol Toxicol 2013; 3. [PMID: 27942535 PMCID: PMC5145265 DOI: 10.11131/2016/101199] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The molecular basis for the regulation of the intestinal barrier is a very fertile research area. A growing body of knowledge supports the targeting of various components of intestinal barrier function as means to treat a variety of diseases, including the inflammatory bowel diseases. Herein, we will summarize the current state of knowledge of key xenobiotic receptor regulators of barrier function, highlighting recent advances, such that the field and its future are succinctly reviewed. We posit that these receptors confer an additional dimension of host-microbe interaction in the gut, by sensing and responding to metabolites released from the symbiotic microbiota, in innate immunity and also in host drug metabolism. The scientific evidence for involvement of the receptors and its molecular basis for the control of barrier function and innate immunity regulation would serve as a rationale towards development of non-toxic probes and ligands as drugs.
Collapse
Affiliation(s)
- Sridhar Mani
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | | | | |
Collapse
|
931
|
Lactobacillus reuteri-specific immunoregulatory gene rsiR modulates histamine production and immunomodulation by Lactobacillus reuteri. J Bacteriol 2013; 195:5567-76. [PMID: 24123819 DOI: 10.1128/jb.00261-13] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human microbiome-derived strains of Lactobacillus reuteri potently suppress proinflammatory cytokines like human tumor necrosis factor (TNF) by converting the amino acid l-histidine to the biogenic amine histamine. Histamine suppresses mitogen-activated protein (MAP) kinase activation and cytokine production by signaling via histamine receptor type 2 (H2) on myeloid cells. Investigations of the gene expression profiles of immunomodulatory L. reuteri ATCC PTA 6475 highlighted numerous genes that were highly expressed during the stationary phase of growth, when TNF suppression is most potent. One such gene was found to be a regulator of genes involved in histidine-histamine metabolism by this probiotic species. During the course of these studies, this gene was renamed the Lactobacillus reuteri-specific immunoregulatory (rsiR) gene. The rsiR gene is essential for human TNF suppression by L. reuteri and expression of the histidine decarboxylase (hdc) gene cluster on the L. reuteri chromosome. Inactivation of rsiR resulted in diminished TNF suppression in vitro and reduced anti-inflammatory effects in vivo in a trinitrobenzene sulfonic acid (TNBS)-induced mouse model of acute colitis. A L. reuteri strain lacking an intact rsiR gene was unable to suppress colitis and resulted in greater concentrations of serum amyloid A (SAA) in the bloodstream of affected animals. The PhdcAB promoter region targeted by rsiR was defined by reporter gene experiments. These studies support the presence of a regulatory gene, rsiR, which modulates the expression of a gene cluster known to mediate immunoregulation by probiotics at the transcriptional level. These findings may point the way toward new strategies for controlling gene expression in probiotics by dietary interventions or microbiome manipulation.
Collapse
|
932
|
Knights D, Lassen KG, Xavier RJ. Advances in inflammatory bowel disease pathogenesis: linking host genetics and the microbiome. Gut 2013; 62:1505-10. [PMID: 24037875 PMCID: PMC3822528 DOI: 10.1136/gutjnl-2012-303954] [Citation(s) in RCA: 324] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Studies of the genetics underlying inflammatory bowel diseases have increased our understanding of the pathways involved in both ulcerative colitis and Crohn's disease and focused attention on the role of the microbiome in these diseases. Full understanding of pathogenesis will require a comprehensive grasp of the delicate homeostasis between gut bacteria and the human host. In this review, we present current evidence of microbiome-gene interactions in the context of other known risk factors and mechanisms, and describe the next steps necessary to pair genetic variant and microbiome sequencing data from patient cohorts. We discuss the concept of dysbiosis, proposing that the functional composition of the gut microbiome may provide a more consistent definition of dysbiosis and may more readily provide evidence of genome-microbiome interactions in future exploratory studies.
