251
|
Gabuzda D, Jamieson BD, Collman RG, Lederman MM, Burdo TH, Deeks SG, Dittmer DP, Fox HS, Funderburg NT, Pahwa SG, Pandrea I, Wilson CC, Hunt PW. Pathogenesis of Aging and Age-related Comorbidities in People with HIV: Highlights from the HIV ACTION Workshop. Pathog Immun 2020; 5:143-174. [PMID: 32856008 PMCID: PMC7449259 DOI: 10.20411/pai.v5i1.365] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
People with HIV (PWH) experience accentuated biological aging, as defined by markers of inflammation, immune dysfunction, and the epigenetic clock. They also have an elevated risk of multiple age-associated comorbidities. To discuss current knowledge, research gaps, and priorities in aging and age-related comorbidities in treated HIV infection, the NIH program staff organized a workshop held in Bethesda, Maryland in September 2019. This review article describes highlights of discussions led by the Pathogenesis/Basic Science Research working group that focused on three high priority topics: immunopathogenesis; the microbiome/virome; and aging and senescence. We summarize knowledge in these fields and describe key questions for research on the pathogenesis of aging and age-related comorbidities in PWH. Understanding the drivers and mechanisms underlying accentuated biological aging is a high priority that will help identify potential therapeutic targets to improve healthspan in older PWH.
Collapse
Affiliation(s)
- Dana Gabuzda
- Department of Cancer Immunology and Virology; Dana-Farber Cancer Institute; Boston, Massachusetts; Department of Neurology; Harvard Medical School; Boston, Massachusetts
| | - Beth D Jamieson
- Department of Medicine; David Geffen School of Medicine; University of California; Los Angeles, California
| | - Ronald G Collman
- Department of Medicine; University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania
| | - Michael M Lederman
- Department of Medicine; Case Western Reserve University School of Medicine; Cleveland, Ohio
| | - Tricia H Burdo
- Department of Neuroscience; Lewis Katz School of Medicine; Temple University; Philadelphia, Pennsylvania
| | - Steven G Deeks
- Department of Medicine; University of California; San Francisco, California
| | - Dirk P Dittmer
- Department of Microbiology and Immunology; University of North Carolina School of Medicine; Chapel Hill, North Carolina
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience; University of Nebraska Medical Center; Omaha, Nebraska
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science; School of Health and Rehabilitation Sciences; Ohio State University College of Medicine; Columbus, Ohio
| | - Savita G Pahwa
- Department of Microbiology and Immunology; University of Miami Miller School of Medicine; Miami, Florida
| | - Ivona Pandrea
- Department of Microbiology and Molecular Genetics; School of Medicine; University of Pittsburgh; Pittsburgh, Pennsylvania
| | - Cara C Wilson
- Department of Medicine; Division of Infectious Diseases; University of Colorado Anschutz Medical Campus; Aurora, Colorado
| | - Peter W Hunt
- Department of Medicine; University of California; San Francisco, California
| |
Collapse
|
252
|
Simionescu G, Ilie OD, Ciobica A, Doroftei B, Maftei R, Grab D, McKenna J, Dhunna N, Mavroudis I, Anton E. Mini-Review on the Possible Interconnections between the Gut-Brain Axis and the Infertility-Related Neuropsychiatric Comorbidities. Brain Sci 2020; 10:brainsci10060384. [PMID: 32560488 PMCID: PMC7349587 DOI: 10.3390/brainsci10060384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/26/2022] Open
Abstract
Both the gut-brain axis (GBA) and the hypothalamic–pituitary–adrenal (HPA) axis remain an intriguing yet obscure network with a strong influence over other systems of organs. Recent reports have sought to describe the multitude of harmful stressors that may impact the HPA axis along with the interconnections between these. This has improved our knowledge of how the underlying mechanisms working to establish homeostasis are affected. A disruption to the HPA axis can amplify the chances of gastrointestinal deficiencies, whilst also increasing the risk of a wide spectrum of neuropsychiatric disorders. Thus, the influence of microorganisms found throughout the digestive tract possess the ability to affect both physiology and behaviour by triggering responses, which may be unfavourable. This is sometimes the case in of infertility. Numerous supplements have been formulated with the intention of rebalancing the gut microflora. Accordingly, the gut flora may alter the pharmacokinetics of drugs used as part of fertility treatments, potentially exacerbating the predisposition for various neurological disorders, regardless of the age and gender.
Collapse
Affiliation(s)
- Gabriela Simionescu
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, No 16, 700115 Iasi, Romania; (G.S.); (D.G.); (E.A.)
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No 34, 700038 Iasi, Romania;
- Origyn Fertility Center, Palace Street, No 3C, 700032 Iasi, Romania
| | - Ovidiu-Dumitru Ilie
- Department of Research, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No 11, 700505 Iasi, Romania; (O.-D.I.); (A.C.)
| | - Alin Ciobica
- Department of Research, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No 11, 700505 Iasi, Romania; (O.-D.I.); (A.C.)
| | - Bogdan Doroftei
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, No 16, 700115 Iasi, Romania; (G.S.); (D.G.); (E.A.)
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No 34, 700038 Iasi, Romania;
- Origyn Fertility Center, Palace Street, No 3C, 700032 Iasi, Romania
- Correspondence:
| | - Radu Maftei
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No 34, 700038 Iasi, Romania;
- Origyn Fertility Center, Palace Street, No 3C, 700032 Iasi, Romania
- Department of Morphostructural Sciences, Faculty of Medicine, University of Medicine and Pharmacy “Grigore. T. Popa” Iasi, University Street, No 16, 700115 Iasi, Romania
| | - Delia Grab
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, No 16, 700115 Iasi, Romania; (G.S.); (D.G.); (E.A.)
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No 34, 700038 Iasi, Romania;
| | - Jack McKenna
- York Hospital, Wigginton Road Clifton, York YO31 8HE, UK;
| | - Nitasha Dhunna
- Mid Yorkshrie Hospitals NHS Trust, Pinderfields Hospital, Wakefield WF1 4DG, UK;
| | - Ioannis Mavroudis
- Leeds Teaching Hospitals NHS Trust, Great George St, Leeds LS1 3EX, UK;
- Laboratory of Neuropathology and Electron Microscopy, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Emil Anton
- Department of Mother and Child Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, No 16, 700115 Iasi, Romania; (G.S.); (D.G.); (E.A.)
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No 34, 700038 Iasi, Romania;
| |
Collapse
|
253
|
Nannini G, Meoni G, Amedei A, Tenori L. Metabolomics profile in gastrointestinal cancers: Update and future perspectives. World J Gastroenterol 2020; 26:2514-2532. [PMID: 32523308 PMCID: PMC7265149 DOI: 10.3748/wjg.v26.i20.2514] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/11/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023] Open
Abstract
Despite recent progress in diagnosis and therapy, gastrointestinal (GI) cancers remain one of the most important causes of death with a poor prognosis due to late diagnosis. Serum tumor markers and detection of occult blood in the stool are the current tests used in the clinic of GI cancers; however, these tests are not useful as diagnostic screening since they have low specificity and low sensitivity. Considering that one of the hallmarks of cancer is dysregulated metabolism and metabolomics is an optimal approach to illustrate the metabolic mechanisms that belong to living systems, is now clear that this -omics could open a new way to study cancer. In the last years, nuclear magnetic resonance (NMR) metabolomics has demonstrated to be an optimal approach for diseases' diagnosis nevertheless a few studies focus on the NMR capability to find new biomarkers for early diagnosis of GI cancers. For these reasons in this review, we will give an update on the status of NMR metabolomic studies for the diagnosis and development of GI cancers using biological fluids.
Collapse
Affiliation(s)
- Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Gaia Meoni
- Giotto Biotech Srl, and CERM (University of Florence), Florence 50019, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi, Florence 50134, Italy
| | - Leonardo Tenori
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Florence 50019, Italy
| |
Collapse
|
254
|
Patil Y, Gooneratne R, Ju XH. Interactions between host and gut microbiota in domestic pigs: a review. Gut Microbes 2020; 11:310-334. [PMID: 31760878 PMCID: PMC7524349 DOI: 10.1080/19490976.2019.1690363] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/16/2019] [Accepted: 11/04/2019] [Indexed: 02/03/2023] Open
Abstract
It is well established that pig gut microbiota plays a critical role in maintaining metabolic homeostasis as well as in a myriad of physiological, neurological and immunological functions; including protection from pathogens and digestion of food materials - some of which would be otherwise indigestible by the pig. A rich and diverse gut microbial ecosystem (balanced microbiota) is the hallmark of good health; while qualitative and quantitative perturbations in the microbial composition can lead to development of various diseases. Alternatively, diseases caused by stressors or other factors have been shown to negatively impact the microbiota. This review focuses primarily on how commensal microorganisms in the gastrointestinal tract of pigs influence biochemical, physiological, immunological, and metabolic processes within the host animal.
Collapse
Affiliation(s)
- Yadnyavalkya Patil
- College of Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, China
- Faculty of Agriculture and Life Sciences, Department of Wine, Food, and Molecular Biosciences, Lincoln University, Lincoln, Canterbury, New Zealand
- Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Ravi Gooneratne
- Faculty of Agriculture and Life Sciences, Department of Wine, Food, and Molecular Biosciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Xiang-Hong Ju
- College of Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, China
- Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| |
Collapse
|
255
|
Chen HT, Huang HL, Li YQ, Xu HM, Zhou YJ. Therapeutic advances in non-alcoholic fatty liver disease: A microbiota-centered view. World J Gastroenterol 2020; 26:1901-1911. [PMID: 32390701 PMCID: PMC7201149 DOI: 10.3748/wjg.v26.i16.1901] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/26/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent metabolic disorder with steadily increasing incidence rates worldwide, especially in the West. There are no drugs available at present to treat NAFLD, and the primary therapeutic options include weight loss and the combination of healthy diet and exercise. Therefore, novel interventions are required that can target the underlying risk factors. Gut microbiota is an "invisible organ" of the human body and vital for normal metabolism and immuno-modulation. The number and diversity of microbes differ across the gastrointestinal tract from the mouth to the anus, and is most abundant in the intestine. Since dysregulated gut microbiota is an underlying pathological factor of NAFLD, it is a viable therapeutic target that can be modulated by antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and microbial metabolites. In this review, we summarize the most recent advances in gut microbiota-targeted therapies against NAFLD in clinical and experimental studies, and critically evaluate novel targets and strategies for treating NAFLD.
Collapse
Affiliation(s)
- Hui-Ting Chen
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong Province, China
| | - Hong-Li Huang
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong Province, China
| | - Yong-Qiang Li
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong Province, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong Province, China
| | - Yong-Jian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, Guangdong Province, China
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong Province, China
| |
Collapse
|
256
|
Sfarti C, Ciobica A, Balmus IM, Ilie OD, Trifan A, Petrea O, Cojocariu C, Gîrleanu I, Sîngeap AM, Stanciu C. Systemic Oxidative Stress Markers in Cirrhotic Patients with Hepatic Encephalopathy: Possible Connections with Systemic Ammoniemia. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:196. [PMID: 32340177 PMCID: PMC7231105 DOI: 10.3390/medicina56040196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/08/2020] [Accepted: 04/18/2020] [Indexed: 02/05/2023]
Abstract
Background and objectives: Oxidative stress shows evidence of dysregulation in cirrhotic patients with hepatic encephalopathy (HE), although there are still controversies regarding the connections between oxidative stress and ammonia in these patients. The aim of this study was to evaluate the oxidative stress implication in overt HE pathogenesis of cirrhotic patients. Materials and Methods: We performed a prospective case-control study, which included 40 patients divided into two groups: group A consisted of 20 cirrhotic patients with HE and increased systemic ammoniemia, and group B consisted of 20 cirrhotic patients with HE and normal systemic ammoniemia. The control group consisted of 21 healthy subjects matched by age and sex. The activity of superoxide dismutase (SOD), glutathione peroxidase (GPx), malondialdehyde (MDA) levels (lipid peroxidation marker), and ammoniemia were evaluated. Results: We found a significant decrease in SOD and GPx activity and also a significant increase of MDA levels in cirrhotic patients with HE as compared to the healthy age-matched control group (1.35 ± 0.08 vs. 0.90 ± 0.08 U/mL, p = 0.002; 0.093 ± 0.06 vs. 0.006 ± 0.008 U/mL, p = 0.001; and 35.94 ± 1.37 vs. 68.90 ± 5.68 nmols/mL, p = 0.0001, respectively). Additionally, we found significant correlations between the main oxidative stress markers and the levels of systemic ammonia (r = 0.452, p = 0.005). Patients from group A had a significant increase of MDA as compared with those from group B (76.93 ± 5.48 vs. 50.06 ± 5.60 nmols/mL, p = 0.019). Also, there was a compensatory increase in the activity of both antioxidant enzymes (SOD and GPx) in patients with increased systemic ammoniemia (group A), as compared to HE patients from group B. Conclusions: Our results demonstrated a significant decrease in antioxidants enzymes activities (SOD and GPx), as well as a significant increase in MDA concentrations, adding new data regarding the influence of oxidative stress in HE pathogenesis in cirrhotic patients.
