1
|
Weber-Stiehl S, Taubenheim J, Järke L, Röcken C, Schreiber S, Aden K, Kaleta C, Rosenstiel P, Sommer F. Hexokinase 2 expression in apical enterocytes correlates with inflammation severity in patients with inflammatory bowel disease. BMC Med 2024; 22:490. [PMID: 39444028 PMCID: PMC11515617 DOI: 10.1186/s12916-024-03710-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Inflammation is characterized by a metabolic switch promoting glycolysis and lactate production. Hexokinases (HK) catalyze the first reaction of glycolysis and inhibition of epithelial HK2 protected from colitis in mice. HK2 expression has been described as elevated in patients with intestinal inflammation; however, there is conflicting data from few cohorts especially with severely inflamed individuals; thus, systematic studies linking disease activity with HK2 levels are needed. METHODS We examined the relationship between HK2 expression and inflammation severity using bulk transcriptome data derived from the mucosa of thoroughly phenotyped inflammatory bowel disease (IBD) patients of two independent cohorts including both subtypes Crohn's disease (CD) and ulcerative colitis (UC). Publicly available single-cell RNA sequencing data were analyzed, and immunofluorescence staining on colonic biopsies of unrelated patients with intestinal inflammation was performed to confirm the RNA-based findings on cellular and protein level. RESULTS HK2 expression gradually increased from mild to intermediate inflammation, yet strongly declined at high inflammation scores. Expression of epithelial marker genes also declined at high inflammation scores, whereas that of candidate immune marker genes increased, indicating a cellular remodeling of the mucosa during inflammation with an infiltration of HK2-negative immune cells and a loss of terminal differentiated epithelial cells in the apical epithelium-the main site of HK2 expression. Normalizing for the enterocyte loss clearly identified epithelial HK2 expression as gradually increasing with disease activity and remaining elevated at high inflammation scores. HK2 protein expression was mostly restricted to brush border enterocytes, and these cells along with HK2 levels vanished with increasing disease severity. CONCLUSIONS Our findings clearly define dysregulated epithelial HK2 expression as an indicator of disease activity in intestinal inflammation and suggest targeted HK2-inhibition as a potential therapeutic avenue.
Collapse
Affiliation(s)
- Saskia Weber-Stiehl
- Institute of Clinical Molecular Biology, University of Kiel, Rosalind-Franklin-Straße 12, Kiel, 24105, Germany
| | - Jan Taubenheim
- Institute of Experimental Medicine, University of Kiel & University Hospital Schleswig-Holstein, Michaelisstr. 5, Kiel, 24105, Germany
| | - Lea Järke
- Institute of Clinical Molecular Biology, University of Kiel, Rosalind-Franklin-Straße 12, Kiel, 24105, Germany
| | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3/House U33, Kiel, 24105, Germany
| | - Stefan Schreiber
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Konrad Aden
- Institute of Clinical Molecular Biology, University of Kiel, Rosalind-Franklin-Straße 12, Kiel, 24105, Germany
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Christoph Kaleta
- Institute of Experimental Medicine, University of Kiel & University Hospital Schleswig-Holstein, Michaelisstr. 5, Kiel, 24105, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, Rosalind-Franklin-Straße 12, Kiel, 24105, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, University of Kiel, Rosalind-Franklin-Straße 12, Kiel, 24105, Germany.
| |
Collapse
|
2
|
Korsten SGPJ, Hartog M, Berends AJ, Koenders MI, Popa CD, Vromans H, Garssen J, van de Ende CHM, Vermeiden JPW, Willemsen LEM. A Sustained-Release Butyrate Tablet Suppresses Ex Vivo T Helper Cell Activation of Osteoarthritis Patients in a Double-Blind Placebo-Controlled Randomized Trial. Nutrients 2024; 16:3384. [PMID: 39408351 PMCID: PMC11478393 DOI: 10.3390/nu16193384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Degenerative joint disease osteoarthritis (OA) is characterized by the degeneration of cartilage, synovial inflammation and low-grade systemic inflammation in association with microbial dysbiosis and intestinal barrier defects. Butyrate is known for its anti-inflammatory and barrier protective effects and might benefit OA patients. In a double-blind placebo-controlled randomized trial, the effects of four to five weeks of oral treatment with sustained-release (SR) butyrate tablets (600 mg/day) on systemic inflammation and immune function were studied in hand OA patients. Serum markers for systemic inflammation and lipopolysaccharide (LPS) leakage were measured and ex vivo stimulation of whole blood or peripheral blood mononuclear cells (PBMCs) was performed at baseline and after treatment. Butyrate treatment did not affect the serum markers nor the cytokine release of ex vivo LPS-stimulated whole blood or PBMCs nor the phenotype of restimulated monocytes. By contrast, butyrate treatment reduced the percentage of activated T helper (Th) cells and the Th17/Treg ratio in αCD3/CD28-activated PBMCs, though cytokine release upon stimulation remained unaffected. Nevertheless, the percentage of CD4+IL9+ cells was reduced by butyrate as compared to the placebo. In both groups, the frequency of Th1, Treg, Th17, activated Th17, CD4+IFNγ+ and CD4+TNFα+ cells was reduced. This study shows a proof of principle of some immunomodulatory effects using a SR butyrate treatment in hand OA patients. The inflammatory phenotype of Th cells was reduced, as indicated by a reduced percentage of Th9 cells, activated Th cells and improved Th17/Treg balance in ex vivo αCD3/CD28-activated PBMCs. Future studies are warranted to further optimize the butyrate dose regime to ameliorate inflammation in OA patients.
Collapse
Affiliation(s)
- Sandra G. P. J. Korsten
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands (J.G.)
- Tiofarma B.V., 3261 ME Oud-Beijerland, The Netherlands
| | - Merel Hartog
- Department of Research, Sint Maartenskliniek, 6574 NA Ubbergen, The Netherlands; (M.H.); (C.H.M.v.d.E.)
- Department of Rheumatology, Sint Maartenskliniek, 6574 NA Ubbergen, The Netherlands;
| | - Alinda J. Berends
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands (J.G.)
| | - Marije I. Koenders
- Department of Rheumatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Calin D. Popa
- Department of Rheumatology, Sint Maartenskliniek, 6574 NA Ubbergen, The Netherlands;
- Department of Rheumatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Herman Vromans
- Tiofarma B.V., 3261 ME Oud-Beijerland, The Netherlands
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands (J.G.)
- Danone/Nutricia Research B.V., 3584 CT Utrecht, The Netherlands
| | - Cornelia H. M. van de Ende
- Department of Research, Sint Maartenskliniek, 6574 NA Ubbergen, The Netherlands; (M.H.); (C.H.M.v.d.E.)
- Department of Rheumatology, Sint Maartenskliniek, 6574 NA Ubbergen, The Netherlands;
| | | | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands (J.G.)
| |
Collapse
|
3
|
Quinn-Bohmann N, Wilmanski T, Sarmiento KR, Levy L, Lampe JW, Gurry T, Rappaport N, Ostrem EM, Venturelli OS, Diener C, Gibbons SM. Microbial community-scale metabolic modelling predicts personalized short-chain fatty acid production profiles in the human gut. Nat Microbiol 2024; 9:1700-1712. [PMID: 38914826 DOI: 10.1038/s41564-024-01728-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/09/2024] [Indexed: 06/26/2024]
Abstract
Microbially derived short-chain fatty acids (SCFAs) in the human gut are tightly coupled to host metabolism, immune regulation and integrity of the intestinal epithelium. However, the production of SCFAs can vary widely between individuals consuming the same diet, with lower levels often associated with disease. A systems-scale mechanistic understanding of this heterogeneity is lacking. Here we use a microbial community-scale metabolic modelling (MCMM) approach to predict individual-specific SCFA production profiles to assess the impact of different dietary, prebiotic and probiotic inputs. We evaluate the quantitative accuracy of our MCMMs using in vitro and ex vivo data, plus published human cohort data. We find that MCMM SCFA predictions are significantly associated with blood-derived clinical chemistries, including cardiometabolic and immunological health markers, across a large human cohort. Finally, we demonstrate how MCMMs can be leveraged to design personalized dietary, prebiotic and probiotic interventions aimed at optimizing SCFA production in the gut. Our model represents an approach to direct gut microbiome engineering for precision health and nutrition.