Collapse
Affiliation(s)
- Dan Knights
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA,Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kara G. Lassen
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ramnik J. Xavier
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA,Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
933
|
Dasgupta S, Kasper DL. Relevance of commensal microbiota in the treatment and prevention of inflammatory bowel disease. Inflamm Bowel Dis 2013; 19:2478-89. [PMID: 23846489 DOI: 10.1097/mib.0b013e318297d884] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Commensal microbiota that reside primarily in the gut of mammals influence the hosts' health to a great extent. Shaping of host immunity locally, a vital component of this influence, can have pro-inflammatory, anti-inflammatory, or neutral outcomes, presumably depending on the composition of the microbiota in an individual and type of molecules expressed in the individual members of the microbiota. Thus, these microbial species can be thought of as a reservoir of molecules that can be used to improve or worsen the condition of patients suffering from immunity or inflammation-driven pathologies like inflammatory bowel disease. In the current review, we elaborate, based on the literature available from murine models of disease and clinical case studies, the need to identify individual members of commensal microbiota that can precipitate or resolve inflammatory bowel disease. Therapeutic approaches could entail enrichment of members of microbiota (or molecules from these microbes), which induces expansion or enhancement of function of regulatory T cells or tolerogenic dendritic cells and reduce members that cause inflammation either directly or indirectly by influencing metabolic and other host molecules. Efficiency of bacteria-driven therapy would potentially be enhanced as we refine our approaches from the use of complete feces as done in fecal transplantation to utilization of microbiota-derived molecules as exemplified by the capsular polysaccharide A from the human gut commensal Bacteroides fragilis. We also highlight the advantages and disadvantages of each approach, defining a natural alternative to the current chemical-based immunosuppressive regimen for patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Suryasarathi Dasgupta
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
934
|
Zhang ZZ, Shang QH, Jin HY, Song B, Oudit GY, Lu L, Zhou T, Xu YL, Gao PJ, Zhu DL, Penninger JM, Zhong JC. Cardiac protective effects of irbesartan via the PPAR-gamma signaling pathway in angiotensin-converting enzyme 2-deficient mice. J Transl Med 2013; 11:229. [PMID: 24067190 PMCID: PMC3850664 DOI: 10.1186/1479-5876-11-229] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/24/2013] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Angiotensin-converting enzyme 2 (ACE2), a monocarboxypeptidase which metabolizes angiotensin II (Ang II) to generate Ang-(1-7), has been shown to prevent cardiac hypertrophy and injury but the mechanism remains elusive. Irbesartan has the dual actions of angiotensin receptor blockade and peroxisome proliferator-activated receptor-γ (PPARγ) activation. We hypothesized that irbesartan would exert its protective effects on ACE2 deficiency-mediated myocardial fibrosis and cardiac injury via the PPARγ signaling. METHODS 10-week-old ACE2 knockout (ACE2KO; Ace2(-/y)) mice received daily with irbesartan (50 mg/kg) or saline for 2 weeks. The wild-type mice (Ace2(+/y)) were used to the normal controls. We examined changes in myocardial ultrastructure, fibrosis-related genes and pathological signaling by real-time PCR gene array, Western blotting, Masson trichrome staining and transmission electron microscope analyses, respectively. RESULTS Compared with the Ace2(+/y) mice, cardiac expression of PPARα and PPARγ were reduced in Ace2(-/y) mice and the myocardial collagen volume fraction (CVF) and expression of fibrosis-related genes were increased, including transforming growth factor-β1 (TGFβ1), connective tissue growth factor (CTGF), collagen I and collagen III. Moreover, ACE2 deficiency triggered cardiac hypertrophy, increased myocardial fibrosis and adverse ultrastructure injury in ACE2KO hearts with higher levels of atrial natriuretic factor (ANF) and phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), without affecting cardiac systolic function. Intriguingly, treatment with irbesartan significantly reversed ACE2 deficiency-mediated pathological hypertrophy and myocardial fibrosis in Ace2(-/y) mice linked with enhancement of plasma Ang-(1-7) level and downregulation of AT1 receptor in heart. Consistent with attenuation of myocardial fibrosis and ultrastructure injury, the myocardial CVF and levels of ANF, TGFβ1, CTGF, collagen I, collagen III and phosphorylated ERK1/2 were lower, and expression of PPARγ was higher in ACE2KO mice in response to irbesartan treatment, without affecting cardiac expression of PPARα, PPARδ, β-myosin heavy chain, TGFβ2 and fibronectin. CONCLUSIONS We conclude that irbesartan prevents ACE2 deficiency-mediated pathological hypertrophy and myocardial fibrosis in ACE2 mutant mice via activation of the PPARγ signaling and suppression of the TGFβ-CTGF-ERK signaling, resulting in attenuation of myocardial injury. Drugs targeting ACE2 and PPARγ represent potential candidates to prevent and treat myocardial injury and related cardiac disorders.