Collapse
Affiliation(s)
- Cătălin Sfarti
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, “St. Spiridon” University Hospital, Institute of Gastroenterology and Hepatology, Independence Avenue, no 1, 700111 Iași, Romania; (C.S.); (A.T.); (O.P.); (I.G.); (A.M.S.)
| | - Alin Ciobica
- Department of Research, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 11, 700505 Iasi, Romania; (I.-M.B.); (O.-D.I.)
| | - Ioana-Miruna Balmus
- Department of Research, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 11, 700505 Iasi, Romania; (I.-M.B.); (O.-D.I.)
- Department of Interdisciplinary Research in Science, Alexandru Ioan Cuza University of Iasi, Carol I Avenue, no. 11, 700506 Iasi, Romania
| | - Ovidiu-Dumitru Ilie
- Department of Research, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no 11, 700505 Iasi, Romania; (I.-M.B.); (O.-D.I.)
| | - Anca Trifan
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, “St. Spiridon” University Hospital, Institute of Gastroenterology and Hepatology, Independence Avenue, no 1, 700111 Iași, Romania; (C.S.); (A.T.); (O.P.); (I.G.); (A.M.S.)
| | - Oana Petrea
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, “St. Spiridon” University Hospital, Institute of Gastroenterology and Hepatology, Independence Avenue, no 1, 700111 Iași, Romania; (C.S.); (A.T.); (O.P.); (I.G.); (A.M.S.)
| | - Camelia Cojocariu
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, “St. Spiridon” University Hospital, Institute of Gastroenterology and Hepatology, Independence Avenue, no 1, 700111 Iași, Romania; (C.S.); (A.T.); (O.P.); (I.G.); (A.M.S.)
| | - Irina Gîrleanu
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, “St. Spiridon” University Hospital, Institute of Gastroenterology and Hepatology, Independence Avenue, no 1, 700111 Iași, Romania; (C.S.); (A.T.); (O.P.); (I.G.); (A.M.S.)
| | - Ana Maria Sîngeap
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, “St. Spiridon” University Hospital, Institute of Gastroenterology and Hepatology, Independence Avenue, no 1, 700111 Iași, Romania; (C.S.); (A.T.); (O.P.); (I.G.); (A.M.S.)
| | - Carol Stanciu
- Romanian Academy, Iasi Branch, Nr. 8, Carol I Avenue, no. 8, 700505 Iasi, Romania;
| |
Collapse
|
257
|
Krause JL, Schäpe SS, Schattenberg F, Müller S, Ackermann G, Rolle-Kampczyk UE, Jehmlich N, Pierzchalski A, von Bergen M, Herberth G. The Activation of Mucosal-Associated Invariant T (MAIT) Cells Is Affected by Microbial Diversity and Riboflavin Utilization in vitro. Front Microbiol 2020; 11:755. [PMID: 32390989 PMCID: PMC7189812 DOI: 10.3389/fmicb.2020.00755] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Recent research has demonstrated that MAIT cells are activated by individual bacterial or yeasts species that possess the riboflavin biosynthesis pathway. However, little is known about the MAIT cell activating potential of microbial communities and the contribution of individual community members. Here, we analyze the MAIT cell activating potential of a human intestinal model community (SIHUMIx) as well as intestinal microbiota after bioreactor cultivation. We determined the contribution of individual SIHUMIx community members to the MAIT cell activating potential and investigated whether microbial stress can influence their MAIT cell activating potential. The MAIT cell activating potential of SIHUMIx was directly related to the relative species abundances in the community. We therefore suggest an additive relationship between the species abundances and their MAIT cell activating potential. In diverse microbial communities, we found that a low MAIT cell activating potential was associated with high microbial diversity and a high level of riboflavin demand and vice versa. We suggest that microbial diversity might affect MAIT cell activation via riboflavin utilization within the community. Microbial acid stress significantly reduced the MAIT cell activating potential of SIHUMIx by impairing riboflavin availability through increasing the riboflavin demand. We show that MAIT cells can perceive microbial stress due to changes in riboflavin utilization and that riboflavin availability might also play a central role for the MAIT cell activating potential of diverse microbiota.
Collapse
Affiliation(s)
- Jannike L Krause
- Department of Environmental Immunology, Helmholtz-Centre for Environmental Research - UFZ Leipzig, Germany
| | - Stephanie S Schäpe
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Florian Schattenberg
- Department of Environmental Microbiology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Susann Müller
- Department of Environmental Microbiology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | | | - Ulrike E Rolle-Kampczyk
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Nico Jehmlich
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Arkadiusz Pierzchalski
- Department of Environmental Immunology, Helmholtz-Centre for Environmental Research - UFZ Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany.,Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Gunda Herberth
- Department of Environmental Immunology, Helmholtz-Centre for Environmental Research - UFZ Leipzig, Germany
| |
Collapse
|
258
|
Stathopoulou C, Nikoleri D, Bertsias G. Immunometabolism: an overview and therapeutic prospects in autoimmune diseases. Immunotherapy 2020; 11:813-829. [PMID: 31120393 DOI: 10.2217/imt-2019-0002] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metabolism is a critical immune regulator under physiologic and pathologic conditions. Culminating evidence has disentangled the contribution of distinct metabolic pathways, namely glucolysis, pentose phosphate, fatty acid oxidation, glutaminolysis, Krebs cycle and oxidative phosphorylation, in modulating innate and adaptive immune cells based on their activation/differentiation state. Metabolic aberrations and changes in the intracellular levels of specific metabolites are linked to the inflammatory phenotype of immune cells implicated in autoimmune disorders such as systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis and diabetes. Notably, targeting metabolism such as the mTOR by rapamycin, hexokinase by 2-deoxy-D-glucose, AMP-activated protein kinase by metformin, may be used to ameliorate autoimmune inflammation. Accordingly, research in immunometabolism is expected to offer novel opportunities for monitoring and treating immune-mediated diseases.
Collapse
Affiliation(s)
- Chrysoula Stathopoulou
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| | - Dimitra Nikoleri
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| | - George Bertsias
- Department of Rheumatology, Clinical Immunology & Allergy, University Hospital of Heraklion, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity & Inflammation, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, N. Plastira 100, 70013 Heraklion, Greece
| |
Collapse
|
259
|
Meng Q, Luo Z, Cao C, Sun S, Ma Q, Li Z, Shi B, Shan A. Weaning Alters Intestinal Gene Expression Involved in Nutrient Metabolism by Shaping Gut Microbiota in Pigs. Front Microbiol 2020; 11:694. [PMID: 32362884 PMCID: PMC7181064 DOI: 10.3389/fmicb.2020.00694] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022] Open
Abstract
Weaning transition usually impairs intestinal architecture and functions and results in gut-associated disorders in pigs. Understanding the changes in intestinal transcriptome and gut microbiota during weaning transition is important for elucidating the underlying mechanism of weaning stress. In the present study, we performed RNA-seq to determine the changes in intestinal transcriptome and 16S rRNA sequencing to measure the gut microbiota changes in the weaning transition. Transcriptome results indicated that weaning transition altered intestinal gene expression involved in nutrient transport and metabolism. Regarding fatty metabolism, fatty acid-binding protein 1 (FABP1), acyl-CoA dehydrogenase (ACADSB), and carnitine palmitoyltransferase 2 (CPT2) expression in the intestine was decreased by weaning. Genes related to bile acid metabolism were increased by weaning, including FABP6, farnesoid X receptor (FXR or NR1H4) and organic solute transporter-α (SLC51A). In addition, genes associated with oxidative stress were altered by weaning transition, including decreased catalase (CAT) and lactate dehydrogenase (LDHA) and increased glutathione peroxidase 2 (GPX2) and superoxide dismutase 3 (SOD3). Results of microbiota composition showed that the Firmicutes abundance and Firmicutes/Bacteroidetes ratio were increased and that the Proteobacteria abundance in the fecal microbiota was decreased by the weaning process; during the weaning transition, the Bacteroides and Fusobacterium abundances decreased markedly, and these bacteria nearly disappeared, while the Prevotella abundance showed a marked increase. Moreover, the levels of the microbial metabolites butyrate and acetate increased with changes in gut microbiota composition. In addition, predictive metagenome by PICRUSt analysis showed that the pathways related to D-glutamine and D-glutamate metabolism, citrate cycle (TCA cycle), peroxisome proliferators-activated receptor (PPAR) signaling, alpha-linolenic acid metabolism were decreased and the pathway related to retinol metabolism was increased in the gut microbiota of piglets during weaning transition. Our results showed that early weaning alters intestinal gene expression involved in nutrient metabolism, which may be due to the changes in microbiota composition.
Collapse
Affiliation(s)
- Qingwei Meng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Zhang Luo
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Chunyu Cao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Shishuai Sun
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Qingquan Ma
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Zhongyu Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
260
|
Guo H, Huang L, Hu S, Chen C, Huang X, Liu W, Wang S, Zhu Y, Zhao Y, Zhang D. Effects of Carbon/Nitrogen Ratio on Growth, Intestinal Microbiota and Metabolome of Shrimp ( Litopenaeus vannamei). Front Microbiol 2020; 11:652. [PMID: 32351483 PMCID: PMC7176362 DOI: 10.3389/fmicb.2020.00652] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/22/2020] [Indexed: 11/20/2022] Open
Abstract
Increasing the C/N ratio of input feed has been reported as a practical approach for improving water quality and enhancing shrimp growth through changing the bacterial community of rearing water. However, little is known about the effects of different C/N ratios of feed input on the intestinal microbiota and metabolome of shrimp. In the present study, the effects of three different C/N ratio levels (CN6, CN10, and CN15) maintained by adding sucrose on the growth, intestinal microbiota and metabolome of Litopenaeus vannamei, and bioflocs formation were investigated after 17 days of feeding. The results indicated that higher C/N ratio (10 and 15), especially CN15, of feed input significantly enhance the length and weight of shrimp individuals accompanied by a significant accumulation of bioflocs, compared to that of CN6. The increase of C/N ratio input decreased the α-diversity of the intestinal microbiota and changed the microbial community structure through increasing the relative abundance of Actinobacteria, Rhodobacteraceae (mainly consist of Roseobacter and Paracoccus groups), Alteromonadaceae, and inhibiting the growth of Cyanobacteria, certain Rhodobacteraceae, Mycoplasmataceae and Vibrio. The change of microbial community caused by increasing C/N ratio input was closely associated with various bioactive metabolites of flavonoids, benzenoids, prenol lipids, and indole derivatives, which are benefit for shrimp growth either as an antimicrobial agent or as a nutrient component. Overall, this study demonstrated that manipulating high C/N ratio of feed input helps to the growth of shrimp through increasing the relative abundance of potential beneficial bacteria and the accumulation of various bioactive metabolites to suppress the growth of detrimental bacteria.