Collapse
Affiliation(s)
- Nick Quinn-Bohmann
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA
| | | | | | - Lisa Levy
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Thomas Gurry
- Pharmaceutical Biochemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Myota GmbH, Berlin, Germany
| | - Noa Rappaport
- Center for Phenomic Health, Buck Institute for Research on Aging, Novato, CA, USA
| | - Erin M Ostrem
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ophelia S Venturelli
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Christian Diener
- Institute for Systems Biology, Seattle, WA, USA.
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria.
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA.
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- eScience Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
4
|
Pietrzak A, Banasiewicz T. Applicability of sodium butyrate preparations from a surgeon's and gastroenterologist's perspective. POLISH JOURNAL OF SURGERY 2024; 96:68-73. [PMID: 38629276 DOI: 10.5604/01.3001.0054.4152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
In recent years, much has been written about the possibilities of using exogenous sodium butyrate in the prevention and treatment of gastrointestinal diseases, in prehabilitation, in peri- and postoperative treatment, as well as its local application. It became possible thanks to the development of a special formulation (microencapsulation technique) enabling the delivery of unstable butyrate compounds to the large intestine, where it is used primarily as a source of energy. It also plays a key role in maintaining body homeostasis by maintaining the integrity of the intestinal epithelium and stimulating the intestinal immune system. There is growing evidence of the effectiveness of sodium butyrate in various areas of health. The following article discusses the possibilities of using microencapsulated sodium butyrate in the prevention and treatment of gastrointestinal diseases from the perspective of a gastroenterologist and gastrointestinal surgeon.
Collapse
Affiliation(s)
- Anna Pietrzak
- 2nd Department of Gastroenterology, Centre of Postgraduate Medical Education, Warsaw, Poland; Department of Gastroenterology, Bielanski Hospital in Warsaw, Poland
| | - Tomasz Banasiewicz
- Chair and Department of General Surgery, Endocrine and Gastroenterological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
5
|
Facchin S, Bertin L, Bonazzi E, Lorenzon G, De Barba C, Barberio B, Zingone F, Maniero D, Scarpa M, Ruffolo C, Angriman I, Savarino EV. Short-Chain Fatty Acids and Human Health: From Metabolic Pathways to Current Therapeutic Implications. Life (Basel) 2024; 14:559. [PMID: 38792581 PMCID: PMC11122327 DOI: 10.3390/life14050559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The gastrointestinal tract is home to trillions of diverse microorganisms collectively known as the gut microbiota, which play a pivotal role in breaking down undigested foods, such as dietary fibers. Through the fermentation of these food components, short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate are produced, offering numerous health benefits to the host. The production and absorption of these SCFAs occur through various mechanisms within the human intestine, contingent upon the types of dietary fibers reaching the gut and the specific microorganisms engaged in fermentation. Medical literature extensively documents the supplementation of SCFAs, particularly butyrate, in the treatment of gastrointestinal, metabolic, cardiovascular, and gut-brain-related disorders. This review seeks to provide an overview of the dynamics involved in the production and absorption of acetate, propionate, and butyrate within the human gut. Additionally, it will focus on the pivotal roles these SCFAs play in promoting gastrointestinal and metabolic health, as well as their current therapeutic implications.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Erica Bonazzi
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Caterina De Barba
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Brigida Barberio
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Marco Scarpa
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Cesare Ruffolo
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Imerio Angriman
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| |
Collapse
|
6
|
Cicchinelli S, Gemma S, Pignataro G, Piccioni A, Ojetti V, Gasbarrini A, Franceschi F, Candelli M. Intestinal Fibrogenesis in Inflammatory Bowel Diseases: Exploring the Potential Role of Gut Microbiota Metabolites as Modulators. Pharmaceuticals (Basel) 2024; 17:490. [PMID: 38675450 PMCID: PMC11053610 DOI: 10.3390/ph17040490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Fibrosis, sustained by the transformation of intestinal epithelial cells into fibroblasts (epithelial-to-mesenchymal transition, EMT), has been extensively studied in recent decades, with the molecular basis well-documented in various diseases, including inflammatory bowel diseases (IBDs). However, the factors influencing these pathways remain unclear. In recent years, the role of the gut microbiota in health and disease has garnered significant attention. Evidence suggests that an imbalanced or dysregulated microbiota, along with environmental and genetic factors, may contribute to the development of IBDs. Notably, microbes produce various metabolites that interact with host receptors and associated signaling pathways, influencing physiological and pathological changes. This review aims to present recent evidence highlighting the emerging role of the most studied metabolites as potential modulators of molecular pathways implicated in intestinal fibrosis and EMT in IBDs. These studies provide a deeper understanding of intestinal inflammation and fibrosis, elucidating the molecular basis of the microbiota role in IBDs, paving the way for future treatments.
Collapse
Affiliation(s)
- Sara Cicchinelli
- Department of Emergency, S.S. Filippo e Nicola Hospital, 67051 Avezzano, Italy;
| | - Stefania Gemma
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giulia Pignataro
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Veronica Ojetti
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
7
|
Williams LM, Cao S. Harnessing and delivering microbial metabolites as therapeutics via advanced pharmaceutical approaches. Pharmacol Ther 2024; 256:108605. [PMID: 38367866 PMCID: PMC10985132 DOI: 10.1016/j.pharmthera.2024.108605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Abstract
Microbial metabolites have emerged as key players in the interplay between diet, the gut microbiome, and host health. Two major classes, short-chain fatty acids (SCFAs) and tryptophan (Trp) metabolites, are recognized to regulate inflammatory, immune, and metabolic responses within the host. Given that many human diseases are associated with dysbiosis of the gut microbiome and consequent reductions in microbial metabolite production, the administration of these metabolites represents a direct, multi-targeted treatment. While a multitude of preclinical studies showcase the therapeutic potential of both SCFAs and Trp metabolites, they often rely on high doses and frequent dosing regimens to achieve systemic effects, thereby constraining their clinical applicability. To address these limitations, a variety of pharmaceutical formulations approaches that enable targeted, delayed, and/or sustained microbial metabolite delivery have been developed. These approaches, including enteric encapsulations, esterification to dietary fiber, prodrugs, and nanoformulations, pave the way for the next generation of microbial metabolite-based therapeutics. In this review, we first provide an overview of the roles of microbial metabolites in maintaining host homeostasis and outline how compromised metabolite production contributes to the pathogenesis of inflammatory, metabolic, autoimmune, allergic, infectious, and cancerous diseases. Additionally, we explore the therapeutic potential of metabolites in these disease contexts. Then, we provide a comprehensive and up-to-date review of the pharmaceutical strategies that have been employed to enhance the therapeutic efficacy of microbial metabolites, with a focus on SCFAs and Trp metabolites.