Collapse
Affiliation(s)
- Zhen-Zhou Zhang
- State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Hypertension, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
935
|
All systems normal. Curr Opin Nephrol Hypertens 2013; 22:531-2. [DOI: 10.1097/mnh.0b013e3283640080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
936
|
Burcelin R, Serino M, Chabo C, Garidou L, Pomié C, Courtney M, Amar J, Bouloumié A. Metagenome and metabolism: the tissue microbiota hypothesis. Diabetes Obes Metab 2013; 15 Suppl 3:61-70. [PMID: 24003922 DOI: 10.1111/dom.12157] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 05/22/2013] [Indexed: 12/14/2022]
Abstract
Over the last decade, the research community has revealed the role of a new organ: the intestinal microbiota. It is considered as a symbiont that is part of our organism since, at birth, it educates the immune system and contributes to the development of the intestinal vasculature and most probably the nervous system. With the advent of new generation sequencing techniques, a catalogue of genes that belong to this microbiome has been established that lists more than 5 million non-redundant genes called the metagenome. Using germ free mice colonized with the microbiota from different origins, it has been formally demonstrated that the intestinal microbiota causes the onset of metabolic diseases. Further to the role of point mutations in our genome, the microbiota can explain the on-going worldwide pandemic of obesity and diabetes, its dissemination and family inheritance, as well as the diversity of the associated metabolic phenotypes. More recently, the discovery of bacterial DNA within host tissues, such as the liver, the adipose tissue and the blood, which establishes a tissue microbiota, introduces new opportunities to identify targets and predictive biomarkers based on the host to microbiota interaction, as well as to define new strategies for pharmacological, immunomodulatory vaccines and nutritional applications.
Collapse
Affiliation(s)
- Rémy Burcelin
- Institut National de Santé et de Recherche Médicale (INSERM), U1048, Toulouse, France.
| | | | | | | | | | | | | | | |
Collapse
|
937
|
Nishida N, Sasaki M, Kurihara M, Ichimaru S, Wakita M, Bamba S, Andoh A, Fujiyama Y, Amagai T. Changes of energy metabolism, nutritional status and serum cytokine levels in patients with Crohn's disease after anti-tumor necrosis factor-α therapy. J Clin Biochem Nutr 2013; 53:122-7. [PMID: 24062610 PMCID: PMC3774925 DOI: 10.3164/jcbn.13-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 04/02/2013] [Indexed: 12/11/2022] Open
Abstract
We investigated the effects of treatment with antibodies against tumor necrosis factor (TNF)-α on energy metabolism, nutritional status, serum cytokine levels in patients with Crohn’s disease (CD). Twelve patients were enrolled. Resting energy expenditure (REE) levels were measured by indirect calorimetry. Crohn’s disease activity index (CDAI) significantly decreased after treatment with anti-TNF-α therapy. Anti-TNF-α therapy did not affect REE, but respiratory quotient (RQ) significantly increased after treatment. Serum interleukin-6 levels were significantly decreased and RQ were significantly increased in high REE (≥25 kcal/kg/day) group as compared to low REE (<25 kcal/kg/day) group. In conclusion, high REE value on admission is a predictive factor for good response to treatment with anti-TNF-α antibodies in active CD patients.