Collapse
Affiliation(s)
- Haipeng Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Lei Huang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Songtao Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Chen Chen
- Zhejiang Mariculture Research Institute, Wenzhou, China
| | - Xiaolin Huang
- Zhejiang Mariculture Research Institute, Wenzhou, China
| | - Wei Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Sipeng Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Yueyue Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Yueji Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| | - Demin Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, China.,School of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
261
|
Knysh OV, Pogorila MS, Voyda YV. In vitro immunomodulatory effect of Bifidobacterium bifidum and Lactobacillus reuteri cell free extracts. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Recent studies have shown that alterations of the immune response in the gastrointestinal mucosa are key components of the mechanism of the probiotic action of beneficial bacteria. Most of the beneficial effects of probiotics are due to the action of their structural components and metabolites. Macrophages are first-line defense cells of the immune system, which not only participate in the detection, phagocytosis and destruction of harmful microorganisms, but also determine the nature of the subsequent immune response by presenting antigens to T-cells and initiating inflammation by releasing cytokines. We researched the effect of two types of cell-free extracts (CFEs) containing probiotic derivatives (structural components and metabolites of bacteria) Bifidobacterium bifidum 1 (BbCFE) and Lactobacillus reuteri DSM 17938 (LrCFE) on the activity of mouse peritoneal macrophages and on the ability of peripheral human blood mononuclear cells to produce cytokines. CFEs were obtained by culturing probiotics in their own disintegrates and then removing cells and cell debris by centrifugation and filtration. Peritoneal macrophages were isolated from mice. Some of them were infected in vitro by Salmonella thyphimurium. Uninfected and infected macrophages were incubated in culture medium containing (30% vol) or not containing CFEs at 37 °С in a microaerobic atmosphere (5% СО2) for 18 hours. After incubation, peritoneal macrophages were lysed. The obtained suspensions were centrifuged and supernatants were carefully collected. Macrophages activity was assessed by the nitrites level, superoxide dismutase (SOD), lactate dehydrogenase (LDH) activity and antiinflammatory cytokines levels in supernatants using spectrophotometric method. Peripheral mononuclear cells were isolated from the blood of healthy volunteers. The ability of peripheral mononuclear blood cells to produce antiinflammatory cytokines was evaluated after cell stimulation with lipopolysaccharide (LPS) and incubation with or without CFEs. Cytokine levels in supernatants were determined using enzyme-linked immunosorbent assay (ELISA). After infection with S. thyphimurium in macrophages, nitrite levels increased 5.5-fold, SOD activity 4.8-fold, and LDH 2-fold. Both studied CFEs exerted a similar effect on the macrophages’ activity. Addition of BbCFE to the incubation medium of infected macrophages resulted in a 4-fold decrease in nitrite levels, and the addition of LrCFE was accompanied by a decrease in nitrite levels to levels in intact cells. Under the influence of both CFEs, the activity of SOD and LDH was significantly reduced and did not differ significantly from the activity of these enzymes in intact cells. BbCFE and LrCFE did not have a significant effect on nitrite levels, SOD and LDH activity in intact macrophages. Under the influence of BbCFE, there was a 2-fold decrease in the production of TNF, a 2-fold increase in IL10 production, and a 30% increase in IL6 production by mononuclear cells. LrCFE caused a decrease in TNF production by 26.7% and IL6 by 36%, and IL10 by 1.9 times. Thus, the studied CFEs normalized the nitrite levels in peritoneal macrophages infected with S. thyphymurium and infection-induced activation of SOD and LDH enzymes. This demonstrates their ability to modulate oxidative processes in macrophages. In addition, under the influence of the investigated CFEs, there was a decrease in the production of pro-inflammatory cytokines (TNFα and IL-6) and increased production of anti-inflammatory cytokine (IL-10) by human peripheral mononuclear cells. The results of the study indicate the ability of CFEs by influencing the functions of innate immunity cells to restrict the inflammatory response and oxidative stress. Based on this, CFEs can be considered as promising agents for the treatment of inflammatory diseases.
Collapse
|
262
|
Dovrolis N, Kolios G, Spyrou GM, Maroulakou I. Computational profiling of the gut-brain axis: microflora dysbiosis insights to neurological disorders. Brief Bioinform 2020; 20:825-841. [PMID: 29186317 DOI: 10.1093/bib/bbx154] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/17/2017] [Indexed: 12/14/2022] Open
Abstract
Almost 2500 years after Hippocrates' observations on health and its direct association to the gastrointestinal tract, a paradigm shift has recently occurred, making the gut and its symbionts (bacteria, fungi, archaea and viruses) a point of convergence for studies. It is nowadays well established that the gut microflora's compositional diversity regulates via its genes (the microbiome) the host's health and provides preliminary insights into disease progression and regulation. The microbiome's involvement is evident in immunological and physiological studies that link changes in its biodiversity to its contributions to the host's phenotype but also in neurological investigations, substantiating the aptly named gut-brain axis. The definitive mechanisms of this last bidirectional interaction will be our main focus because it presents researchers with a new conundrum. In this review, we prospect current literature for computational analysis methodologies that accommodate the need for better understanding of the microbiome-gut-brain interactions and neurological disorder onset and progression, through cross-disciplinary systems biology applications. We will present bioinformatics tools used in exploring these synergies that help build and interpret microbial 16S ribosomal RNA data sets, produced by shotgun and high-throughput sequencing of healthy and neurological disorder samples stored in biological databases. These approaches provide alternative means for researchers to form hypotheses to their inquests faster, cheaper and swith precision. The goal of these studies relies on the integration of combined metagenomics and metabolomics assessments. An accurate characterization of the microbiome and its functionality can support new diagnostic, prognostic and therapeutic strategies for neurological disorders, customized for each individual host.
Collapse
|
263
|
Abstract
The intestinal tract harbors a diverse community of microbes that have co-evolved with the host immune system. Although many of these microbes execute functions that are critical for host physiology, the host immune system must control the microbial community so that the dynamics of this interdependent relationship is maintained. To facilitate host homeostasis, the immune system ensures that the microbial load is tolerated, but anatomically contained, while remaining reactive to microbial invasion. Although the microbiota is required for intestinal immune development, immune responses regulate the structure and composition of the intestinal microbiota by evolving unique immune adaptations that manage this high-bacterial load. The immune mechanisms work together to ensure that commensal bacteria rarely breach the intestinal barrier and that any that do invade should be killed rapidly to prevent penetration to systemic sites. The communication between microbiota and the immune system is mediated by the interaction of bacterial components with pattern recognition receptors expressed by intestinal epithelium and various antigen-presenting cells resulting in activation of both innate and adaptive immune responses. Interaction between the microbial community and host plays a crucial role in the mucosal homeostasis and health status of the host. In addition to providing a home to numerous microbial inhabitants, the intestinal tract is an active immunological organ, with more resident immune cells than anywhere else in the body, organized in lymphoid structures called Peyer's patches and isolated lymphoid follicles such as the cecal tonsils. Macrophages, dendritic cells, various subsets of T cells, B cells and the secretory immunoglobulin A (IgA) they produce, all contribute to the generation of a proper immune response to invading pathogens while keeping the resident microbial community in check without generating an overt inflammatory response to it. IgA-producing plasma cells, intraepithelial lymphocytes, and γδT cell receptor-expressing T cells are lymphocytes that are uniquely present in the mucosa. In addition, of the γδT cells in the intestinal lamina propria, there are significant numbers of IL-17-producing T cells and regulatory T cells. The accumulation and function of these mucosal leukocytes are regulated by the presence of intestinal microbiota, which regulate these immune cells and enhance the mucosal barrier function allowing the host to mount robust immune responses against invading pathogens, and simultaneously maintains immune homeostasis.
Collapse
Affiliation(s)
- Michael H Kogut
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX, 77845 USA.
| | - Annah Lee
- Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX, 77845 USA; Department of Poultry Science, Texas A&M University, College Station, TX, 77845 USA
| | - Elizabeth Santin
- Universidade Federal Do Paraná, Department of Veterinary Medicine, Curitiba, 80035-050 Brazil
| |
Collapse
|
264
|
Dimitri-Pinheiro S, Soares R, Barata P. The Microbiome of the Nose-Friend or Foe? ALLERGY & RHINOLOGY 2020; 11:2152656720911605. [PMID: 32206384 PMCID: PMC7074508 DOI: 10.1177/2152656720911605] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently, multiple studies regarding the human microbiota and its role on the development of disease have emerged. Current research suggests that the nasal cavity is a major reservoir for opportunistic pathogens, which can then spread to other sections of the respiratory tract and be involved in the development of conditions such as allergic rhinitis, chronic rhinosinusitis, asthma, pneumonia, and otitis media. However, our knowledge of how nasal microbiota changes originate nasopharyngeal and respiratory conditions is still incipient. Herein, we describe how the nasal microbiome in healthy individuals varies with age and explore the effect of nasal microbiota changes in a range of infectious and immunological conditions. We also describe the potential health benefits of human microbiota modulation through probiotic use, both in disease prevention and as adjuvant therapy. Current research suggests that patients with different chronic rhinosinusitis phenotypes possess distinct nasal microbiota profiles, which influence immune response and may be used in the future as biomarkers of disease progression. Probiotic intervention may also have a promising role in the prevention and adjunctive treatment of acute respiratory tract infections and allergic rhinitis, respectively. However, further studies are needed to define the role of probiotics in the chronic rhinosinusitis.
Collapse
Affiliation(s)
- Sofia Dimitri-Pinheiro
- Hospital Centre of Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.,Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Raquel Soares
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal.,I3S-Institute for Innovation and Health Research, University of Porto, Porto, Portugal
| | - Pedro Barata
- I3S-Institute for Innovation and Health Research, University of Porto, Porto, Portugal.,Faculty of Health Sciences, University of Fernando Pessoa, Porto, Portugal
| |
Collapse
|
265
|
Beyond Heat Stress: Intestinal Integrity Disruption and Mechanism-Based Intervention Strategies. Nutrients 2020; 12:nu12030734. [PMID: 32168808 PMCID: PMC7146479 DOI: 10.3390/nu12030734] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
The current climate changes have increased the prevalence and intensity of heat stress (HS) conditions. One of the initial consequences of HS is the impairment of the intestinal epithelial barrier integrity due to hyperthermia and hypoxia following blood repartition, which often results in a leaky gut followed by penetration and transfer of luminal antigens, endotoxins, and pathogenic bacteria. Under extreme conditions, HS may culminate in the onset of “heat stroke”, a potential lethal condition if remaining untreated. HS-induced alterations of the gastrointestinal epithelium, which is associated with a leaky gut, are due to cellular oxidative stress, disruption of intestinal integrity, and increased production of pro-inflammatory cytokines. This review summarizes the possible resilience mechanisms based on in vitro and in vivo data and the potential interventions with a group of nutritional supplements, which may increase the resilience to HS-induced intestinal integrity disruption and maintain intestinal homeostasis.
Collapse
|
266
|
Shah RM, McKenzie EJ, Rosin MT, Jadhav SR, Gondalia SV, Rosendale D, Beale DJ. An Integrated Multi-Disciplinary Perspectivefor Addressing Challenges of the Human Gut Microbiome. Metabolites 2020; 10:E94. [PMID: 32155792 PMCID: PMC7143645 DOI: 10.3390/metabo10030094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
Our understanding of the human gut microbiome has grown exponentially. Advances in genome sequencing technologies and metagenomics analysis have enabled researchers to study microbial communities and their potential function within the context of a range of human gut related diseases and disorders. However, up until recently, much of this research has focused on characterizing the gut microbiological community structure and understanding its potential through system wide (meta) genomic and transcriptomic-based studies. Thus far, the functional output of these microbiomes, in terms of protein and metabolite expression, and within the broader context of host-gut microbiome interactions, has been limited. Furthermore, these studies highlight our need to address the issues of individual variation, and of samples as proxies. Here we provide a perspective review of the recent literature that focuses on the challenges of exploring the human gut microbiome, with a strong focus on an integrated perspective applied to these themes. In doing so, we contextualize the experimental and technical challenges of undertaking such studies and provide a framework for capitalizing on the breadth of insight such approaches afford. An integrated perspective of the human gut microbiome and the linkages to human health will pave the way forward for delivering against the objectives of precision medicine, which is targeted to specific individuals and addresses the issues and mechanisms in situ.