Collapse
Affiliation(s)
- Lindsey M Williams
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Shijie Cao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
8
|
Liu S, Li F, Cai Y, Ren L, Sun L, Gang X, Wang G. Bacteroidaceae, Bacteroides, and Veillonella: emerging protectors against Graves' disease. Front Cell Infect Microbiol 2024; 14:1288222. [PMID: 38404289 PMCID: PMC10884117 DOI: 10.3389/fcimb.2024.1288222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
Background Graves' disease (GD) is the most common cause of hyperthyroidism, and its pathogenesis remains incompletely elucidated. Numerous studies have implicated the gut microbiota in the development of thyroid disorders. This study employs Mendelian randomization analysis to investigate the characteristics of gut microbiota in GD patients, aiming to offer novel insights into the etiology and treatment of Graves' disease. Methods Two-sample Mendelian randomization (MR) analysis was employed to assess the causal relationship between Graves' disease and the gut microbiota composition. Gut microbiota data were sourced from the international consortium MiBioGen, while Graves' disease data were obtained from FINNGEN. Eligible single nucleotide polymorphisms (SNPs) were selected as instrumental variables. Multiple analysis methods, including inverse variance-weighted (IVW), MR-Egger regression, weighted median, weighted mode, and MR-RAPS, were utilized. Sensitivity analyses were conducted employing MR-Egger intercept test, Cochran's Q test, and leave-one-out analysis as quality control measures. Results The Mendelian randomization study conducted in a European population revealed a decreased risk of Graves' disease associated with Bacteroidaceae (Odds ratio (OR) [95% confidence interval (CI)]: 0.89 [0.89 ~ 0.90], adjusted P value: <0.001), Bacteroides (OR: [95% CI]: 0.555 [0.437 ~ 0.706], adjusted P value: <0.001), and Veillonella (OR [95% CI]: 0.632 [0.492 ~ 0.811], adjusted P value: 0.016). No significant evidence of heterogeneity, or horizontal pleiotropy was detected. Furthermore, the preliminary MR analysis identified 13 bacterial species including Eubacterium brachy group and Family XIII AD3011 group, exhibiting significant associations with Graves' disease onset, suggesting potential causal effects. Conclusion A causal relationship exists between gut microbiota and Graves' disease. Bacteroidaceae, Bacteroides, and Veillonella emerge as protective factors against Graves' disease development. Prospective probiotic supplementation may offer a novel avenue for adjunctive treatment in the management of Graves' disease in the future.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Jilin, Changchun, China
| | - Fan Li
- Department of Gastroenterology, The First Hospital of Jilin University, Jilin, Changchun, China
| | - Yunjia Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Jilin, Changchun, China
| | - Linan Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Jilin, Changchun, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Jilin, Changchun, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Jilin, Changchun, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Jilin, Changchun, China
| |
Collapse
|
9
|
Perricone V, Schokker D, Bossers A, de Bruijn A, Kar SK, Te Pas MFW, Rebel JMJ, Wouters IM, de Jong IC. Dietary strategies can increase cloacal endotoxin levels and modulate the resident microbiota in broiler chickens. Poult Sci 2024; 103:103312. [PMID: 38100944 PMCID: PMC10762469 DOI: 10.1016/j.psj.2023.103312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
Endotoxins released from poultry feces have been associated with impaired human health. Because endotoxins are released from gram-negative intestinal bacteria, it was hypothesized that dietary strategies may influence endotoxin excretion via modulation of gut microbiota. We therefore tested dietary strategies that could potentially reduce cloacal endotoxin levels in broiler chickens. One-day-old male Ross 308 (N = 1,344) broilers were housed in 48 pens (N = 8 pens/treatment, 28 chickens per pen) and fed 1 of 6 diets for 35 days (d) in a 3-phase feeding program: a basic diet (CON) that served as the reference diet, or basic diet supplemented with butyrate (BUT), inulin (INU), medium-chain fatty acids (MCFA) or Original XPC™LS (XPC), or a high-fiber-low-protein (HF-LP) diet. A significant (P < 0.05) increase in cloacal endotoxin concentration at d 35 was observed in BUT as compared to CON. Analysis of cloacal microbiota showed a trend (P < 0.07) for a higher gram-negative/gram-positive ratio and for a higher relative abundance of gram-negative bacteria at d 35 (P ≤ 0.08) in BUT and HF-LP as compared to CON. A significant (P < 0.05) increase in average daily gain (ADG) and improved feed conversion ratio (P < 0.05) were observed in MCFA during the grower phase (d 14-28), and a significant (P < 0.05) increase in average daily feed intake (ADFI) was observed in MCFA during d 0 to 28. Broilers fed HF-LP had a significantly (P < 0.05) higher FCR and lower ADG throughout the rearing period. No treatment effects were found on footpad dermatitis, but BUT had worst hock burn scores at d 35 (P < 0.01) and MCFA had worst cleanliness scores at d 21 but not at d 35 (treatment*age P < 0.05), while INU had better cleanliness as compared to CON at d 35 (P < 0.05). In conclusion, especially BUT and HF-LP were able to modulate resident microbiota and BUT also increased cloacal endotoxin levels, which was opposite to our hypothesis. The present study indicates that cloacal endotoxin release can be affected by the diet but further study is needed to find dietary treatments that can reduce cloacal endotoxin release.
Collapse
Affiliation(s)
- Vera Perricone
- Wageningen Livestock Research, Wageningen University and Research, 6700 AH Wageningen, the Netherlands
| | - Dirkjan Schokker
- Wageningen Livestock Research, Wageningen University and Research, 6700 AH Wageningen, the Netherlands; Wageningen Bioveterinary Research, Wageningen University and Research, 8221 RA Lelystad, the Netherlands
| | - Alex Bossers
- Wageningen Bioveterinary Research, Wageningen University and Research, 8221 RA Lelystad, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3508 TD Utrecht, the Netherlands
| | - Anne de Bruijn
- Wageningen Livestock Research, Wageningen University and Research, 6700 AH Wageningen, the Netherlands
| | - Soumya K Kar
- Wageningen Livestock Research, Wageningen University and Research, 6700 AH Wageningen, the Netherlands
| | - Marinus F W Te Pas
- Wageningen Livestock Research, Wageningen University and Research, 6700 AH Wageningen, the Netherlands
| | - Johanna M J Rebel
- Wageningen Livestock Research, Wageningen University and Research, 6700 AH Wageningen, the Netherlands; Wageningen Bioveterinary Research, Wageningen University and Research, 8221 RA Lelystad, the Netherlands
| | - Inge M Wouters
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3508 TD Utrecht, the Netherlands
| | - Ingrid C de Jong
- Wageningen Livestock Research, Wageningen University and Research, 6700 AH Wageningen, the Netherlands.
| |
Collapse
|
10
|
Ambat A, Antony L, Maji A, Ghimire S, Mattiello S, Kashyap PC, More S, Sebastian V, Scaria J. Enhancing recovery from gut microbiome dysbiosis and alleviating DSS-induced colitis in mice with a consortium of rare short-chain fatty acid-producing bacteria. Gut Microbes 2024; 16:2382324. [PMID: 39069899 PMCID: PMC11290756 DOI: 10.1080/19490976.2024.2382324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 05/24/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024] Open
Abstract
The human gut microbiota is a complex community comprising hundreds of species, with a few present in high abundance and the vast majority in low abundance. The biological functions and effects of these low-abundant species on their hosts are not yet fully understood. In this study, we assembled a bacterial consortium (SC-4) consisting of B. paravirosa, C. comes, M. indica, and A. butyriciproducens, which are low-abundant, short-chain fatty acid (SCFA)-producing bacteria isolated from healthy human gut, and tested its effect on host health using germ-free and human microbiota-associated colitis mouse models. The selection also favored these four bacteria being reduced in abundance in either Ulcerative Colitis (UC) or Crohn's disease (CD) metagenome samples. Our findings demonstrate that SC-4 can colonize germ-free (GF) mice, increasing mucin thickness by activating MUC-1 and MUC-2 genes, thereby protecting GF mice from Dextran Sodium Sulfate (DSS)-induced colitis. Moreover, SC-4 aided in the recovery of human microbiota-associated mice from DSS-induced colitis, and intriguingly, its administration enhanced the alpha diversity of the gut microbiome, shifting the community composition closer to control levels. The results showed enhanced phenotypes across all measures when the mice were supplemented with inulin as a dietary fiber source alongside SC-4 administration. We also showed a functional redundancy existing in the gut microbiome, resulting in the low abundant SCFA producers acting as a form of insurance, which in turn accelerates recovery from the dysbiotic state upon the administration of SC-4. SC-4 colonization also upregulated iNOS gene expression, further supporting its ability to produce an increasing number of goblet cells. Collectively, our results provide evidence that low-abundant SCFA-producing species in the gut may offer a novel therapeutic approach to IBD.