Collapse
Affiliation(s)
- Nao Nishida
- Department of Food Science and Nutrition Major, Graduate School of Human Environment Sciences, Mukogawa Women's University Graduate School, 6-46 Ikebiraki-cho, Nishinomiya 663-8558, Japan ; Division of Clinical Nutrition, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
938
|
The role of KCNQ1 in mouse and human gastrointestinal cancers. Oncogene 2013; 33:3861-8. [PMID: 23975432 PMCID: PMC3935979 DOI: 10.1038/onc.2013.350] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/07/2013] [Accepted: 07/04/2013] [Indexed: 12/19/2022]
Abstract
Kcnq1, which encodes for the pore-forming alpha subunit of a voltage-gated potassium channel, was identified as a gastrointestinal (GI) tract cancer susceptibility gene in multiple Sleeping Beauty DNA transposon-based forward genetic screens in mice. To confirm that Kcnq1 has a functional role in GI tract cancer we created ApcMin mice that carried a targeted deletion mutation in Kcnq1. Results demonstrated that Kcnq1 is a tumor suppressor gene as Kcnq1 mutant mice developed significantly more intestinal tumors, especially in the proximal small intestine and colon, some of these tumors progressed to become aggressive adenocarcinomas. Gross tissue abnormalities were also observed in the rectum, pancreas and stomach. Colon organoid formation was significantly increased in organoids created from Kcnq1 mutant mice compared with wildtype littermate controls, suggesting a role for Kcnq1 in regulation of the intestinal crypt stem cell compartment. To identify gene expression changes due to loss of Kcnq1 we carried out microarray studies in colon and proximal small intestine. We identified altered genes involved in innate immune responses, goblet and Paneth cell function, ion channels, intestinal stem cells, EGFR and other growth regulatory signaling pathways. We also found genes implicated in inflammation and in cellular detoxification. Pathway analysis using Ingenuity Pathway Analysis (IPA) and gene set enrichment analysis (GSEA) confirmed the importance of these gene clusters and further identified significant overlap with genes regulated by MUC2 and CFTR, two important regulators of intestinal homeostasis. To investigate the role of KCNQ1 in human colorectal cancer (CRC) we measured protein levels of KCNQ1 by immunohistochemistry in tissue microarrays containing samples from CRC patients with liver metastases who had undergone hepatic resection. Results showed that low expression of KCNQ1 expression was significantly associated with poor overall survival (OS).
Collapse
|
939
|
Ostaff MJ, Stange EF, Wehkamp J. Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol Med 2013; 5:1465-83. [PMID: 24039130 PMCID: PMC3799574 DOI: 10.1002/emmm.201201773] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/19/2013] [Accepted: 07/04/2013] [Indexed: 12/17/2022] Open
Abstract
We survive because we adapted to a world of microorganisms. All our epithelial surfaces participate in keeping up an effective barrier against microbes while not initiating ongoing inflammatory processes and risking collateral damage to the host. Major players in this scenario are antimicrobial peptides (AMPs). Such broad-spectrum innate antibiotics are in part produced by specialized cells but also widely sourced from all epithelia as well as circulating inflammatory cells. AMPs belong to an ancient defense system found in all organisms and participated in a preservative co-evolution with a complex microbiome. Particularly interesting interactions between host barrier and microbiota can be found in the gut. The intestinal cell lining not only has to maintain a tightly regulated homeostasis during its high-throughput regeneration, but also a balanced relationship towards an extreme number of mutualistic or commensal inhabitants. Recent research suggests that advancing our understanding of the circumstances of such balanced and sometimes imbalanced interactions between gut microbiota and host AMPs should have therapeutic implications for different intestinal disorders.
Collapse
Affiliation(s)
- Maureen J Ostaff
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Germany
| | | | | |
Collapse
|
940
|
Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D'Angelo C, Massi-Benedetti C, Fallarino F, Carvalho A, Puccetti P, Romani L. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013; 39:372-85. [PMID: 23973224 DOI: 10.1016/j.immuni.2013.08.003] [Citation(s) in RCA: 1635] [Impact Index Per Article: 136.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 05/24/2013] [Indexed: 02/07/2023]
Abstract
Endogenous tryptophan (Trp) metabolites have an important role in mammalian gut immune homeostasis, yet the potential contribution of Trp metabolites from resident microbiota has never been addressed experimentally. Here, we describe a metabolic pathway whereby Trp metabolites from the microbiota balance mucosal reactivity in mice. Switching from sugar to Trp as an energy source (e.g., under conditions of unrestricted Trp availability), highly adaptive lactobacilli are expanded and produce an aryl hydrocarbon receptor (AhR) ligand-indole-3-aldehyde-that contributes to AhR-dependent Il22 transcription. The resulting IL-22-dependent balanced mucosal response allows for survival of mixed microbial communities yet provides colonization resistance to the fungus Candida albicans and mucosal protection from inflammation. Thus, the microbiota-AhR axis might represent an important strategy pursued by coevolutive commensalism for fine tuning host mucosal reactivity contingent on Trp catabolism.