Collapse
Affiliation(s)
- Rohan M. Shah
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia;
- Land and Water, Commonwealth Scientific and Industrial Research Organization (CSIRO), Dutton Park, QLD 4102, Australia
| | - Elizabeth J. McKenzie
- Liggins Institute, The University of Auckland, Grafton, Auckland 1142, New Zealand; (E.J.M.); (M.T.R.)
| | - Magda T. Rosin
- Liggins Institute, The University of Auckland, Grafton, Auckland 1142, New Zealand; (E.J.M.); (M.T.R.)
| | - Snehal R. Jadhav
- Centre for Advanced Sensory Science, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC 3125, Australia;
| | - Shakuntla V. Gondalia
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia;
| | | | - David J. Beale
- Land and Water, Commonwealth Scientific and Industrial Research Organization (CSIRO), Dutton Park, QLD 4102, Australia
| |
Collapse
|
267
|
Albillos A, de Gottardi A, Rescigno M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J Hepatol 2020; 72:558-577. [PMID: 31622696 DOI: 10.1016/j.jhep.2019.10.003] [Citation(s) in RCA: 1208] [Impact Index Per Article: 241.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/14/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023]
Abstract
The gut-liver axis refers to the bidirectional relationship between the gut and its microbiota, and the liver, resulting from the integration of signals generated by dietary, genetic and environmental factors. This reciprocal interaction is established by the portal vein which enables transport of gut-derived products directly to the liver, and the liver feedback route of bile and antibody secretion to the intestine. The intestinal mucosal and vascular barrier is the functional and anatomical structure that serves as a playground for the interactions between the gut and the liver, limiting the systemic dissemination of microbes and toxins while allowing nutrients to access the circulation and to reach the liver. The control of microbial communities is critical to maintaining homeostasis of the gut-liver axis, and as part of this bidirectional communication the liver shapes intestinal microbial communities. Alcohol disrupts the gut-liver axis at multiple interconnected levels, including the gut microbiome, mucus barrier, epithelial barrier and at the level of antimicrobial peptide production, which increases microbial exposure and the proinflammatory environment of the liver. Growing evidence indicates the pathogenetic role of microbe-derived metabolites, such as trimethylamine, secondary bile acids, short-chain fatty acids and ethanol, in the pathogenesis of non-alcoholic fatty liver disease. Cirrhosis by itself is associated with profound alterations in gut microbiota and damage at the different levels of defence of the intestinal barrier, including the epithelial, vascular and immune barriers. The relevance of the severe disturbance of the intestinal barrier in cirrhosis has been linked to translocation of live bacteria, bacterial infections and disease progression. The identification of the elements of the gut-liver axis primarily damaged in each chronic liver disease offers possibilities for intervention. Beyond antibiotics, upcoming therapies centred on the gut include new generations of probiotics, bacterial metabolites (postbiotics), faecal microbial transplantation, and carbon nanoparticles. FXR-agonists target both the gut and the liver and are currently being tested in different liver diseases. Finally, synthetic biotic medicines, phages that target specific bacteria or therapies that create physical barriers between the gut and the liver offer new therapeutic approaches.
Collapse
Affiliation(s)
- Agustín Albillos
- Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, IRYCIS, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| | - Andrea de Gottardi
- Hepatology, Inselspital and Department of Biomedical Research, University of Bern, Switzerland; Servizio di Gastroenterología e Epatologia, Ente Ospedaliero Cantonale, Università della Svizzera Italiana, Lugano, Switzerland
| | - María Rescigno
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele (Mi), Italy; Humanitas Clinical and Research Center, IRCCS, 20089 Rozzano (Mi), Italy
| |
Collapse
|
268
|
Ilie OD, Ciobica A, McKenna J, Doroftei B, Mavroudis I. Minireview on the Relations between Gut Microflora and Parkinson's Disease: Further Biochemical (Oxidative Stress), Inflammatory, and Neurological Particularities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4518023. [PMID: 32089768 PMCID: PMC7025076 DOI: 10.1155/2020/4518023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/20/2019] [Accepted: 01/04/2020] [Indexed: 02/07/2023]
Abstract
The aetiology of Parkinson's disease (PD) is a highly debated topic. Despite the progressive increase in the number of patients diagnosed with PD over the last couple of decades, the causes remain largely unknown. This report is aimed at highlighting the main features of the microbial communities which have been termed "the second brain" that may be a major participant in the etiopathophysiology of PD. It is possible that dysbiosis could be caused by an overactivity of proinflammatory cytokines which act on the gastrointestinal tract as well as infections. The majority of patients who are diagnosed with PD display gastrointestinal symptoms as one of the earliest features. In addition, an unbalanced cycle of oxidative stress caused by dysbacteriosis may have the effect of gradually promoting PD's specific phenotype. Thus, it seems that bacteria possess the ability to manipulate the brain by initiating specific responses, defining their capability to configure the human body, with oxidative stress playing a pivotal role in preventing infections but also in activating related signalling pathways.
Collapse
Affiliation(s)
- Ovidiu-Dumitru Ilie
- Department of Research, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no. 11, 700505 Iasi, Romania
| | - Alin Ciobica
- Department of Research, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, no. 11, 700505 Iasi, Romania
| | - Jack McKenna
- Leeds Teaching Hospitals NHS Trust, Great George St., Leeds LS1 3EX, UK
| | - Bogdan Doroftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, no. 16, 700115 Iasi, Romania
- Origyn Fertility Center, Palace Street, no. 3C, 700032 Iasi, Romania
| | - Ioannis Mavroudis
- Leeds Teaching Hospitals NHS Trust, Great George St., Leeds LS1 3EX, UK
- Laboratory of Neuropathology and Electron Microscopy, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| |
Collapse
|
269
|
Davoodi S, Foley E. Host-Microbe-Pathogen Interactions: A Review of Vibrio cholerae Pathogenesis in Drosophila. Front Immunol 2020; 10:3128. [PMID: 32038640 PMCID: PMC6993214 DOI: 10.3389/fimmu.2019.03128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
Most animals maintain mutually beneficial symbiotic relationships with their intestinal microbiota. Resident microbes in the gastrointestinal tract breakdown indigestible food, provide essential nutrients, and, act as a barrier against invading microbes, such as the enteric pathogen Vibrio cholerae. Over the last decades, our knowledge of V. cholerae pathogenesis, colonization, and transmission has increased tremendously. A number of animal models have been used to study how V. cholerae interacts with host-derived resources to support gastrointestinal colonization. Here, we review studies on host-microbe interactions and how infection with V. cholerae disrupts these interactions, with a focus on contributions from the Drosophila melanogaster model. We will discuss studies that highlight the connections between symbiont, host, and V. cholerae metabolism; crosstalk between V. cholerae and host microbes; and the impact of the host immune system on the lethality of V. cholerae infection. These studies suggest that V. cholerae modulates host immune-metabolic responses in the fly and improves Vibrio fitness through competition with intestinal microbes.
Collapse
Affiliation(s)
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
270
|
Guo K, Ruan G, Fan W, Fang L, Wang Q, Luo M, Yi T. The effect of nitrite and sulfide on the antioxidant capacity and microbial composition of the intestines of red swamp crayfish, Procambarus clarkii. FISH & SHELLFISH IMMUNOLOGY 2020; 96:290-296. [PMID: 31765791 DOI: 10.1016/j.fsi.2019.11.052] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 06/10/2023]
Abstract
Nitrite and sulfide are harmful pollutants in water ecosystems that negatively influence the survival and growth of crayfish. It is currently known that the intestine of crustaceans acts as a significant immune organ, serving as the front line of defense against diseases. In this study, we investigated how the oxidative damage parameters, antioxidant status and microbial composition of the intestine of Procambarus clarkii were influenced under acute nitrite (60 mg/L) and sulfide (18 mg/L) stress for 72 h. Compared with the control, after exposure to nitrite and sulfide stress, the production of reactive oxygen species, and the lipid peroxide and malondialdehyde contents increased in the intestines and were significantly higher after 72 h of exposure. The superoxide dismutase, catalase and glutathione peroxidase activities increased to maximum levels at 6, 24 and 12 h, respectively. These activities then decreased gradually and were significantly lower than those of the control after 48 or 72 h of exposure. In the crayfish exposed to stress, the expression of antioxidant genes including heat shock protein 70, ferritin and metallothionein increased to their maximum values at 12, 48 and 12 h, respectively. The expression levels then decreased gradually, and after 72 h, were lower than, or lacked significant differences with, the expression levels in the control. Additionally, nitrite and sulfide exposure restructured the intestinal microbial community of P. clarkii. This led to decreases in the abundance of some genera such as Citrobacter. However, the abundance of other genera, such as Shewanella and Acinetobacter, increased. Therefore, the health of P. clarkii was seriously impaired when exposed to nitrite and sulfide stress.
Collapse
Affiliation(s)
- Kun Guo
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, 434000, PR China
| | - Guoliang Ruan
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, 434000, PR China; Hubei Research Institute of Aquatic Industry Technology, Jingzhou, 434000, PR China.
| | - Wenhao Fan
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, 434000, PR China
| | - Liu Fang
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, 434000, PR China; Hubei Research Institute of Aquatic Industry Technology, Jingzhou, 434000, PR China
| | - Qian Wang
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, 434000, PR China
| | - Mingzhong Luo
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, 434000, PR China
| | - Tilin Yi
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, 434000, PR China; Hubei Research Institute of Aquatic Industry Technology, Jingzhou, 434000, PR China
| |
Collapse
|
271
|
Olsen I, Hicks SD. Oral microbiota and autism spectrum disorder (ASD). J Oral Microbiol 2019; 12:1702806. [PMID: 31893019 PMCID: PMC6913665 DOI: 10.1080/20002297.2019.1702806] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is associated with several oropharyngeal abnormalities, including dysbiosis in the oral microbiota. Since the oral cavity is the start of the gastrointestinal tract, this strengthens and extends the notion of a microbial gut-brain axis in ASD and even raises the question whether a microbial oral-brain axis exists. It is clear that oral bacteria can find their way to the brain through a number of pathways following routine dental procedures. A connection between the oral microbiota and a number of other brain disorders has been reported. As the evidence so far for an association between the oral microbiota and ASDs rests on a few reports only, further studies in this field are necessary. The current review discusses a possible relationship between oral bacteria and the biologic and symptomologic aspects of ASD, focusing on the clinical implications for diagnostic and therapeutic development.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Steven D. Hicks
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
272
|
Miller CJ, Bates ST, Gielda LM, Creighton JC. Examining transmission of gut bacteria to preserved carcass via anal secretions in Nicrophorus defodiens. PLoS One 2019; 14:e0225711. [PMID: 31790470 PMCID: PMC6886834 DOI: 10.1371/journal.pone.0225711] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 11/11/2019] [Indexed: 12/31/2022] Open
Abstract
Direct transmission of bacteria to subsequent generations highlights the beneficial nature of host-bacteria relationships. In insects, this process is often mediated by the production of microbe-containing secretions. The objective of this study was to determine if the burying beetle, Nicrophorus defodiens, utilizes anal secretions to transmit adult digestive tract bacteria onto a small vertebrate carcass; thus creating the potential to aid in carcass preservation or pass digestive tract bacteria to their larval offspring. Using high-throughput Illumina sequencing of the 16S rRNA gene, we characterized bacterial communities of adult beetle digestive tracts, their anal secretions, and prepared mouse carcasses. We also examined unprepared carcass bacterial communities as a means to interpret community shifts that take place during carcass preservation. We found a vast reduction in diversity on prepared carcasses after anal secretion application. Overall, there was little similarity in bacterial communities among adult digestive tracts, anal secretions, and prepared carcasses, suggesting bacterial communities found in adult digestive tracts do not successfully colonize and achieve dominance on prepared carcasses by way of beetle anal secretions. We concluded that N. defodiens does not transmit their digestive tract bacterial communities to prepared carcasses in a wholesale manner, but may transmit key microbes, including core microbiome members, to preserved carcasses that may ultimately act to sustain larvae and serve as inocula for larval digestive tracts.
Collapse
Affiliation(s)
- Christopher James Miller
- Department of Biological Sciences, Purdue University Northwest, Hammond, IN, United States of America
| | - Scott Thomas Bates
- Department of Biological Sciences, Purdue University Northwest, Westville, IN, United States of America
| | - Lindsay M. Gielda
- Department of Biological Sciences, Purdue University Northwest, Westville, IN, United States of America
| | - J. Curtis Creighton
- Department of Biological Sciences, Purdue University Northwest, Hammond, IN, United States of America
| |
Collapse
|
273
|
Abstract
Despite the development of new drugs and therapeutic strategies, mortality and morbidity related to heart failure (HF) remains high. It is also the leading cause of global mortality. Several concepts have been proposed to explore the underlying pathogenesis of HF, but there is still a strong need for more specific and complementary therapeutic options. In recent years, accumulating evidence has demonstrated that changes in the composition of gut microbiota, referred to as dysbiosis, might play a pivotal role in the development of several diseases, including HF. HF-associated decreased cardiac output, resulting in bowell wall oedema and intestine ischaemia, can alter gut structure, peamibility and function. These changes would favour bacterial translocation, exacerbating HF pathogenesis at least partly through activation of systemic inflammation. Although our knowledge of the precise molecular mechanisms by which gut dysbiosis influance HF is still limited, a growing body of evidence has recently demonstrated the impact of a series of gut microbiome-derived metabolites, such as trimetylamine N-oxide, short-chain fatty acids or secondary bile acids, which have been shown to play critical roles in cardiac health and disease. This review will summarize the role of gut microbiota and its metabolites in the pathogenesis of HF. Current and future preventive and therapeutic strategies to prevent HF by an adequate modulation of the microbiome and its derived metabolites are also discussed.