Collapse
Affiliation(s)
- Achuthan Ambat
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Linto Antony
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Abhijit Maji
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Sudeep Ghimire
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Samara Mattiello
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Purna C. Kashyap
- Enteric Neuroscience Program, Department of Medicine and Physiology, Mayo Clinic, Rochester, MN, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Vanessa Sebastian
- Department of Pathology, Jubilee Mission Medical College and Research Institute, Thrissur, India
| | - Joy Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
11
|
Duan H, Wang L, Huangfu M, Li H. The impact of microbiota-derived short-chain fatty acids on macrophage activities in disease: Mechanisms and therapeutic potentials. Biomed Pharmacother 2023; 165:115276. [PMID: 37542852 DOI: 10.1016/j.biopha.2023.115276] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) derived from the fermentation of carbohydrates by gut microbiota play a crucial role in regulating host physiology. Among them, acetate, propionate, and butyrate are key players in various biological processes. Recent research has revealed their significant functions in immune and inflammatory responses. For instance, butyrate reduces the development of interferon-gamma (IFN-γ) generating cells while promoting the development of regulatory T (Treg) cells. Propionate inhibits the initiation of a Th2 immune response by dendritic cells (DCs). Notably, SCFAs have an inhibitory impact on the polarization of M2 macrophages, emphasizing their immunomodulatory properties and potential for therapeutics. In animal models of asthma, both butyrate and propionate suppress the M2 polarization pathway, thus reducing allergic airway inflammation. Moreover, dysbiosis of gut microbiota leading to altered SCFA production has been implicated in prostate cancer progression. SCFAs trigger autophagy in cancer cells and promote M2 polarization in macrophages, accelerating tumor advancement. Manipulating microbiota- producing SCFAs holds promise for cancer treatment. Additionally, SCFAs enhance the expression of hypoxia-inducible factor 1 (HIF-1) by blocking histone deacetylase, resulting in increased production of antibacterial effectors and improved macrophage-mediated elimination of microorganisms. This highlights the antimicrobial potential of SCFAs and their role in host defense mechanisms. This comprehensive review provides an in-depth analysis of the latest research on the functional aspects and underlying mechanisms of SCFAs in relation to macrophage activities in a wide range of diseases, including infectious diseases and cancers. By elucidating the intricate interplay between SCFAs and macrophage functions, this review aims to contribute to the understanding of their therapeutic potential and pave the way for future interventions targeting SCFAs in disease management.
Collapse
Affiliation(s)
- Hongliang Duan
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - LiJuan Wang
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Mingmei Huangfu
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - Hanyang Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
12
|
Nahm DH. Regulatory T Cell-Targeted Immunomodulatory Therapy for Long-Term Clinical Improvement of Atopic Dermatitis: Hypotheses and Perspectives. Life (Basel) 2023; 13:1674. [PMID: 37629531 PMCID: PMC10455293 DOI: 10.3390/life13081674] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Atopic dermatitis (AD) is a chronically relapsing inflammatory skin disorder characterized by itching and eczematous lesions. It is often associated with a personal or familial history of allergic diseases. Allergic inflammation induced by immunoglobulin E and T-helper type 2 (Th2) cell responses to common environmental agents has been suggested to play an essential role in AD pathogenesis. The standard therapies for AD, including topical or systemic agents, focus on controlling skin inflammation. Recently developed monoclonal antibody to interleukin-4 receptor alpha or Janus kinase inhibitors can provide significant clinical improvements in patients with AD by inhibiting Th2 cell-mediated skin inflammation. However, the clinical efficacy of the Th2 cell-targeted therapy is transient and incomplete in patients with AD. Patients with AD are seeking a permanent cure. Therefore, the development of novel immunomodulatory strategies that can improve a long-term clinical outcome and provide a long-term treatment-free clinical remission of AD (disease-modifying therapy) is needed. Regulatory T (Treg) cells play a critical role in the maintenance of immune tolerance and suppress the development of autoimmune and allergic diseases. This review provides three working hypotheses and perspectives for the treatment of AD by Treg cell activation. (1) A decreased number or function of Treg cells is a critical event that causes the activation of Th2 cells, leading to the development and maintenance of AD. (2) Activation of Treg cells is an effective therapeutic approach for AD. (3) Many different immunomodulatory strategies activating Treg cells can provide a long-term clinical improvement of AD by induction of immune tolerance. The Treg cell-targeted immunomodulatory therapies for AD include allergen immunotherapy, microbiota, vitamin D, polyvalent human immunoglobulin G, monoclonal antibodies to the surface antigens of T cell or antigen-presenting cell, and adoptive transfer of autologous Treg cells or genetically engineered Treg cells expanded in vitro.
Collapse
Affiliation(s)
- Dong-Ho Nahm
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
13
|
Yan X, Li J, Wu D. The Role of Short-Chain Fatty Acids in Acute Pancreatitis. Molecules 2023; 28:4985. [PMID: 37446647 DOI: 10.3390/molecules28134985] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Acute pancreatitis (AP) is a digestive emergency and can develop into a systematic illness. The role of the gut in the progression and deterioration of AP has drawn much attention from researchers, and areas of interest include dysbiosis of the intestinal flora, weakened intestinal barrier function, and bacterial and endotoxin translocation. Short-chain fatty acids (SCFAs), as one of the metabolites of gut microbiota, have been proven to be depleted in AP patients. SCFAs help restore gut homeostasis by rebuilding gut flora, stabilizing the intestinal epithelial barrier, and regulating inflammation. SCFAs can also suppress systematic inflammatory responses, improve the injured pancreas, and prevent and protect other organ dysfunctions. Based on multiple beneficial effects, increasing SCFAs is an essential idea of gut protective treatment in AP. Specific strategies include the direct use of butyrate or indirect supplementation through fiber, pre/pro/synbiotics, or fecal microbiota transplantation as a promising adjective therapy to enteral nutrition.
Collapse
Affiliation(s)
- Xiaxiao Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianing Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Dong Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Clinical Epidemiology Unit, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
14
|
Recharla N, Geesala R, Shi XZ. Gut Microbial Metabolite Butyrate and Its Therapeutic Role in Inflammatory Bowel Disease: A Literature Review. Nutrients 2023; 15:2275. [PMID: 37242159 PMCID: PMC10221771 DOI: 10.3390/nu15102275] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Background and objective: Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a chronic inflammatory disorder characterized by aberrant immune responses and compromised barrier function in the gastrointestinal tract. IBD is associated with altered gut microbiota and their metabolites in the colon. Butyrate, a gut microbial metabolite, plays a crucial role in regulating immune function, epithelial barrier function, and intestinal homeostasis. In this review, we aim to present an overview of butyrate synthesis and metabolism and the mechanism of action of butyrate in maintaining intestinal homeostasis and to discuss the therapeutic implications of butyrate in IBD. Methods: We searched the literature up to March 2023 through PubMed, Web of Science, and other sources using search terms such as butyrate, inflammation, IBD, Crohn's disease, and ulcerative colitis. Clinical studies in patients and preclinical studies in rodent models of IBD were included in the summary of the therapeutic implications of butyrate. Results: Research in the last two decades has shown the beneficial effects of butyrate on gut immune function and epithelial barrier function. Most of the preclinical and clinical studies have shown the positive effect of butyrate oral supplements in reducing inflammation and maintaining remission in colitis animal models and IBD patients. However, butyrate enema showed mixed effects. Butyrogenic diets, including germinated barley foodstuff and oat bran, are found to increase fecal butyrate concentrations and reduce the disease activity index in both animal models and IBD patients. Conclusions: The current literature suggests that butyrate is a potential add-on therapy to reduce inflammation and maintain IBD remission. Further clinical studies are needed to determine if butyrate administration alone is an effective therapeutic treatment for IBD.
Collapse
Affiliation(s)
| | | | - Xuan-Zheng Shi
- Department of Internal Medicine, The University of Texas Medical Branch, 301 University Blvd, 4.106 Basic Science Building, Galveston, TX 77555-0655, USA; (N.R.); (R.G.)
| |
Collapse
|
15
|
De Filippo C, Costa A, Becagli MV, Monroy MM, Provensi G, Passani MB. Gut microbiota and oleoylethanolamide in the regulation of intestinal homeostasis. Front Endocrinol (Lausanne) 2023; 14:1135157. [PMID: 37091842 PMCID: PMC10113643 DOI: 10.3389/fendo.2023.1135157] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
A vast literature strongly suggests that the endocannabinoid (eCB) system and related bioactive lipids (the paracannabinoid system) contribute to numerous physiological processes and are involved in pathological conditions such as obesity, type 2 diabetes, and intestinal inflammation. The gut paracannabinoid system exerts a prominent role in gut physiology as it affects motility, permeability, and inflammatory responses. Another important player in the regulation of host metabolism is the intestinal microbiota, as microorganisms are indispensable to protect the intestine against exogenous pathogens and potentially harmful resident microorganisms. In turn, the composition of the microbiota is regulated by intestinal immune responses. The intestinal microbial community plays a fundamental role in the development of the innate immune system and is essential in shaping adaptive immunity. The active interplay between microbiota and paracannabinoids is beginning to appear as potent regulatory system of the gastrointestinal homeostasis. In this context, oleoylethanolamide (OEA), a key component of the physiological systems involved in the regulation of dietary fat consumption, energy homeostasis, intestinal motility, and a key factor in modulating eating behavior, is a less studied lipid mediator. In the small intestine namely duodenum and jejunum, levels of OEA change according to the nutrient status as they decrease during food deprivation and increase upon refeeding. Recently, we and others showed that OEA treatment in rodents protects against inflammatory events and changes the intestinal microbiota composition. In this review, we briefly define the role of OEA and of the gut microbiota in intestinal homeostasis and recapitulate recent findings suggesting an interplay between OEA and the intestinal microorganisms.