Collapse
MESH Headings
- Animals
- Basic Helix-Loop-Helix Transcription Factors/deficiency
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Candida albicans/immunology
- Candidiasis/immunology
- Energy Metabolism
- Female
- Gastrointestinal Tract/immunology
- Gastrointestinal Tract/metabolism
- Gastrointestinal Tract/microbiology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoles/metabolism
- Interleukin-17/deficiency
- Interleukin-17/genetics
- Interleukins/metabolism
- Limosilactobacillus reuteri/growth & development
- Limosilactobacillus reuteri/immunology
- Limosilactobacillus reuteri/metabolism
- Metagenome
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Myeloid Differentiation Factor 88/deficiency
- Myeloid Differentiation Factor 88/genetics
- Probiotics
- Receptors, Aryl Hydrocarbon/deficiency
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Toll-Like Receptor 2/deficiency
- Toll-Like Receptor 2/genetics
- Tryptophan/chemistry
- Tryptophan/metabolism
- Interleukin-22
Collapse
Affiliation(s)
- Teresa Zelante
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia 06132, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
941
|
Perlot T, Penninger JM. ACE2 - from the renin-angiotensin system to gut microbiota and malnutrition. Microbes Infect 2013; 15:866-73. [PMID: 23962453 PMCID: PMC7110844 DOI: 10.1016/j.micinf.2013.08.003] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/06/2013] [Indexed: 02/07/2023]
Abstract
The renin-angiotensin system (RAS) is a complex network that regulates blood pressure, electrolyte and fluid homeostasis, as well as the function of several organs. Angiotensin-converting enzyme 2 (ACE2) was identified as an enzyme that negatively regulates the RAS by converting Ang II, the main bioactive molecule of the RAS, to Ang 1-7. Thus, ACE2 counteracts the role of angiotensin-converting enzyme (ACE) which generates Ang II from Ang I. ACE and ACE2 have been implicated in several pathologies such as cardiovascular and renal disease or acute lung injury. In addition, ACE2 has functions independent of the RAS: ACE2 is the receptor for the SARS coronavirus and ACE2 is essential for expression of neutral amino acid transporters in the gut. In this context, ACE2 modulates innate immunity and influences the composition of the gut microbiota, which can explain diarrhea and intestinal inflammation observed in Hartnup disorder, Pellagra, or under conditions of severe malnutrition. Here we review and discuss the diverse functions of ACE2 and its relevance to human pathologies.
Collapse
Affiliation(s)
- Thomas Perlot
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | | |
Collapse
|
942
|
Kumar JS, Subramanian VS, Kapadia R, Kashyap ML, Said HM. Mammalian colonocytes possess a carrier-mediated mechanism for uptake of vitamin B3 (niacin): studies utilizing human and mouse colonic preparations. Am J Physiol Gastrointest Liver Physiol 2013; 305:G207-13. [PMID: 23744738 PMCID: PMC3742858 DOI: 10.1152/ajpgi.00148.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Niacin (vitamin B3; nicotinic acid) plays an important role in maintaining redox state of cells and is obtained from endogenous and exogenous sources. The latter source has generally been assumed to be the dietary niacin, but another exogenous source that has been ignored is the niacin that is produced by the normal microflora of the large intestine. For this source of niacin to be bioavailable, it needs to be absorbed, but little is known about the ability of the large intestine to absorb niacin and the mechanism involved. Here we addressed these issues using the nontransformed human colonic epithelial NCM460 cells, native human colonic apical membrane vesicles (AMV) isolated from organ donors, and mouse colonic loops in vivo as models. Uptake of ³H-nicotinic acid by NCM460 cells was: 1) acidic pH (but not Na⁺) dependent; 2) saturable (apparent Km = 2.5 ± 0.8 μM); 3) inhibited by unlabeled nicotinic acid, nicotinamide, and probenecid; 4) neither affected by other bacterially produced monocarboxylates, monocarboxylate transport inhibitor, or by substrates of the human organic anion transporter-10; 5) affected by modulators of the intracellular protein tyrosine kinase- and Ca²⁺-calmodulin-regulatory pathways; and 6) adaptively regulated by extracellular nicotinate level. Uptake of nicotinic acid by human colonic AMV in vitro and by mouse colonic loops in vivo was also carrier mediated. These findings report, for the first time, that mammalian colonocytes possess a high-affinity carrier-mediated mechanism for nicotinate uptake and show that the process is affected by intracellular and extracellular factors.