Collapse
Affiliation(s)
- Maxime Branchereau
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse, France
| | - Rémy Burcelin
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse, France
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse, France.
- INSERM U1048 - Institute of Cardiovascular and Metabolic Diseases - I2MC, 1 avenue Jean Poulhès - BP 84225, 31432, Toulouse Cedex 4, France.
| |
Collapse
|
274
|
Intestinal microbiome composition and its relation to joint pain and inflammation. Nat Commun 2019; 10:4881. [PMID: 31653850 PMCID: PMC6814863 DOI: 10.1038/s41467-019-12873-4] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Macrophage-mediated inflammation is thought to have a causal role in osteoarthritis-related pain and severity, and has been suggested to be triggered by endotoxins produced by the gastrointestinal microbiome. Here we investigate the relationship between joint pain and the gastrointestinal microbiome composition, and osteoarthritis-related knee pain in the Rotterdam Study; a large population based cohort study. We show that abundance of Streptococcus species is associated with increased knee pain, which we validate by absolute quantification of Streptococcus species. In addition, we replicate these results in 867 Caucasian adults of the Lifelines-DEEP study. Finally we show evidence that this association is driven by local inflammation in the knee joint. Our results indicate the microbiome is a possible therapeutic target for osteoarthritis-related knee pain.
Collapse
|
275
|
Negi S, Das DK, Pahari S, Nadeem S, Agrewala JN. Potential Role of Gut Microbiota in Induction and Regulation of Innate Immune Memory. Front Immunol 2019; 10:2441. [PMID: 31749793 PMCID: PMC6842962 DOI: 10.3389/fimmu.2019.02441] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/01/2019] [Indexed: 12/14/2022] Open
Abstract
The gut microbiota significantly regulates the development and function of the innate and adaptive immune system. The attribute of immunological memory has long been linked only with adaptive immunity. Recent evidence indicates that memory is also present in the innate immune cells such as monocytes/macrophages and natural killer cells. These cells exhibit pattern recognition receptors (PRRs) that recognize microbe- or pathogen-associated molecular patterns (MAMPs or PAMPs) expressed by the microbes. Interaction between PRRs and MAMPs is quite crucial since it triggers the sequence of signaling events and epigenetic rewiring that not only play a cardinal role in modulating the activation and function of the innate cells but also impart a sense of memory response. We discuss here how gut microbiota can influence the generation of innate memory and functional reprogramming of bone marrow progenitors that helps in protection against infections. This article will broaden our current perspective of association between the gut microbiome and innate memory. In the future, this knowledge may pave avenues for development and designing of novel immunotherapies and vaccination strategies.
Collapse
Affiliation(s)
- Shikha Negi
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Gastroenterology Division, Washington University in St. Louis, St. Louis, MO, United States
| | - Deepjyoti Kumar Das
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Susanta Pahari
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Immunology Division, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Sajid Nadeem
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Center for Biomedical Engineering, Indian Institute of Technology-Ropar, Rupnagar, India
| |
Collapse
|
276
|
Smith RP, Easson C, Lyle SM, Kapoor R, Donnelly CP, Davidson EJ, Parikh E, Lopez JV, Tartar JL. Gut microbiome diversity is associated with sleep physiology in humans. PLoS One 2019; 14:e0222394. [PMID: 31589627 PMCID: PMC6779243 DOI: 10.1371/journal.pone.0222394] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022] Open
Abstract
The human gut microbiome can influence health through the brain-gut-microbiome axis. Growing evidence suggests that the gut microbiome can influence sleep quality. Previous studies that have examined sleep deprivation and the human gut microbiome have yielded conflicting results. A recent study found that sleep deprivation leads to changes in gut microbiome composition while a different study found that sleep deprivation does not lead to changes in gut microbiome. Accordingly, the relationship between sleep physiology and the gut microbiome remains unclear. To address this uncertainty, we used actigraphy to quantify sleep measures coupled with gut microbiome sampling to determine how the gut microbiome correlates with various measures of sleep physiology. We measured immune system biomarkers and carried out a neurobehavioral assessment as these variables might modify the relationship between sleep and gut microbiome composition. We found that total microbiome diversity was positively correlated with increased sleep efficiency and total sleep time, and was negatively correlated with wake after sleep onset. We found positive correlations between total microbiome diversity and interleukin-6, a cytokine previously noted for its effects on sleep. Analysis of microbiome composition revealed that within phyla richness of Bacteroidetes and Firmicutes were positively correlated with sleep efficiency, interleukin-6 concentrations and abstract thinking. Finally, we found that several taxa (Lachnospiraceae, Corynebacterium, and Blautia) were negatively correlated with sleep measures. Our findings initiate linkages between gut microbiome composition, sleep physiology, the immune system and cognition. They may lead to mechanisms to improve sleep through the manipulation of the gut microbiome.
Collapse
Affiliation(s)
- Robert P. Smith
- Department of Biological Sciences, Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Fort Lauderdale FL, United States of America
| | - Cole Easson
- Department of Biological Sciences, Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Fort Lauderdale FL, United States of America
- Biology Department, Middle Tennessee State University, Murfreesboro, TN, United States of America
| | - Sarah M. Lyle
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| | - Ritishka Kapoor
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| | - Chase P. Donnelly
- Department of Biological Sciences, Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Fort Lauderdale FL, United States of America
| | - Eileen J. Davidson
- Department of Biological Sciences, Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Fort Lauderdale FL, United States of America
| | - Esha Parikh
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| | - Jose V. Lopez
- Department of Biological Sciences, Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Fort Lauderdale FL, United States of America
| | - Jaime L. Tartar
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
- * E-mail:
| |
Collapse
|
277
|
Wang G, Huang S, Wang Y, Cai S, Yu H, Liu H, Zeng X, Zhang G, Qiao S. Bridging intestinal immunity and gut microbiota by metabolites. Cell Mol Life Sci 2019; 76:3917-3937. [PMID: 31250035 PMCID: PMC6785585 DOI: 10.1007/s00018-019-03190-6] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/06/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023]
Abstract
The gastrointestinal tract is the site of nutrient digestion and absorption and is also colonized by diverse, highly mutualistic microbes. The intestinal microbiota has diverse effects on the development and function of the gut-specific immune system, and provides some protection from infectious pathogens. However, interactions between intestinal immunity and microorganisms are very complex, and recent studies have revealed that this intimate crosstalk may depend on the production and sensing abilities of multiple bioactive small molecule metabolites originating from direct produced by the gut microbiota or by the metabolism of dietary components. Here, we review the interplay between the host immune system and the microbiota, how commensal bacteria regulate the production of metabolites, and how these microbiota-derived products influence the function of several major innate and adaptive immune cells involved in modulating host immune homeostasis.
Collapse
Affiliation(s)
- Gang Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China
| | - Shuo Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China
| | - Yuming Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China
| | - Shuang Cai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China
| | - Hongbing Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK, 74074, USA
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
- Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
278
|
Hagan T, Cortese M, Rouphael N, Boudreau C, Linde C, Maddur MS, Das J, Wang H, Guthmiller J, Zheng NY, Huang M, Uphadhyay AA, Gardinassi L, Petitdemange C, McCullough MP, Johnson SJ, Gill K, Cervasi B, Zou J, Bretin A, Hahn M, Gewirtz AT, Bosinger SE, Wilson PC, Li S, Alter G, Khurana S, Golding H, Pulendran B. Antibiotics-Driven Gut Microbiome Perturbation Alters Immunity to Vaccines in Humans. Cell 2019; 178:1313-1328.e13. [PMID: 31491384 PMCID: PMC6750738 DOI: 10.1016/j.cell.2019.08.010] [Citation(s) in RCA: 417] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 06/21/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022]
Abstract
Emerging evidence indicates a central role for the microbiome in immunity. However, causal evidence in humans is sparse. Here, we administered broad-spectrum antibiotics to healthy adults prior and subsequent to seasonal influenza vaccination. Despite a 10,000-fold reduction in gut bacterial load and long-lasting diminution in bacterial diversity, antibody responses were not significantly affected. However, in a second trial of subjects with low pre-existing antibody titers, there was significant impairment in H1N1-specific neutralization and binding IgG1 and IgA responses. In addition, in both studies antibiotics treatment resulted in (1) enhanced inflammatory signatures (including AP-1/NR4A expression), observed previously in the elderly, and increased dendritic cell activation; (2) divergent metabolic trajectories, with a 1,000-fold reduction in serum secondary bile acids, which was highly correlated with AP-1/NR4A signaling and inflammasome activation. Multi-omics integration revealed significant associations between bacterial species and metabolic phenotypes, highlighting a key role for the microbiome in modulating human immunity.
Collapse
Affiliation(s)
- Thomas Hagan
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mario Cortese
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Decatur, GA 30030, USA
| | - Carolyn Boudreau
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Caitlin Linde
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Mohan S Maddur
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Jishnu Das
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Hong Wang
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Jenna Guthmiller
- Department of Medicine, Section of Rheumatology, Knapp Center for Lupus and Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Nai-Ying Zheng
- Department of Medicine, Section of Rheumatology, Knapp Center for Lupus and Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Min Huang
- Department of Medicine, Section of Rheumatology, Knapp Center for Lupus and Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Amit A Uphadhyay
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Luiz Gardinassi
- Department of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Caroline Petitdemange
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | | | - Sara Jo Johnson
- Hope Clinic of the Emory Vaccine Center, Decatur, GA 30030, USA
| | - Kiran Gill
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Barbara Cervasi
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Jun Zou
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Alexis Bretin
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Megan Hahn
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Steve E Bosinger
- Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Patrick C Wilson
- Department of Medicine, Section of Rheumatology, Knapp Center for Lupus and Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Shuzhao Li
- Department of Medicine, Emory University, Atlanta, GA 30303, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
279
|
Zhang Y, Zhao R, Shi D, Sun S, Ren H, Zhao H, Wu W, Jin L, Sheng J, Shi Y. Characterization of the circulating microbiome in acute-on-chronic liver failure associated with hepatitis B. Liver Int 2019; 39:1207-1216. [PMID: 30864226 DOI: 10.1111/liv.14097] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with hepatitis B-related acute-on-chronic liver failure (HB-ACLF) may have an increased circulating microbial burden. This study aimed to assess circulating microbial load and composition and to explore the association between the circulating microbiome and both systemic inflammation (SI) and clinical outcome in HB-ACLF. METHODS Plasma from 50 HB-ACLF patients, 23 healthy controls and 25 patients with compensated liver cirrhosis (C-LC) was analysed for chemokines/cytokines and bacterial DNA and further analysed by 16S rDNApyrosequencing. Linear discriminant analysis effect size (LEfSe) and inferred metagenomics analyses were performed. RESULTS The circulating bacterial DNA was significantly increased in HB-ACLF patients compared to that in the control groups. The overall microbial diversity was significantly decreased in HB-ACLF patients. HB-ACLF patients were enriched with Moraxellaceae, Sulfurovum, Comamonas and Burkholderiaceae but were depleted in Actinobacteria, Deinococcus-Thermus, Alphaproteobacteria, Xanthomonadaceae and Enterobacteriaceae compared to controls. Network analysis revealed a direct positive correlation between Burkholderiaceae and chemokine IP-10 in HB-ACLF patients. The relative abundance of Prevotellaceae independently predicted 28-day mortality. Inferred functional metagenomics predicted an enrichment of bacteria with genes related to methane, alanine, aspartate, glutamate, pyrimidine, purine and energy metabolism. CONCLUSIONS HB-ACLF patients display increased circulating microbial burden, altered microbiome composition and a shift in microbiome functionality. The alteration in circulating microbiota is associated with SI and clinical outcome in HB-ACLF.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, Hangzhou, China
| | - Ruihong Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shanshan Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haotang Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linfeng Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
280
|
A long-distance relationship: the commensal gut microbiota and systemic viruses. Curr Opin Virol 2019; 37:44-51. [PMID: 31226645 DOI: 10.1016/j.coviro.2019.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/14/2022]
Abstract
Recent advances defining the role of the commensal gut microbiota in the development, education, induction, function, and maintenance of the mammalian immune system inform our understanding of how immune responses govern the outcome of systemic virus infection. While characterization of the impact of the local oral, respiratory, dermal and genitourinary microbiota on host immune responses and systemic virus infection is in its infancy, the gut microbiota interacts with host immunity systemically and at distal non-gastrointestinal tract sites to modulate the pathogenesis of systemic viruses. Gut microbes, microbe-associated molecular patterns, and microbe-derived metabolites engage receptors expressed on the cell surface, in the endosome, or in the cytoplasm to orchestrate optimal innate and adaptive immune responses important for controlling systemic virus infection.