Collapse
Affiliation(s)
- Carlotta De Filippo
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Alessia Costa
- Dipartimento di Scienze della Salute, Università di Firenze, Firenze, Italy
| | | | - Mariela Mejia Monroy
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Gustavo Provensi
- Dipartimento di Neurofarba, Università di Firenze, Firenze, Italy
- *Correspondence: Maria Beatrice Passani, ; Gustavo Provensi,
| | - Maria Beatrice Passani
- Dipartimento di Scienze della Salute, Università di Firenze, Firenze, Italy
- *Correspondence: Maria Beatrice Passani, ; Gustavo Provensi,
| |
Collapse
|
16
|
Dimov I, Mollova D, Vasileva T, Bivolarski V, Nikolova M, Bivolarska A, Iliev I. Metabolic profiling of probiotic strain Lactobacillus delbrueckii subsp. bulgaricus L14 cultivated in presence of prebiotic oligosaccharides and polysaccharides in simulating in vitro gastrointestinal tract system. BIOTECHNOL BIOTEC EQ 2023. [DOI: 10.1080/13102818.2023.2178825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Affiliation(s)
- Ivica Dimov
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Daniela Mollova
- Department of Biochemistry and Microbiology, Faculty of Biology, Plovdiv University “Paisii Hilendarski”, Plovdiv, Bulgaria
| | - Tonka Vasileva
- Department of Biochemistry and Microbiology, Faculty of Biology, Plovdiv University “Paisii Hilendarski”, Plovdiv, Bulgaria
| | - Veselin Bivolarski
- Department of Biochemistry and Microbiology, Faculty of Biology, Plovdiv University “Paisii Hilendarski”, Plovdiv, Bulgaria
| | - Mariana Nikolova
- Department of Biochemistry and Microbiology, Faculty of Biology, Plovdiv University “Paisii Hilendarski”, Plovdiv, Bulgaria
| | - Anelia Bivolarska
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Ilia Iliev
- Department of Biochemistry and Microbiology, Faculty of Biology, Plovdiv University “Paisii Hilendarski”, Plovdiv, Bulgaria
| |
Collapse
|
17
|
Yoon JH, Do JS, Velankanni P, Lee CG, Kwon HK. Gut Microbial Metabolites on Host Immune Responses in Health and Disease. Immune Netw 2023; 23:e6. [PMID: 36911800 PMCID: PMC9995988 DOI: 10.4110/in.2023.23.e6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 03/07/2023] Open
Abstract
Intestinal microorganisms interact with various immune cells and are involved in gut homeostasis and immune regulation. Although many studies have discussed the roles of the microorganisms themselves, interest in the effector function of their metabolites is increasing. The metabolic processes of these molecules provide important clues to the existence and function of gut microbes. The interrelationship between metabolites and T lymphocytes in particular plays a significant role in adaptive immune functions. Our current review focuses on 3 groups of metabolites: short-chain fatty acids, bile acids metabolites, and polyamines. We collated the findings of several studies on the transformation and production of these metabolites by gut microbes and explained their immunological roles. Specifically, we summarized the reports on changes in mucosal immune homeostasis represented by the Tregs and Th17 cells balance. The relationship between specific metabolites and diseases was also analyzed through latest studies. Thus, this review highlights microbial metabolites as the hidden treasure having potential diagnostic markers and therapeutic targets through a comprehensive understanding of the gut-immune interaction.
Collapse
Affiliation(s)
- Jong-Hwi Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jun-Soo Do
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Priyanka Velankanni
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
| | - Choong-Gu Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) School, University of Science and Technology, Seoul 02792, Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
- Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
18
|
Dugan B, Conway J, Duggal NA. Inflammaging as a target for healthy ageing. Age Ageing 2023; 52:7024516. [PMID: 36735849 DOI: 10.1093/ageing/afac328] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 11/11/2022] [Indexed: 02/05/2023] Open
Abstract
Life expectancy has been on the rise for the past few decades, but healthy life expectancy has not kept pace, leading to a global burden of age-associated disorders. Advancing age is accompanied by a chronic increase in basal systemic inflammation, termed inflammaging, contributing towards an increased risk of developing chronic diseases in old age. This article reviews the recent literature to formulate hypotheses regarding how age-associated inflammaging plays a crucial role in driving chronic diseases and ill health in older adults. Here, we discuss how non-pharmacological intervention strategies (diet, nutraceutical supplements, phytochemicals, physical activity, microbiome-based therapies) targeting inflammaging restore health in older adults. We also consider alternative existing pharmacological interventions (Caloric restriction mimetics, p38 mitogen-activated protein kinase inhibitors) and explore novel targets (senolytics) aimed at combating inflammaging and optimising the ageing process to increase healthy lifespan.
Collapse
Affiliation(s)
- Ben Dugan
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Jessica Conway
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Niharika A Duggal
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
19
|
Okamura T, Hamaguchi M, Hasegawa Y, Hashimoto Y, Majima S, Senmaru T, Ushigome E, Nakanishi N, Asano M, Yamazaki M, Sasano R, Nakanishi Y, Seno H, Takano H, Fukui M. Oral Exposure to Polystyrene Microplastics of Mice on a Normal or High-Fat Diet and Intestinal and Metabolic Outcomes. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:27006. [PMID: 36821708 PMCID: PMC9945580 DOI: 10.1289/ehp11072] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
BACKGROUND Microplastics (MPs) are small particles of plastic (≤5mm in diameter). In recent years, oral exposure to MPs in living organisms has been a cause of concern. Leaky gut syndrome (LGS), associated with a high-fat diet (HFD) in mice, can increase the entry of foreign substances into the body through the intestinal mucosa. OBJECTIVES We aimed to evaluate the pathophysiology of intestinal outcomes associated with consuming a high-fat diet and simultaneous intake of MPs, focusing on endocrine and metabolic systems. METHODS C57BL6/J mice were fed a normal diet (ND) or HFD with or without polystyrene MP for 4 wk to investigate differences in glucose tolerance, intestinal permeability, gut microbiota, as well as metabolites in serum, feces, and liver. RESULTS In comparison with HFD mice, mice fed the HFD with MPs had higher blood glucose, serum lipid concentrations, and nonalcoholic fatty liver disease (NAFLD) activity scores. Permeability and goblet cell count of the small intestine (SI) in HFD-fed mice were higher and lower, respectively, than in ND-fed mice. There was no obvious difference in the number of inflammatory cells in the SI lamina propria between mice fed the ND and mice fed the ND with MP, but there were more inflammatory cells and fewer anti-inflammatory cells in mice fed the HFD with MPs in comparison with mice fed the HFD without MPs. The expression of genes related to inflammation, long-chain fatty acid transporter, and Na+/glucose cotransporter was significantly higher in mice fed the HFD with MPs than in mice fed the HFD without MPs. Furthermore, the genus Desulfovibrio was significantly more abundant in the intestines of mice fed the HFD with MPs in comparison with mice fed the HFD without MPs. Muc2 gene expression was decreased when palmitic acid and microplastics were added to the murine intestinal epithelial cell line MODE-K cells, and Muc2 gene expression was increased when IL-22 was added. DISCUSSION Our findings suggest that in this study, MP induced metabolic disturbances, such as diabetes and NAFLD, only in mice fed a high-fat diet. These findings suggest that LGS might have been triggered by HFD, causing MPs to be deposited in the intestinal mucosa, resulting in inflammation of the intestinal mucosal intrinsic layer and thereby altering nutrient absorption. These results highlight the need for reducing oral exposure to MPs through remedial environmental measures to improve metabolic disturbance under high-fat diet conditions. https://doi.org/10.1289/EHP11072.