Collapse
Affiliation(s)
- Jeyan S. Kumar
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine; ,2Department of Medical Research, Veterans Affairs Medical Center, Long Beach, California
| | - Veedamali S. Subramanian
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine; ,2Department of Medical Research, Veterans Affairs Medical Center, Long Beach, California
| | - Rubina Kapadia
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine; ,2Department of Medical Research, Veterans Affairs Medical Center, Long Beach, California
| | - Moti L. Kashyap
- 2Department of Medical Research, Veterans Affairs Medical Center, Long Beach, California
| | - Hamid M. Said
- 1Departments of Medicine, Physiology and Biophysics, University of California, Irvine; ,2Department of Medical Research, Veterans Affairs Medical Center, Long Beach, California
| |
Collapse
|
943
|
Patel SK, Velkoska E, Burrell LM. Emerging markers in cardiovascular disease: Where does angiotensin-converting enzyme 2 fit in? Clin Exp Pharmacol Physiol 2013; 40:551-9. [DOI: 10.1111/1440-1681.12069] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/13/2013] [Accepted: 02/19/2013] [Indexed: 01/21/2023]
Affiliation(s)
- Sheila K Patel
- Department of Medicine; University of Melbourne; Austin Health; Melbourne Victoria Australia
| | - Elena Velkoska
- Department of Medicine; University of Melbourne; Austin Health; Melbourne Victoria Australia
| | - Louise M Burrell
- Department of Medicine; University of Melbourne; Austin Health; Melbourne Victoria Australia
| |
Collapse
|
944
|
Moss BG, Chugani DC. Increased risk of very low birth weight, rapid postnatal growth, and autism in underweight and obese mothers. Am J Health Promot 2013; 28:181-8. [PMID: 23875984 DOI: 10.4278/ajhp.120705-quan-325] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE To determine whether prepregnancy weight was associated with children's birth weight, early physical growth, and autism diagnosis. DESIGN Early Childhood Longitudinal Study-Birth Cohort data. SETTING United States. SUBJECTS Representative sample of U.S. children followed from birth through kindergarten (n = 4800). Also, a subpopulation of the very low birth weight children was examined (n = 500). MEASURES Maternal variables included age and prepregnancy body mass index. Changes in children's height, weight, and head circumference between 9 months and 2 years were used as growth metrics. Children's sex, age, birth weight, and reported autism were also considered. ANALYSIS Logistic and multinomial logistic models assessed the impact of prepregnancy weight on birth weight and children's subsequent rate of physical growth and autism. RESULTS Children born to underweight or obese mothers had increased odds of very low birth weight. Very low birth weight was related to rapid height and weight growth and more than twice the likelihood to subsequently be diagnosed with autism. For the subgroup of very low birth weight children, rapid head growth was related to a fivefold increase in the odds of autism. After accounting for the impact birth weight and growth rates, we found prepregnancy weight indirectly impacted autism risk. CONCLUSION Being underweight or obese during prepregnancy indirectly increased risk for autism from increased odds of low birth weight and accelerated postnatal growth.
Collapse
|
945
|
Blankfield A. Kynurenine Pathway Pathologies: do Nicotinamide and Other Pathway Co-Factors have a Therapeutic Role in Reduction of Symptom Severity, Including Chronic Fatigue Syndrome (CFS) and Fibromyalgia (FM). Int J Tryptophan Res 2013; 6:39-45. [PMID: 23922501 PMCID: PMC3729338 DOI: 10.4137/ijtr.s11193] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The definition of dual tryptophan pathways has increased the understanding of the mind-body, body-mind dichotomy. The serotonergic pathway highlights the primary (endogenous) psychiatric disorders. The up-regulation of the kynurenine pathway by physical illnesses can cause neuropathic and immunological disorders1 associated with secondary neuropsychiatric symptoms. Tryptophan and nicotinamide deficiencies fall within the protein energy malnutrition (PEM) spectrum. They can arise if the kynurenine pathway is stressed by primary or secondary inflammatory conditions and the consequent imbalance of available catabolic/anabolic substrates may adversely influence convalescent phase efficiency. The replacement of depleted or reduced NAD+ levels and other cofactors can perhaps improve the clinical management of these disorders. Chronic fatigue syndrome (CFS) and fibromyalgia (FM) appear to meet the criteria of a tryptophan-kynurenine pathway disorder with potential neuroimmunological sequelae. Aspects of some of the putative precipitating factors have been previously outlined.2,3 An analysis of the areas of metabolic dysfunction will focus on future directions for research and management.