Collapse
|
281
|
Guthrie L, Wolfson S, Kelly L. The human gut chemical landscape predicts microbe-mediated biotransformation of foods and drugs. eLife 2019; 8:42866. [PMID: 31184303 PMCID: PMC6559788 DOI: 10.7554/elife.42866] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 05/10/2019] [Indexed: 12/14/2022] Open
Abstract
Microbes are nature's chemists, capable of producing and metabolizing a diverse array of compounds. In the human gut, microbial biochemistry can be beneficial, for example vitamin production and complex carbohydrate breakdown; or detrimental, such as the reactivation of an inactive drug metabolite leading to patient toxicity. Identifying clinically relevant microbiome metabolism requires linking microbial biochemistry and ecology with patient outcomes. Here we present MicrobeFDT, a resource which clusters chemically similar drug and food compounds and links these compounds to microbial enzymes and known toxicities. We demonstrate that compound structural similarity can serve as a proxy for toxicity, enzyme sharing, and coarse-grained functional similarity. MicrobeFDT allows users to flexibly interrogate microbial metabolism, compounds of interest, and toxicity profiles to generate novel hypotheses of microbe-diet-drug-phenotype interactions that influence patient outcomes. We validate one such hypothesis experimentally, using MicrobeFDT to reveal unrecognized gut microbiome metabolism of the ovarian cancer drug altretamine.
Collapse
Affiliation(s)
- Leah Guthrie
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York, United States
| | - Sarah Wolfson
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York, United States
| | - Libusha Kelly
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York, United States.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, United States
| |
Collapse
|
282
|
Abstract
Over the last decade, our understanding of the composition and functions of the gut microbiota has greatly increased. To a large extent, this has been due to the development of high-throughput genomic analyses of microbial communities, which have identified the critical contributions of the microbiome to human health. Over the last decade, our understanding of the composition and functions of the gut microbiota has greatly increased. To a large extent, this has been due to the development of high-throughput genomic analyses of microbial communities, which have identified the critical contributions of the microbiome to human health. Consequently, the intestinal microbiota has emerged as an attractive therapeutic target. The large majority of microbiota-targeted therapies aim at engineering the intestinal ecosystem by means of probiotics or prebiotics. Recently, a novel therapeutic approach has emerged which focuses on molecules that are secreted, modulated, or degraded by the microbiome and act directly on the host. Here, we discuss the advantages and challenges associated with the metabolite-based “postbiotic” approach, highlighting recent progress and the areas that need intensive attention and investigation over the next 5 years. The time is ripe for postbiotic therapies to be developed in the near future.
Collapse
|
283
|
Wondraczek L, Pohnert G, Schacher FH, Köhler A, Gottschaldt M, Schubert US, Küsel K, Brakhage AA. Artificial Microbial Arenas: Materials for Observing and Manipulating Microbial Consortia. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1900284. [PMID: 30993782 DOI: 10.1002/adma.201900284] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/28/2019] [Indexed: 06/09/2023]
Abstract
From the smallest ecological niche to global scale, communities of microbial life present a major factor in system regulation and stability. As long as laboratory studies remain restricted to single or few species assemblies, however, very little is known about the interaction patterns and exogenous factors controlling the dynamics of natural microbial communities. In combination with microfluidic technologies, progress in the manufacture of functional and stimuli-responsive materials makes artificial microbial arenas accessible. As habitats for natural or multispecies synthetic consortia, they are expected to not only enable detailed investigations, but also the training and the directed evolution of microbial communities in states of balance and disturbance, or under the effects of modulated stimuli and spontaneous response triggers. Here, a perspective on how materials research will play an essential role in generating answers to the most pertinent questions of microbial engineering is presented, and the concept of adaptive microbial arenas and possibilities for their construction from particulate microniches to 3D habitats is introduced. Materials as active and tunable components at the interface of living and nonliving matter offer exciting opportunities in this field. Beyond forming the physical horizon for microbial cultivates, they will enable dedicated intervention, training, and observation of microbial consortia.
Collapse
Affiliation(s)
- Lothar Wondraczek
- Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Fraunhoferstrasse 6, 07743, Jena, Germany
- Center of Energy and Environmental Chemistry Jena (CEEC Jena), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Microverse Cluster, Friedrich Schiller University Jena, Neugasse 23, 07743, Jena, Germany
| | - Georg Pohnert
- Microverse Cluster, Friedrich Schiller University Jena, Neugasse 23, 07743, Jena, Germany
- Institute of Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Lessingstrasse 8, 07743, Jena, Germany
- Max Planck Institute for Chemical Ecology, Hans-Knöll-Strasse 8, 07745, Jena, Germany
| | - Felix H Schacher
- Center of Energy and Environmental Chemistry Jena (CEEC Jena), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Microverse Cluster, Friedrich Schiller University Jena, Neugasse 23, 07743, Jena, Germany
- Institute of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Angela Köhler
- Microverse Cluster, Friedrich Schiller University Jena, Neugasse 23, 07743, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology (HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| | - Michael Gottschaldt
- Institute of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Ulrich S Schubert
- Center of Energy and Environmental Chemistry Jena (CEEC Jena), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Microverse Cluster, Friedrich Schiller University Jena, Neugasse 23, 07743, Jena, Germany
- Institute of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Kirsten Küsel
- Microverse Cluster, Friedrich Schiller University Jena, Neugasse 23, 07743, Jena, Germany
- Institute of Biodiversity, Aquatic Geomicrobiology, Friedrich Schiller University, Dornburger Str. 159, 07743, Jena, Germany
- German Center for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Deutscher Platz 5E, 04103, Leipzig, Germany
| | - Axel A Brakhage
- Microverse Cluster, Friedrich Schiller University Jena, Neugasse 23, 07743, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology (HKI), Adolf-Reichwein-Str. 23, 07745, Jena, Germany
| |
Collapse
|
284
|
A tryptophan metabolite of the skin microbiota attenuates inflammation in patients with atopic dermatitis through the aryl hydrocarbon receptor. J Allergy Clin Immunol 2019; 143:2108-2119.e12. [DOI: 10.1016/j.jaci.2018.11.036] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/13/2018] [Accepted: 11/09/2018] [Indexed: 12/20/2022]
|
285
|
Zhang Y, Chen H, Zhu W, Yu K. Cecal Infusion of Sodium Propionate Promotes Intestinal Development and Jejunal Barrier Function in Growing Pigs. Animals (Basel) 2019; 9:ani9060284. [PMID: 31141995 PMCID: PMC6617143 DOI: 10.3390/ani9060284] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Microbial-derived short-chain fatty acids can exert influence on intestinal development and intestinal barrier function. Usually, it is well known that short-chain fatty acid butyrate provides energy for the colonic cell turnover and maintains the integrity of the colonic epithelium. However, the effect of short-chain fatty acid propionate on intestinal development and jejunal barrier function is given less attention. In this study, we found that cecal infusion of propionate promoted development of the jejunum and colon, and selectively enhanced jejunal tight junction protein expression. These results suggest that propionate by microbial fermentation in the hindgut has an important role in intestinal development and gut health. Abstract Short-chain fatty acids (SCFAs) produced by microbial fermentation facilitate the differentiation and proliferation of intestinal epithelium. However, the role of individual SCFAs, such as propionate, on intestinal development is still unclear. In the present study, sixteen barrows fitted with a cecal fistula were randomly divided into two groups for cecal infusion of either saline (control group) or sodium propionate (propionate group). After 28 days, the length and the relative weight of intestinal segments were calculated, the intestinal morphology was assessed, and the expression of tight junction protein was measured using qPCR and Western blotting. Compared to the saline group, the length of the colon was significantly increased in the propionate group (p < 0.05). The jejunal villi length and villi/crypt ratio in the propionate group were significantly higher than in the saline group (p < 0.05). Furthermore, propionate infusion significantly upregulated the mRNA levels of Claudin-4 and the expression of Claudin-1, Claudin-4, and Occludin protein in the jejunal mucosa (p < 0.05). Collectively, these findings revealed that the short-chain fatty acid propionate in the hindgut contributed to intestinal development, and selectively enhanced jejunal tight junction protein expression.
Collapse
Affiliation(s)
- Yanan Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China.
| | - Huizi Chen
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China.
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China.
- National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China.
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China.
- National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
286
|
Tang ZZ, Chen G, Hong Q, Huang S, Smith HM, Shah RD, Scholz M, Ferguson JF. Multi-Omic Analysis of the Microbiome and Metabolome in Healthy Subjects Reveals Microbiome-Dependent Relationships Between Diet and Metabolites. Front Genet 2019; 10:454. [PMID: 31164901 PMCID: PMC6534069 DOI: 10.3389/fgene.2019.00454] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022] Open
Abstract
The human microbiome has been associated with health status, and risk of disease development. While the etiology of microbiome-mediated disease remains to be fully elucidated, one mechanism may be through microbial metabolism. Metabolites produced by commensal organisms, including in response to host diet, may affect host metabolic processes, with potentially protective or pathogenic consequences. We conducted multi-omic phenotyping of healthy subjects (N = 136), in order to investigate the interaction between diet, the microbiome, and the metabolome in a cross-sectional sample. We analyzed the nutrient composition of self-reported diet (3-day food records and food frequency questionnaires). We profiled the gut and oral microbiome (16S rRNA) from stool and saliva, and applied metabolomic profiling to plasma and stool samples in a subset of individuals (N = 75). We analyzed these multi-omic data to investigate the relationship between diet, the microbiome, and the gut and circulating metabolome. On a global level, we observed significant relationships, particularly between long-term diet, the gut microbiome and the metabolome. Intake of plant-derived nutrients as well as consumption of artificial sweeteners were associated with significant differences in circulating metabolites, particularly bile acids, which were dependent on gut enterotype, indicating that microbiome composition mediates the effect of diet on host physiology. Our analysis identifies dietary compounds and phytochemicals that may modulate bacterial abundance within the gut and interact with microbiome composition to alter host metabolism.