Collapse
Affiliation(s)
- Takuro Okamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Yuka Hasegawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Yoshitaka Hashimoto
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Saori Majima
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Takafumi Senmaru
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Mai Asano
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Masahiro Yamazaki
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | | | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohisa Takano
- Environmental Health Sciences, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
20
|
Hodgkinson K, El Abbar F, Dobranowski P, Manoogian J, Butcher J, Figeys D, Mack D, Stintzi A. Butyrate's role in human health and the current progress towards its clinical application to treat gastrointestinal disease. Clin Nutr 2023; 42:61-75. [PMID: 36502573 DOI: 10.1016/j.clnu.2022.10.024] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/17/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Butyrate is a key energy source for colonocytes and is produced by the gut microbiota through fermentation of dietary fiber. Butyrate is a histone deacetylase inhibitor and also signals through three G-protein coupled receptors. It is clear that butyrate has an important role in gastrointestinal health and that butyrate levels can impact both host and microbial functions that are intimately coupled with each other. Maintaining optimal butyrate levels improves gastrointestinal health in animal models by supporting colonocyte function, decreasing inflammation, maintaining the gut barrier, and promoting a healthy microbiome. Butyrate has also shown protective actions in the context of intestinal diseases such as inflammatory bowel disease, graft-versus-host disease of the gastrointestinal tract, and colon cancer, whereas lower levels of butyrate and/or the microbes which are responsible for producing this metabolite are associated with disease and poorer health outcomes. However, clinical efforts to increase butyrate levels in humans and reverse these negative outcomes have generated mixed results. This article discusses our current understanding of the molecular mechanisms of butyrate action with a focus on the gastrointestinal system, the links between host and microbial factors, and the efforts that are currently underway to apply the knowledge gained from the bench to bedside.
Collapse
Affiliation(s)
- Kendra Hodgkinson
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Faiha El Abbar
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Peter Dobranowski
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Juliana Manoogian
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - James Butcher
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daniel Figeys
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - David Mack
- Department of Paediatrics, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L1, Canada; Children's Hospital of Eastern Ontario Inflammatory Bowel Disease Centre and Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Alain Stintzi
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
21
|
Gut Microbial-Derived Metabolites as Immune Modulators of T Helper 17 and Regulatory T Cells. Int J Mol Sci 2023; 24:ijms24021806. [PMID: 36675320 PMCID: PMC9867388 DOI: 10.3390/ijms24021806] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The gut microbiota and its derived metabolites greatly impact the host immune system, both innate and adaptive responses. Gut dysbiosis and altered levels of microbiota-derived metabolites have been described in several immune-related and immune-mediated diseases such as intestinal bowel disease, multiple sclerosis, or colorectal cancer. Gut microbial-derived metabolites are synthesized from dietary compounds ingested by the host or host-produced metabolites, and additionally, some bacterial products can be synthesized de novo. In this review, we focus on the two first metabolites families including short-chain fatty acids, indole metabolites, polyamines, choline-derived compounds, and secondary bile acids. They all have been described as immunoregulatory molecules that specifically affect the adaptive immune system and T helper 17 and regulatory T cells. We discuss the mechanisms of action and the consequences in health and diseases related to these gut microbial-derived metabolites. Finally, we propose that the exogenous administration of these molecules or other compounds that bind to their immunoregulatory receptors in a homologous manner could be considered therapeutic approaches.
Collapse
|
22
|
Alam MZ, Maslanka JR, Abt MC. Immunological consequences of microbiome-based therapeutics. Front Immunol 2023; 13:1046472. [PMID: 36713364 PMCID: PMC9878555 DOI: 10.3389/fimmu.2022.1046472] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023] Open
Abstract
The complex network of microscopic organisms living on and within humans, collectively referred to as the microbiome, produce wide array of biologically active molecules that shape our health. Disruption of the microbiome is associated with susceptibility to a range of diseases such as cancer, diabetes, allergy, obesity, and infection. A new series of next-generation microbiome-based therapies are being developed to treat these diseases by transplanting bacteria or bacterial-derived byproducts into a diseased individual to reset the recipient's microbiome and restore health. Microbiome transplantation therapy is still in its early stages of being a routine treatment option and, with a few notable exceptions, has had limited success in clinical trials. In this review, we highlight the successes and challenges of implementing these therapies to treat disease with a focus on interactions between the immune system and microbiome-based therapeutics. The immune activation status of the microbiome transplant recipient prior to transplantation has an important role in supporting bacterial engraftment. Following engraftment, microbiome transplant derived signals can modulate immune function to ameliorate disease. As novel microbiome-based therapeutics are developed, consideration of how the transplants will interact with the immune system will be a key factor in determining whether the microbiome-based transplant elicits its intended therapeutic effect.
Collapse
Affiliation(s)
| | | | - Michael C. Abt
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
23
|
Limketkai BN, Hamideh M, Shah R, Sauk JS, Jaffe N. Dietary Patterns and Their Association With Symptoms Activity in Inflammatory Bowel Diseases. Inflamm Bowel Dis 2022; 28:1627-1636. [PMID: 35092268 DOI: 10.1093/ibd/izab335] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Existing studies on diet and inflammatory bowel disease (IBD) have largely focused on evaluating the effects of single nutrients or whole predesigned diets but not on evaluating the effects of diverse dietary patterns. This study applied unsupervised methods to identify dietary patterns of individuals with IBD and evaluated their association with symptoms activity. METHODS This retrospective study of adults with IBD collected current clinical data and typical diet recalled from the time when in clinical remission. Discrete dietary structures were defined by k-means clustering. Multivariable logistic regression evaluated the relationship between diet clusters and the presence of active symptoms, while adjusting for age, sex, disease duration, disease behavior, and medication use. RESULTS Of 691 participants, 36% had Crohn's disease (CD) and 64% had ulcerative colitis (UC) or IBD-unclassified. Five major dietary clusters were identified: 2 resembled a Western diet (WD) (WD1, WD2), 1 resembled a balanced diet, and 2 resembled a plant-based diet (PB) (PB1, PB2). Compared with WD1, PB2 was associated with lower odds of active symptoms for CD (odds ratio [OR], 0.32; 95% confidence interval [CI], 0.12-0.83) and UC (OR, 0.31; 95% CI, 0.15-0.62). PB1 was associated with lower odds of active symptoms for participants with UC (OR, 0.45; 95% CI, 0.23-0.90) but not for participants with CD (OR, 0.95; 95% CI, 0.36-2.51). CONCLUSIONS Diets with increased intake of fruits and vegetables, reduction of processed meats and refined carbohydrates, and preference of water for hydration were associated with lower risk of active symptoms with IBD, although increased intake of fruits and vegetables alone did not reduce risk of symptoms with CD.
Collapse
Affiliation(s)
- Berkeley N Limketkai
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Mohamed Hamideh
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rishabh Shah
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jenny S Sauk
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nancee Jaffe
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
24
|
KIANI AYSHAKARIM, BONETTI GABRIELE, DONATO KEVIN, BERTELLI MATTEO. Dietary supplements for intestinal inflammation. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2022; 63:E214-E220. [PMID: 36479492 PMCID: PMC9710413 DOI: 10.15167/2421-4248/jpmh2022.63.2s3.2763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Intestinal inflammation leads to various chronic diseases, collectively known as inflammatory bowel disease (IBD). IBD mainly affects the large intestine, but it can also affect the gastrointestinal tract as a whole. Its major symptoms are pain, diarrhea, and weight loss, and it is usually associated with deficiencies of both macro- and micronutrients. Unluckily, after some time the body develops resistance against the already available drugs: thus, many patients fail to maintain remission, which is achieved in less than 50% of cases. Diet is a major determinant of gut inflammation. An unbalanced diet can affect the gut microbiota and cause dysbiosis, which is related to a dysregulated host immune response. The Mediterranean Diet its renowned for its anti-inflammatory effects and for preventing dysbiosis. In order to improve management and treatment of intestinal inflammatory diseases, it should become common practice to integrate the patient's diet with dietary supplements with anti-inflammatory effects (probiotics, butyrate, phosphatidylcholine, lactoferrin, palmitoylethanolamide, silymarin, and omega 3), which maintain the stability of the intestinal microbial cohort and strengthen the mucosal barrier, thus preventing or soothing IBD symptoms. Dietary supplements may help fight the high costs, the adverse side effects, and the recurrent relapses typical of drug use.