Collapse
|
946
|
Kunisawa J, Kiyono H. Vitamin-mediated regulation of intestinal immunity. Front Immunol 2013; 4:189. [PMID: 23874335 PMCID: PMC3708512 DOI: 10.3389/fimmu.2013.00189] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 06/26/2013] [Indexed: 12/17/2022] Open
Abstract
The intestine is exposed continuously to complex environments created by numerous injurious and beneficial non-self antigens. The unique mucosal immune system in the intestine maintains the immunologic homeostasis between the host and the external environment. Crosstalk between immunocompetent cells and endogenous (e.g., cytokines and chemokines) as well as exogenous factors (e.g., commensal bacteria and dietary materials) achieves the vast diversity of intestinal immune functions. In addition to their vital roles as nutrients, vitamins now also are known to have immunologically crucial functions, specifically in regulating host immune responses. In this review, we focus on the immunologic functions of vitamins in regulating intestinal immune responses and their roles in moderating the fine balance between physiologic and pathologic conditions of the intestine.
Collapse
Affiliation(s)
- Jun Kunisawa
- Laboratory of Vaccine Materials, National Institute of Biomedical Innovation , Osaka , Japan
| | | |
Collapse
|
947
|
Bindels LB, Delzenne NM. Muscle wasting: the gut microbiota as a new therapeutic target? Int J Biochem Cell Biol 2013; 45:2186-90. [PMID: 23831839 DOI: 10.1016/j.biocel.2013.06.021] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/19/2013] [Accepted: 06/25/2013] [Indexed: 12/25/2022]
Abstract
Muscle wasting is characterized by a loss of muscle mass and strength, and occurs in several pathological conditions such as cancer, chronic heart failure, chronic infection and malnutrition. Muscle wasting can be caused by inflammation and inappropriate nutritional status. Interestingly, gut microbiota has recently been proposed as an environmental factor involved, among others, in energy sparing from the diet, and in the regulation of host immunity and metabolism. This review presents evidence supporting the existence of a gut microbiota-muscle axis and discusses the potential role and therapeutic interest of gut microbiota in muscle wasting, specifically in the context of cancer and malnutrition. This review also proposes possible molecular mechanisms underlying the gut microbiota-muscle axis. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
948
|
Abstract
The gut flora is composed of a huge number of diverse, well-adapted symbionts that interact with epithelial lining throughout the host's entire life. Not all commensals have the same ability to maintain quiescent, protective inflammation. Importantly, instability in the composition of gut microbial communities (referred to as dysbiosis) has been linked to loss of gut barrier in the context of common human illnesses with increasing socio-economic impacts, such as Crohn disease and colorectal cancer. Our recent findings suggest that disease-predisposing dysbiosis can now be intentionally manipulated by targeting the major Crohn disease-predisposing NOD2 gene. That knowledge will not only add a new dimension to the often overlooked microbiology of Crohn disease and colorectal cancer, but will also have a broad impact on biomedical sciences worldwide.
Collapse
Affiliation(s)
- Thomas Secher
- INRA; Institut National de la Santé et de la Recherche Médicale; UMR 1043; Toulouse, France
| | - Sylvain Normand
- University Lille Nord de France; Lille, France,Institut Pasteur de Lille; Center for Infection and Immunity of Lille; Lille, France,Centre National de la Recherche Scientifique; Unité Mixte de Recherche 8204; Lille, France,Institut National de la Santé et de la Recherche Médicale; U1019; Lille, France
| | - Mathias Chamaillard
- University Lille Nord de France; Lille, France,Institut Pasteur de Lille; Center for Infection and Immunity of Lille; Lille, France,Centre National de la Recherche Scientifique; Unité Mixte de Recherche 8204; Lille, France,Institut National de la Santé et de la Recherche Médicale; U1019; Lille, France,Correspondence to: Mathias Chamaillard,
| |
Collapse
|
949
|
Gong HL, Shi Y, Zhou L, Wu CP, Cao PY, Tao L, Xu C, Hou DS, Wang YZ. The Composition of Microbiome in Larynx and the Throat Biodiversity between Laryngeal Squamous Cell Carcinoma Patients and Control Population. PLoS One 2013; 8:e66476. [PMID: 23824228 PMCID: PMC3688906 DOI: 10.1371/journal.pone.0066476] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/07/2013] [Indexed: 01/13/2023] Open
Abstract