Collapse
Affiliation(s)
- Zheng-Zheng Tang
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, Madison, WI, United States
| | - Guanhua Chen
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, WI, United States
| | - Qilin Hong
- Department of Statistics, University of Wisconsin–Madison, Madison, WI, United States
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Holly M. Smith
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rachana D. Shah
- Division of Pediatric Endocrinology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Matthew Scholz
- Vanderbilt Technologies for Advanced Genomics (VANTAGE), Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Translational and Clinical Cardiovascular Research Center (VTRACC), Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
287
|
Chen G, Chen R, Chen D, Ye H, Hu B, Zeng X, Liu Z. Tea Polysaccharides as Potential Therapeutic Options for Metabolic Diseases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:5350-5360. [PMID: 30474370 DOI: 10.1021/acs.jafc.8b05338] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Tea polysaccharides (TPS) are regarded as some of the main bioactive constituents of tea made from the leaves and buds of the tea plant ( Camellia sinensis L.). An increasing number of studies have demonstrated that TPS can reduce the risk of type 2 diabetes, obesity, and other metabolic diseases. However, the potential mechanisms responsible for antidiabetic and antiobesogenic activities of TPS remain unclear. Therefore, the cellular and physiological mechanisms that underlie the antidiabetic and antiobesogenic effects, including antioxidant and anti-inflammation effects, inhibition of digestive enzymes, prevention of macronutrient absorption, and expression of gene and protein, were summarized in this review. Furthermore, the gastrointestinal functions of TPS and the role of gut microbiota in the prevention and treatment of metabolic diseases were discussed. It is expected that the present review will be helpful for enhancing our knowledge about the health-promoting effects of TPS on metabolic diseases and stimulating further works on TPS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhonghua Liu
- Key Laboratory of Ministry of Education for Tea Science , Hunan Agricultural University , Changsha , Hunan 410128 , People's Republic of China
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients , Changsha , Hunan 410128 , People's Republic of China
| |
Collapse
|
288
|
Duan Y, Wang Y, Liu Q, Zhang J, Xiong D. Changes in the intestine barrier function of Litopenaeus vannamei in response to pH stress. FISH & SHELLFISH IMMUNOLOGY 2019; 88:142-149. [PMID: 30807860 DOI: 10.1016/j.fsi.2019.02.047] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 06/09/2023]
Abstract
pH of water environment affects the survival of aquatic animals. Intestine barrier function influences the health of animals, which is related to its mucosa structure, immune components, and microbial communities. In this study, we investigated the histological structure, digestive and metabolic capacity, immune responses, and microbial composition in the intestine of Litopenaeus vanmei under three different conditions: control (pH 8.3), low pH stress (pH 6.9), and high pH stress (pH 9.7) for 72 h. The results showed both low and high pH stress disrupted the intestine morphological structure, and induced variations in the activities of digestive (AMS, LPS, Tryp, and Pep) and metabolic (HK, PK, CCO, and LDH) enzymes. Low and high pH stress also increased oxidative stress (MDA, LPO, PC, and ·O2- generation), and decreased the antioxidant enzyme activities (T-AOC, SOD, and GST); shrimp enhanced CAT activity and HSP70, Trx, MT and Fer gene transcripts as defense mechanism. Additionally, Immune confusion was also found in the shrimp intestine in response to low and high pH stress, including the antibacterial ability (T-NOS, PO, proPO, ALF, and Lys), pathogen recognition (TLR and Lec), apoptosis (Casp, IAP and p53), and mucus homeostasis (Muc-1, Muc-2, Muc-5AC, Muc-5B, and Muc-19). pH exposure also decreased the diversity of the intestine bacterial, disturbed the composition of microbiota, and decreased the microbial metabolite SCFA contents. Our results indicated that acute pH stress can impair the intestine barrier function of white shrimp, probably via destroying mucosa structure, confusing digestion and metabolism, inducing oxidative stress, disordering immunity, and disrupting the microbial composition.
Collapse
Affiliation(s)
- Yafei Duan
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, PR China
| | - Yun Wang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, PR China
| | - Qingsong Liu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, PR China
| | - Jiasong Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, PR China.
| | - Dalin Xiong
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, PR China
| |
Collapse
|
289
|
Dovrolis N, Filidou E, Kolios G. Systems biology in inflammatory bowel diseases: on the way to precision medicine. Ann Gastroenterol 2019; 32:233-246. [PMID: 31040620 PMCID: PMC6479645 DOI: 10.20524/aog.2019.0373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic and recurrent inflammatory disorders of the gastrointestinal tract. The elucidation of their etiopathology requires complex and multiple approaches. Systems biology has come to fulfill this need in approaching the pathogenetic mechanisms of IBD and its etiopathology, in a comprehensive way, by combining data from different scientific sources. In combination with bioinformatics and network medicine, it uses principles from computer science, mathematics, physics, chemistry, biology, medicine and computational tools to achieve its purposes. Systems biology utilizes scientific sources that provide data from omics studies (e.g., genomics, transcriptomics, etc.) and clinical observations, whose combined analysis leads to network formation and ultimately to a more integrative image of disease etiopathogenesis. In this review, we analyze the current literature on the methods and the tools utilized by systems biology in order to cover an innovative and exciting field: IBD-omics.
Collapse
Affiliation(s)
- Nikolas Dovrolis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Correspondence to: Prof. George Kolios, MD PhD, Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Dragana, Alexandroupolis, 68100, Greece, e-mail:
| |
Collapse
|
290
|
Emergence of Extended-Spectrum Beta-Lactamase-Producing Enterobacteriaceae Colonization: What Termites Can Teach Us About the Gut Microbiota. Crit Care Med 2019; 45:752-754. [PMID: 28291103 DOI: 10.1097/ccm.0000000000002279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
291
|
Duan Y, Wang Y, Liu Q, Dong H, Li H, Xiong D, Zhang J. Changes in the intestine microbial, digestion and immunity of Litopenaeus vannamei in response to dietary resistant starch. Sci Rep 2019; 9:6464. [PMID: 31015554 PMCID: PMC6478684 DOI: 10.1038/s41598-019-42939-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/11/2019] [Indexed: 01/07/2023] Open
Abstract
Resistant starch (RS) is a constituent of dietary fibre that has beneficial effects on the intestine physiological function of animals. However, the roles of RS on shrimp intestine health is unknown. In this study, we investigated the the effects of dietary RS on the microbial composition, and digestive and immune-related indices in the intestine of Litopenaeus vannamei. The shrimp were fed with diets containing different levels of RS: 0 g/kg (Control), 10 g/kg (RS1), 30 g/kg (RS2) and 50 g/kg (RS3) for 56 days. The results showed that dietary RS improved the morphology of the intestine mucosa. RS also increased the activity of digestive enzymes (AMS, LPS, Tryp, and Pep) and immune enzymes (PO, T-AOC, T-NOS, and NO), and the expression levels of immune-related genes (proPO, ALF, Lys, HSP70, Trx, Muc-1, Muc-2, Muc-5AC, Muc-5B, and Muc-19). A microbiome analysis indicated that dietary RS increased the short-chain fatty acids (SCFAs) contents and altered the composition of the intestine microbial. Specifically, RS increased the abundances of Proteobacteria and decreased the abundance of Bacteroidetes. At the genus level, the beneficial bacteria (Lutimonas, Ruegeria, Shimia, Mesoflavibacter, and Mameliella) were enriched, which might be involved in degrading toxins and producing beneficial metabolites; while potential pathogens (Formosa and Pseudoalteromonas) were decreased in response to dietary RS. Our results revealed that dietary RS could improve the intestine health of L. vannamei, probably via modulating the intestine microbial composition and SCFAs contents, and enhancing the digestion and immunity of the shrimp.
Collapse
Affiliation(s)
- Yafei Duan
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P.R. China
| | - Yun Wang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P.R. China
| | - Qingsong Liu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P.R. China
| | - Hongbiao Dong
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P.R. China
| | - Hua Li
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P.R. China
| | - Dalin Xiong
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P.R. China
| | - Jiasong Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fishery Ecology and Environment, Guangdong Province, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P.R. China.
| |
Collapse
|
292
|
Toward rational selection criteria for selection of probiotics in pigs. ADVANCES IN APPLIED MICROBIOLOGY 2019; 107:83-112. [PMID: 31128749 DOI: 10.1016/bs.aambs.2019.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
An accurate understanding of properties of probiotics is a prerequisite for selecting probiotic organisms for use in swine production. This review aims to review selection criteria for probiotic organism in swine. The systematically investigated ecological history rather than the source of isolates should be regarded as the natural origin of probiotic strains, which helps to correct the inconsistencies arising from incorrect identification of the source. Moreover, in vivo studies are suggested as follow-up assessment to validate the characteristics of probiotic predicted by in vitro experiments. In addition, the intended probiotic effect depends on the age of the animal and disease prevention in young animals may require different probiotic strains when compared to growth promotion in older animals. With adequate selection criteria, the inclusion of probiotic in feed supplementation is a promising way to exert positive effects on sows, newborns, weanling animals and grower-finisher pigs. Both host-adapted probiotics and nomadic probiotics can be applied for pathogen inhibition but host adapted organisms appear to have a different mode of action. Host-adapted probiotic strains are likely to be associated with exclusive colonization while the nomadic or environmental strain exert better immune stimulating functions. Strains with potent enzymatic activity are fitter for grower pigs favoring feed digestion and enhancing growth performance.
Collapse
|
293
|
Neonatal intestinal immune regulation by the commensal bacterium, P. UF1. Mucosal Immunol 2019; 12:434-444. [PMID: 30647410 PMCID: PMC6375783 DOI: 10.1038/s41385-018-0125-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/23/2018] [Accepted: 12/07/2018] [Indexed: 02/04/2023]
Abstract
Newborns are highly susceptible to pathogenic infections with significant worldwide morbidity possibly due to an immature immune system. Recently, we reported that Propionibacterium strain, P. UF1, isolated from the gut microbiota of preterm infants, induced the differentiation of bacteria-specific Th17 cells. Here, we demonstrate that P. UF1 significantly increased the number of protective Th17 cells and maintained IL-10+ regulatory T cells (Tregs) in newborn mice. In addition, P. UF1 protected mice from intestinal Listeria monocytogenes (L. m) infection. P. UF1 also functionally sustained the gut microbiota and induced critical B vitamin metabolites implicated in the regulation of T cell immunity during L. m intestinal infection. Transcriptomic analysis of P. UF1-induced Th17 cells revealed genes involved in the differentiation and regulation of these cells. These results illustrate the potency of P. UF1 in the enhancement of neonatal host defense against intestinal pathogen infection.
Collapse
|
294
|
Vu D, Groenewald M, de Vries M, Gehrmann T, Stielow B, Eberhardt U, Al-Hatmi A, Groenewald J, Cardinali G, Houbraken J, Boekhout T, Crous P, Robert V, Verkley G. Large-scale generation and analysis of filamentous fungal DNA barcodes boosts coverage for kingdom fungi and reveals thresholds for fungal species and higher taxon delimitation. Stud Mycol 2019; 92:135-154. [PMID: 29955203 PMCID: PMC6020082 DOI: 10.1016/j.simyco.2018.05.001] [Citation(s) in RCA: 487] [Impact Index Per Article: 81.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Species identification lies at the heart of biodiversity studies that has in recent years favoured DNA-based approaches. Microbial Biological Resource Centres are a rich source for diverse and high-quality reference materials in microbiology, and yet the strains preserved in these biobanks have been exploited only on a limited scale to generate DNA barcodes. As part of a project funded in the Netherlands to barcode specimens of major national biobanks, sequences of two nuclear ribosomal genetic markers, the Internal Transcribed Spaces and 5.8S gene (ITS) and the D1/D2 domain of the 26S Large Subunit (LSU), were generated as DNA barcode data for ca. 100 000 fungal strains originally assigned to ca. 17 000 species in the CBS fungal biobank maintained at the Westerdijk Fungal Biodiversity Institute, Utrecht. Using more than 24 000 DNA barcode sequences of 12 000 ex-type and manually validated filamentous fungal strains of 7 300 accepted species, the optimal identity thresholds to discriminate filamentous fungal species were predicted as 99.6 % for ITS and 99.8 % for LSU. We showed that 17 % and 18 % of the species could not be discriminated by the ITS and LSU genetic markers, respectively. Among them, ∼8 % were indistinguishable using both genetic markers. ITS has been shown to outperform LSU in filamentous fungal species discrimination with a probability of correct identification of 82 % vs. 77.6 %, and a clustering quality value of 84 % vs. 77.7 %. At higher taxonomic classifications, LSU has been shown to have a better discriminatory power than ITS. With a clustering quality value of 80 %, LSU outperformed ITS in identifying filamentous fungi at the ordinal level. At the generic level, the clustering quality values produced by both genetic markers were low, indicating the necessity for taxonomic revisions at genus level and, likely, for applying more conserved genetic markers or even whole genomes. The taxonomic thresholds predicted for filamentous fungal identification at the genus, family, order and class levels were 94.3 %, 88.5 %, 81.2 % and 80.9 % based on ITS barcodes, and 98.2 %, 96.2 %, 94.7 % and 92.7 % based on LSU barcodes. The DNA barcodes used in this study have been deposited to GenBank and will also be publicly available at the Westerdijk Institute's website as reference sequences for fungal identification, marking an unprecedented data release event in global fungal barcoding efforts to date.