Collapse
Affiliation(s)
| | - GABRIELE BONETTI
- MAGI’S LAB, Rovereto (TN), Italy
- Correspondence: Gabriele Bonetti, MAGI’S LAB, Rovereto (TN), 38068, Italy. E-mail:
| | | | - MATTEO BERTELLI
- MAGI Euregio, Bolzano, Italy
- MAGI’S LAB, Rovereto (TN), Italy
- MAGISNAT, Peachtree Corners (GA), USA
| |
Collapse
|
25
|
Effect of DSS-Induced Ulcerative Colitis and Butyrate on the Cytochrome P450 2A5: Contribution of the Microbiome. Int J Mol Sci 2022; 23:ijms231911627. [PMID: 36232929 PMCID: PMC9569822 DOI: 10.3390/ijms231911627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
Several studies have indicated the beneficial anti-inflammatory effect of butyrate in inflammatory bowel disease (IBD) therapy implying attempts to increase butyrate production in the gut through orally administered dietary supplementation. Through the gut-liver axis, however, butyrate may reach directly the liver and influence the drug-metabolizing ability of hepatic enzymes, and, indirectly, also the outcome of applied pharmacotherapy. The focus of our study was on the liver microsomal cytochrome P450 (CYP) 2A5, which is a mouse orthologue of human CYP2A6 responsible for metabolism of metronidazole, an antibiotic used to treat IBD. Our findings revealed that specific pathogen-free (SPF) and germ-free (GF) mice with dextran sulfate sodium (DSS)-induced colitis varied markedly in enzyme activity of CYP2A and responded differently to butyrate pre-treatment. A significant decrease (to 50%) of the CYP2A activity was observed in SPF mice with colitis; however, an administration of butyrate prior to DSS reversed this inhibition effect. This phenomenon was not observed in GF mice. The results highlight an important role of gut microbiota in the regulation of CYP2A under inflammatory conditions. Due to the role of CYP2A in metronidazole metabolism, this phenomenon may have an impact on the IBD therapy. Butyrate administration, hence, brings promising therapeutic potential for improving symptoms of gut inflammation; however, possible interactions with drug metabolism need to be further studied.
Collapse
|
26
|
Spivak I, Fluhr L, Elinav E. Local and systemic effects of microbiome‐derived metabolites. EMBO Rep 2022; 23:e55664. [PMID: 36031866 PMCID: PMC9535759 DOI: 10.15252/embr.202255664] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 12/12/2022] Open
Abstract
Commensal microbes form distinct ecosystems within their mammalian hosts, collectively termed microbiomes. These indigenous microbial communities broadly expand the genomic and functional repertoire of their host and contribute to the formation of a “meta‐organism.” Importantly, microbiomes exert numerous biochemical reactions synthesizing or modifying multiple bioactive small molecules termed metabolites, which impact their host's physiology in a variety of contexts. Identifying and understanding molecular mechanisms of metabolite–host interactions, and how their disrupted signaling can contribute to diseases, may enable their therapeutic application, a modality termed “postbiotic” therapy. In this review, we highlight key examples of effects of bioactive microbe‐associated metabolites on local, systemic, and immune environments, and discuss how these may impact mammalian physiology and associated disorders. We outline the challenges and perspectives in understanding the potential activity and function of this plethora of microbially associated small molecules as well as possibilities to harness them toward the promotion of personalized precision therapeutic interventions.
Collapse
Affiliation(s)
- Igor Spivak
- Systems Immunology Department Weizmann Institute of Science Rehovot Israel
- Medical Clinic III University Hospital Aachen Aachen Germany
| | - Leviel Fluhr
- Systems Immunology Department Weizmann Institute of Science Rehovot Israel
| | - Eran Elinav
- Systems Immunology Department Weizmann Institute of Science Rehovot Israel
- Microbiome & Cancer Division, DKFZ Heidelberg Germany
| |
Collapse
|
27
|
Pravda J. Evidence-based pathogenesis and treatment of ulcerative colitis: A causal role for colonic epithelial hydrogen peroxide. World J Gastroenterol 2022; 28:4263-4298. [PMID: 36159014 PMCID: PMC9453768 DOI: 10.3748/wjg.v28.i31.4263] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/19/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
In this comprehensive evidence-based analysis of ulcerative colitis (UC), a causal role is identified for colonic epithelial hydrogen peroxide (H2O2) in both the pathogenesis and relapse of this debilitating inflammatory bowel disease. Studies have shown that H2O2 production is significantly increased in the non-inflamed colonic epithelium of individuals with UC. H2O2 is a powerful neutrophilic chemotactic agent that can diffuse through colonic epithelial cell membranes creating an interstitial chemotactic molecular “trail” that attracts adjacent intravascular neutrophils into the colonic epithelium leading to mucosal inflammation and UC. A novel therapy aimed at removing the inappropriate H2O2 mediated chemotactic signal has been highly effective in achieving complete histologic resolution of colitis in patients experiencing refractory disease with at least one (biopsy-proven) histologic remission lasting 14 years to date. The evidence implies that therapeutic intervention to prevent the re-establishment of a pathologic H2O2 mediated chemotactic signaling gradient will indefinitely preclude neutrophilic migration into the colonic epithelium constituting a functional cure for this disease. Cumulative data indicate that individuals with UC have normal immune systems and current treatment guidelines calling for the suppression of the immune response based on the belief that UC is caused by an underlying immune dysfunction are not supported by the evidence and may cause serious adverse effects. It is the aim of this paper to present experimental and clinical evidence that identifies H2O2 produced by the colonic epithelium as the causal agent in the pathogenesis of UC. A detailed explanation of a novel therapeutic intervention to normalize colonic H2O2, its rationale, components, and formulation is also provided.
Collapse
Affiliation(s)
- Jay Pravda
- Disease Pathogenesis, Inflammatory Disease Research Centre, Palm Beach Gardens, FL 33410, United States
| |
Collapse
|
28
|
Rudiansyah M, Abdalkareem Jasim S, S Azizov B, Samusenkov V, Kamal Abdelbasset W, Yasin G, Mohammad HJ, Jawad MA, Mahmudiono T, Hosseini-Fard SR, Mirzaei R, Karampoor S. The emerging microbiome-based approaches to IBD therapy: From SCFAs to urolithin A. J Dig Dis 2022; 23:412-434. [PMID: 36178158 DOI: 10.1111/1751-2980.13131] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel disease (IBD) is a group of chronic gastrointestinal inflammatory conditions which can be life-threatening, affecting both children and adults. Crohn's disease and ulcerative colitis are the two main forms of IBD. The pathogenesis of IBD is complex and involves genetic background, environmental factors, alteration in gut microbiota, aberrant immune responses (innate and adaptive), and their interactions, all of which provide clues to the identification of innovative diagnostic or prognostic biomarkers and the development of novel treatments. Gut microbiota provide significant benefits to its host, most notably via maintaining immunological homeostasis. Furthermore, changes in gut microbial populations may promote immunological dysregulation, resulting in autoimmune diseases, including IBD. Investigating the interaction between gut microbiota and immune system of the host may lead to a better understanding of the pathophysiology of IBD as well as the development of innovative immune- or microbe-based therapeutics. In this review we summarized the most recent findings on innovative therapeutics for IBD, including microbiome-based therapies such as fecal microbiota transplantation, probiotics, live biotherapeutic products, short-chain fatty acids, bile acids, and urolithin A.