The throat is an ecological assemblage involved human cells and microbiota, and the colonizing bacteria are important factors in balancing this environment. However, this bacterial community profile has thus been poorly investigated. The purpose of this study was to investigate the microbial biology of the larynx and to analyze the throat biodiversity in laryngeal carcinoma patients compared to a control population in a case-control study. Barcoded pyrosequencing analysis of the 16S rRNA gene was used. We collected tissue samples from 29 patients with laryngeal carcinoma and 31 control patients with vocal cord polyps. The findings of high-quality sequence datasets revealed 218 genera from 13 phyla in the laryngeal mucosa. The predominant communities of phyla in the larynx were Firmicutes (54%), Fusobacteria (17%), Bacteroidetes (15%), Proteobacteria (11%), and Actinobacteria (3%). The leading genera were Streptococcus (36%), Fusobacterium (15%), Prevotella (12%), Neisseria (6%), and Gemella (4%). The throat bacterial compositions were highly different between laryngeal carcinoma subjects and control population (p = 0.006). The abundance of the 26 genera was significantly different between the laryngeal cancer and control groups by metastats analysis (p<0.05). Fifteen genera may be associated with laryngeal carcinoma by partial least squares discriminant analysis (p<0.001). In summary, this study revealed the microbiota profiles in laryngeal mucosa from tissue specimens. The compositions of bacteria community in throat were different between laryngeal cancer patients and controls, and probably were related with this carcinoma. The disruption of this bio-ecological niche might be a risk factor for laryngeal carcinoma.
Collapse
Affiliation(s)
- Hong-Li Gong
- Department of Otolaryngology, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai, China
| | - Yi Shi
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- * E-mail: (LZ); (YS)
| | - Liang Zhou
- Department of Otolaryngology, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai, China
- * E-mail: (LZ); (YS)
| | - Chun-Ping Wu
- Department of Otolaryngology, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai, China
| | - Peng-Yu Cao
- Department of Otolaryngology, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai, China
| | - Lei Tao
- Department of Otolaryngology, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai, China
| | - Chen Xu
- Shanghai Key Laboratory for Reproductive Medicine, Department of Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong-Sheng Hou
- Shanghai Key Laboratory for Reproductive Medicine, Department of Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue-Zhu Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Sequencing Centre, Shanghai, China
| |
Collapse
|
950
|
Faria AMC, Gomes-Santos AC, Gonçalves JL, Moreira TG, Medeiros SR, Dourado LPA, Cara DC. Food components and the immune system: from tonic agents to allergens. Front Immunol 2013; 4:102. [PMID: 23730302 PMCID: PMC3656403 DOI: 10.3389/fimmu.2013.00102] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/20/2013] [Indexed: 12/13/2022] Open
Abstract
The intestinal mucosa is the major site of contact with antigens, and it houses the largest lymphoid tissue in the body. In physiological conditions, microbiota and dietary antigens are the natural sources of stimulation for the gut-associated lymphoid tissues (GALT) and for the immune system as a whole. Germ-free models have provided some insights on the immunological role of gut antigens. However, most of the GALT is not located in the large intestine, where gut microbiota is prominent. It is concentrated in the small intestine where protein absorption takes place. In this review, we will address the involvement of food components in the development and the function of the immune system. Studies in mice have already shown that dietary proteins are critical elements for the developmental shift of the immature neonatal immune profile into a fully developed immune system. The immunological effects of other food components (such as vitamins and lipids) will also be addressed. Most of the cells in the GALT are activated and local pro-inflammatory mediators are abundant. Regulatory elements are known to provide a delicate yet robust balance that maintains gut homeostasis. Usually antigenic contact in the gut induces two major immune responses, oral tolerance and production of secretory IgA. However, under pathological conditions mucosal homeostasis is disturbed resulting in inflammatory reactions such as food hypersensitivity. Food allergy development depends on many factors such as genetic predisposition, biochemical features of allergens, and a growing array of environmental elements. Neuroimmune interactions are also implicated in food allergy and they are examples of the high complexity of the phenomenon. Recent findings on the gut circuits triggered by food components will be reviewed to show that, far beyond their role as nutrients, they are critical players in the operation of the immune system in health and disease.
Collapse
Affiliation(s)
- Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Minas Gerais, Brazil ; Instituto de Investigação em Imunologia (iii) São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|