Collapse
Affiliation(s)
- D. Vu
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - M. Groenewald
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - M. de Vries
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - T. Gehrmann
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - B. Stielow
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - U. Eberhardt
- Staatliches Museum f. Naturkunde Stuttgart, Abt. Botanik, Rosenstein 1, D-70191 Stuttgart, Germany
| | - A. Al-Hatmi
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - J.Z. Groenewald
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - G. Cardinali
- University of Perugia, Dept. of Pharmaceutical Sciences, Via Borgo 20 Giugno 74, I 06121 Perugia, Italy
| | - J. Houbraken
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - T. Boekhout
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, The Netherlands
| | - P.W. Crous
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Wageningen University and Research Centre (WUR), Laboratory of Phytopathology, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Department of Genetics, Biochemistry and Microbiology, Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria 0028, South Africa
| | - V. Robert
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - G.J.M. Verkley
- Westerdijk Fungal Biodiversity Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
295
|
Swaggerty CL, Callaway TR, Kogut MH, Piva A, Grilli E. Modulation of the Immune Response to Improve Health and Reduce Foodborne Pathogens in Poultry. Microorganisms 2019; 7:E65. [PMID: 30823445 PMCID: PMC6462950 DOI: 10.3390/microorganisms7030065] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/19/2019] [Accepted: 02/21/2019] [Indexed: 11/16/2022] Open
Abstract
Salmonella and Campylobacter are the two leading causes of bacterial-induced foodborne illness in the US. Food production animals including cattle, swine, and chickens are transmission sources for both pathogens. The number of Salmonella outbreaks attributed to poultry has decreased. However, the same cannot be said for Campylobacter where 50⁻70% of human cases result from poultry products. The poultry industry selects heavily on performance traits which adversely affects immune competence. Despite increasing demand for poultry, regulations and public outcry resulted in the ban of antibiotic growth promoters, pressuring the industry to find alternatives to manage flock health. One approach is to incorporate a program that naturally enhances/modulates the bird's immune response. Immunomodulation of the immune system can be achieved using a targeted dietary supplementation and/or feed additive to alter immune function. Science-based modulation of the immune system targets ways to reduce inflammation, boost a weakened response, manage gut health, and provide an alternative approach to prevent disease and control foodborne pathogens when conventional methods are not efficacious or not available. The role of immunomodulation is just one aspect of an integrated, coordinated approach to produce healthy birds that are also safe and wholesome products for consumers.
Collapse
Affiliation(s)
- Christina L Swaggerty
- United States Department of Agriculture/ARS, 2881 F and B Road, College Station, TX 77845, USA.
| | - Todd R Callaway
- Department of Animal and Dairy Science, University of Georgia, 425 River Road, Athens, GA 30602, USA.
| | - Michael H Kogut
- United States Department of Agriculture/ARS, 2881 F and B Road, College Station, TX 77845, USA.
| | - Andrea Piva
- Vetagro S.p.A., Via Porro 2, 42124, Reggio Emilia, Italy.
| | - Ester Grilli
- Vetagro S.p.A., Via Porro 2, 42124, Reggio Emilia, Italy.
| |
Collapse
|
296
|
Xie Y, Zhou G, Wang C, Xu X, Li C. Specific Microbiota Dynamically Regulate the Bidirectional Gut-Brain Axis Communications in Mice Fed Meat Protein Diets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:1003-1017. [PMID: 30541283 DOI: 10.1021/acs.jafc.8b05654] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The purpose of this study was to characterize the dynamic changes of different protein diets to gut microbiota and explore the influence on communications between the gut and the brain. C57BL/6J mice were fed casein, soy protein, and four kinds of processed meat proteins at a normal dose of 20% for 8 months. Bacteroidales S24-7 abundance increased from 4 to 8 months, whereas the abundances of six genera including Akkermansia decreased remarkably. Lachnospiraceae Unclassified abundance in the emulsion-type sausage protein and stewed pork protein groups showed an opposite change from 4 to 8 months. Twenty-eight and 48 specific operational taxonomy units in cecum and colon respectively were involved in regulating serotonin, peptide YY, leptin, and insulin levels. Specific microbiota was involved, directly or indirectly through signaling molecules, in the regulation of body metabolism, which may affect the communications between the gut and brain and cause different growth performances.
Collapse
Affiliation(s)
- Yunting Xie
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MOA , Nanjing Agricultural University , Nanjing 210095 , P.R. China
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MOA , Nanjing Agricultural University , Nanjing 210095 , P.R. China
- Joint International Research Laboratory of Animal Health and Food Safety, MOE , Nanjing Agricultural University , Nanjing 210095 , P.R. China
| | - Chao Wang
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MOA , Nanjing Agricultural University , Nanjing 210095 , P.R. China
| | - Xinglian Xu
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MOA , Nanjing Agricultural University , Nanjing 210095 , P.R. China
- Joint International Research Laboratory of Animal Health and Food Safety, MOE , Nanjing Agricultural University , Nanjing 210095 , P.R. China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Key Laboratory of Meat Products Processing, MOA , Nanjing Agricultural University , Nanjing 210095 , P.R. China
- Joint International Research Laboratory of Animal Health and Food Safety, MOE , Nanjing Agricultural University , Nanjing 210095 , P.R. China
- National Center for International Research on Animal Gut Nutrition , Nanjing Agricultural University , Nanjing 210095 , P.R. China
| |
Collapse
|
297
|
Lee KH, Gordon A, Shedden K, Kuan G, Ng S, Balmaseda A, Foxman B. The respiratory microbiome and susceptibility to influenza virus infection. PLoS One 2019; 14:e0207898. [PMID: 30625134 PMCID: PMC6326417 DOI: 10.1371/journal.pone.0207898] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/06/2018] [Indexed: 12/27/2022] Open
Abstract
Influenza is a major cause of morbidity and mortality worldwide. However, vaccine effectiveness has been low to moderate in recent years and vaccine coverage remains low, especially in low- and middle-income countries. Supplementary methods of prevention should be explored to reduce the high burden of influenza. A potential target is the respiratory tract microbiome, complex microbial communities which envelop the respiratory epithelium and play an important role in shaping host immunity. Using a household transmission study, we examined whether the nose/throat microbiota was associated with influenza susceptibility among participants exposed to influenza virus in the household. Further, we characterized changes in the nose/throat microbiota to explore whether community stability was influenced by influenza virus infection. Using a generalized linear mixed effects model, we found a nasal/oropharyngeal community state type (CST) associated with decreased susceptibility to influenza. The CST was rare and transitory among young children but a prevalent and stable CST among adults. Using boosting and linear mixed effects models, we found associations between the nose/throat microbiota and influenza also existed at the taxa level, specifically with the relative abundance of Alloprevotella, Prevotella, and Bacteroides oligotypes. We found high rates of change between bacterial community states among both secondary cases and household contacts who were not infected during follow up. Further work is needed to separate the effect of influenza virus infection from the considerable short-term changes that occur even in the absence of virus. Lastly, age was strongly associated with susceptibility to influenza and the nose/throat bacterial community structure. Although additional studies are needed to determine causality, our results suggest the nose/throat microbiome may be a potential target for reducing the burden of influenza.
Collapse
Affiliation(s)
- Kyu Han Lee
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kerby Shedden
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Statistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Guillermina Kuan
- Centro de Salud Sócrates Flores Vivas, Ministry of Health, Managua, Nicaragua
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Sophia Ng
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Betsy Foxman
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
298
|
Cao W, Wang C, Chin Y, Chen X, Gao Y, Yuan S, Xue C, Wang Y, Tang Q. DHA-phospholipids (DHA-PL) and EPA-phospholipids (EPA-PL) prevent intestinal dysfunction induced by chronic stress. Food Funct 2019; 10:277-288. [DOI: 10.1039/c8fo01404c] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
DHA-PL and EPA-PL may effectively protect mice against intestinal dysfunction under chronic stress exposure.
Collapse
Affiliation(s)
- Wanxiu Cao
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Chengcheng Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Yaoxian Chin
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Xin Chen
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Yuan Gao
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Shihan Yuan
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Changhu Xue
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Yuming Wang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| | - Qingjuan Tang
- College of Food Science and Engineering
- Ocean University of China
- Qingdao
- China
| |
Collapse
|
299
|
Grainger J, Daw R, Wemyss K. Systemic instruction of cell-mediated immunity by the intestinal microbiome. F1000Res 2018; 7:F1000 Faculty Rev-1910. [PMID: 30631436 PMCID: PMC6290979 DOI: 10.12688/f1000research.14633.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2018] [Indexed: 12/11/2022] Open
Abstract
Recent research has shed light on the plethora of mechanisms by which the gastrointestinal commensal microbiome can influence the local immune response in the gut (in particular, the impact of the immune system on epithelial barrier homeostasis and ensuring microbial diversity). However, an area that is much less well explored but of tremendous therapeutic interest is the impact the gut microbiome has on systemic cell-mediated immune responses. In this commentary, we highlight some key studies that are beginning to broadly examine the different mechanisms by which the gastrointestinal microbiome can impact the systemic immune compartment. Specifically, we discuss the effects of the gut microbiome on lymphocyte polarisation and trafficking, tailoring of resident immune cells in the liver, and output of circulating immune cells from the bone marrow. Finally, we explore contexts in which this new understanding of long-range effects of the gut microbiome can have implications, including cancer therapies and vaccination.
Collapse
Affiliation(s)
- John Grainger
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Collaborative Centre for Inflammation Research, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Rufus Daw
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Collaborative Centre for Inflammation Research, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Kelly Wemyss
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Collaborative Centre for Inflammation Research, Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
300
|
Lamarche D, Johnstone J, Zytaruk N, Clarke F, Hand L, Loukov D, Szamosi JC, Rossi L, Schenck LP, Verschoor CP, McDonald E, Meade MO, Marshall JC, Bowdish DME, Karachi T, Heels-Ansdell D, Cook DJ, Surette MG. Microbial dysbiosis and mortality during mechanical ventilation: a prospective observational study. Respir Res 2018; 19:245. [PMID: 30526610 PMCID: PMC6286574 DOI: 10.1186/s12931-018-0950-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022] Open
Abstract
Background Host-associated microbial communities have important roles in tissue homeostasis and overall health. Severe perturbations can occur within these microbial communities during critical illness due to underlying diseases and clinical interventions, potentially influencing patient outcomes. We sought to profile the microbial composition of critically ill mechanically ventilated patients, and to determine whether microbial diversity is associated with illness severity and mortality. Methods We conducted a prospective, observational study of mechanically ventilated critically ill patients with a high incidence of pneumonia in 2 intensive care units (ICUs) in Hamilton, Canada, nested within a randomized trial for the prevention of healthcare-associated infections. The microbial profiles of specimens from 3 anatomical sites (respiratory, and upper and lower gastrointestinal tracts) were characterized using 16S ribosomal RNA gene sequencing. Results We collected 65 specimens from 34 ICU patients enrolled in the trial (29 endotracheal aspirates, 26 gastric aspirates and 10 stool specimens). Specimens were collected at a median time of 3 days (lower respiratory tract and gastric aspirates; interquartile range [IQR] 2–4) and 6 days (stool; IQR 4.25–6.75) following ICU admission. We observed a loss of biogeographical distinction between the lower respiratory tract and gastrointestinal tract microbiota during critical illness. Moreover, microbial diversity in the respiratory tract was inversely correlated with APACHE II score (r = − 0.46, p = 0.013) and was associated with hospital mortality (Median Shannon index: Discharged alive; 1.964 vs. Deceased; 1.348, p = 0.045). Conclusions The composition of the host-associated microbial communities is severely perturbed during critical illness. Reduced microbial diversity reflects high illness severity and is associated with mortality. Microbial diversity may be a biomarker of prognostic value in mechanically ventilated patients. Trial registration ClinicalTrials.gov ID NCT01782755. Registered February 4 2013. Electronic supplementary material The online version of this article (10.1186/s12931-018-0950-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daphnée Lamarche
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Jennie Johnstone
- Department of Medicine, University of Toronto, Toronto, ON, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Public Health Ontario, Toronto, ON, Canada
| | - Nicole Zytaruk
- St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - France Clarke
- St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Lori Hand
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Hamilton Health Sciences, Hamilton, ON, Canada
| | - Dessi Loukov
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, Hamilton, ON, Canada
| | - Jake C Szamosi
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Laura Rossi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Louis P Schenck
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Chris P Verschoor
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Ellen McDonald
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Maureen O Meade
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Hamilton Health Sciences, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - John C Marshall
- Department of Surgery, University of Toronto, Toronto, ON, Canada.,Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada
| | - Dawn M E Bowdish
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, Hamilton, ON, Canada
| | - Tim Karachi
- Hamilton Health Sciences, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Diane Heels-Ansdell
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Deborah J Cook
- St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada. .,Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada. .,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada. .,Department of Medicine, McMaster University, Health Sciences Bldg, 3N8F, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
| | | | | | | |
Collapse
|