Collapse
Affiliation(s)
- Mohammad Rudiansyah
- Division of Nephrology & Hypertension, Department of Internal Medicine, Faculty of Medicine, Universitas Lambung Mangkurat, Ulin Hospital, Banjarmasin, Indonesia
| | - Saade Abdalkareem Jasim
- Al-Maarif University College Medical Laboratory Techniques Department Al-Anbar-Ramadi, Ramadi, Iraq
| | - Bakhadir S Azizov
- Department of Therapeutic Disciplines No.1, Tashkent State Dental Institute, Tashkent, Uzbekistan
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Ghulam Yasin
- Department of Botany University of Bahauddin Zakariya University, Multan, Pakistan
| | | | | | - Trias Mahmudiono
- Department of Nutrition Faculty of Public Health Universitas, Airlangga, Indonesia
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Jourova L, Satka S, Frybortova V, Zapletalova I, Anzenbacher P, Anzenbacherova E, Hermanova PP, Drabonova B, Srutkova D, Kozakova H, Hudcovic T. Butyrate Treatment of DSS-Induced Ulcerative Colitis Affects the Hepatic Drug Metabolism in Mice. Front Pharmacol 2022; 13:936013. [PMID: 35928257 PMCID: PMC9343805 DOI: 10.3389/fphar.2022.936013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/15/2022] [Indexed: 12/18/2022] Open
Abstract
The development of inflammatory bowel disease (IBD) is associated with alterations in the gut microbiota. There is currently no universal treatment for this disease, thus emphasizing the importance of developing innovative therapeutic approaches. Gut microbiome-derived metabolite butyrate with its well-known anti-inflammatory effect in the gut is a promising candidate. Due to increased intestinal permeability during IBD, butyrate may also reach the liver and influence liver physiology, including hepatic drug metabolism. To get an insight into this reason, the aim of this study was set to clarify not only the protective effects of the sodium butyrate (SB) administration on colonic inflammation but also the effects of SB on hepatic drug metabolism in experimental colitis induced by dextran sodium sulfate (DSS) in mice. It has been shown here that the butyrate pre-treatment can alleviate gut inflammation and reduce the leakiness of colonic epithelium by restoration of the assembly of tight-junction protein Zonula occludens-1 (ZO-1) in mice with DSS-induced colitis. In this article, butyrate along with inflammation has also been shown to affect the expression and enzyme activity of selected cytochromes P450 (CYPs) in the liver of mice. In this respect, CYP3A enzymes may be very sensitive to gut microbiome-targeted interventions, as significant changes in CYP3A expression and activity in response to DSS-induced colitis and/or butyrate treatment have also been observed. With regard to medications used in IBD and microbiota-targeted therapeutic approaches, it is important to deepen our knowledge of the effect of gut inflammation, and therapeutic interventions were followed concerning the ability of the organism to metabolize drugs. This gut–liver axis, mediated through inflammation as well as microbiome-derived metabolites, may affect the response to IBD therapy.
Collapse
Affiliation(s)
- Lenka Jourova
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
- *Correspondence: Lenka Jourova,
| | - Stefan Satka
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Veronika Frybortova
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Iveta Zapletalova
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Pavel Anzenbacher
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Eva Anzenbacherova
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Petra Petr Hermanova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Barbora Drabonova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Hana Kozakova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Tomas Hudcovic
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| |
Collapse
|
30
|
Weber-Stiehl S, Järke L, Castrillón-Betancur JC, Gilbert F, Sommer F. Mitochondrial Function and Microbial Metabolites as Central Regulators of Intestinal Immune Responses and Cancer. Front Microbiol 2022; 13:919424. [PMID: 35847099 PMCID: PMC9277123 DOI: 10.3389/fmicb.2022.919424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/24/2022] [Indexed: 11/24/2022] Open
Abstract
Energy and anabolic metabolism are essential for normal cellular homeostasis but also play an important role in regulating immune responses and cancer development as active immune and cancer cells show an altered metabolic profile. Mitochondria take a prominent position in these metabolic reactions. First, most key energetic reactions take place within or in conjunction with mitochondria. Second, mitochondria react to internal cues from within the cell but also to external cues originating from the microbiota, a vast diversity of associated microorganisms. The impact of the microbiota on host physiology has been largely investigated in the last decade revealing that the microbiota contributes to the extraction of calories from the diet, energy metabolism, maturation of the immune system and cellular differentiation. Thus, changes in the microbiota termed dysbiosis have been associated with disease development including metabolic diseases, inflammation and cancer. Targeting the microbiota to modulate interactions with the mitochondria and cellular metabolism to delay or inhibit disease development and pathogenesis appears an attractive therapeutic approach. Here, we summarize recent advances in developing the therapeutic potential of microbiota-mitochondria interactions for inflammation and cancer.
Collapse
|
31
|
Dietary polysaccharides from guavira pomace, a co-product from the fruit pulp industry, display therapeutic application in gut disorders. Food Res Int 2022; 156:111291. [DOI: 10.1016/j.foodres.2022.111291] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022]
|
32
|
Intestinal Microbiota-Derived Short Chain Fatty Acids in Host Health and Disease. Nutrients 2022; 14:nu14091977. [PMID: 35565943 PMCID: PMC9105144 DOI: 10.3390/nu14091977] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023] Open
Abstract
Intestinal microbiota has its role as an important component of human physiology. It produces metabolites that module key functions to establish a symbiotic crosstalk with their host. Among them, short chain fatty acids (SCFAs), produced by intestinal bacteria during the fermentation of partially and non-digestible polysaccharides, play key roles in regulating colon physiology and changing intestinal environment. Recent research has found that SCFAs not only influence the signal transduction pathway in the gut, but they also reach tissues and organs outside of the gut, through their circulation in the blood. Growing evidence highlights the importance of SCFAs level in influencing health maintenance and disease development. SCFAs are probably involved in the management of host health in a complicated (positive or negative) way. Here, we review the current understanding of SCFAs effects on host physiology and discuss the potential prevention and therapeutics of SCFAs in a variety of disorders. It provides a systematic theoretical basis for the study of mechanisms and precise intake level of SCFAs to promote human health.
Collapse
|
33
|
Conway J, Certo M, Lord JM, Mauro C, Duggal NA. Understanding the role of host metabolites in the induction of immune senescence: Future strategies for keeping the ageing population healthy. Br J Pharmacol 2022; 179:1808-1824. [PMID: 34435354 DOI: 10.1111/bph.15671] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
Advancing age is accompanied by significant remodelling of the immune system, termed immune senescence, and increased systemic inflammation, termed inflammageing, both of which contribute towards an increased risk of developing chronic diseases in old age. Age-associated alterations in metabolic homeostasis have been linked with changes in a range of physiological functions, but their effects on immune senescence remains poorly understood. In this article, we review the recent literature to formulate hypotheses as to how an age-associated dysfunctional metabolism, driven by an accumulation of key host metabolites (saturated fatty acids, cholesterol, ceramides and lactate) and loss of other metabolites (glutamine, tryptophan and short-chain fatty acids), might play a role in driving immune senescence and inflammageing, ultimately leading to diseases of old age. We also highlight the potential use of metabolic immunotherapeutic strategies targeting these processes in counteracting immune senescence and restoring immune homeostasis in older adults. LINKED ARTICLES: This article is part of a themed issue on Inflammation, Repair and Ageing. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.9/issuetoc.
Collapse
Affiliation(s)
- Jessica Conway
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Janet M Lord
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham and University of Birmingham, Birmingham, UK
| | - Claudio Mauro
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Niharika A Duggal
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
34
|
The role of short-chain fatty acids in Clostridioides difficile infection: A review. Anaerobe 2022; 75:102585. [DOI: 10.1016/j.anaerobe.2022.102585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/20/2022]
|
35
|
O'Riordan KJ, Collins MK, Moloney GM, Knox EG, Aburto MR, Fülling C, Morley SJ, Clarke G, Schellekens H, Cryan JF. Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol 2022; 546:111572. [PMID: 35066114 DOI: 10.1016/j.mce.2022.111572] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 02/08/2023]
Abstract
The role of the intestinal microbiota as a regulator of gut-brain axis signalling has risen to prominence in recent years. Understanding the relationship between the gut microbiota, the metabolites it produces, and the brain will be critical for the subsequent development of new therapeutic approaches, including the identification of novel psychobiotics. A key focus in this regard have been the short-chain fatty acids (SCFAs) produced by bacterial fermentation of dietary fibre, which include butyrate, acetate, and propionate. Ongoing research is focused on the entry of SCFAs into systemic circulation from the gut lumen, their migration to cerebral circulation and across the blood brain barrier, and their potential to exert acute and chronic effects on brain structure and function. This review aims to discuss our current mechanistic understanding of the direct and indirect influence that SCFAs have on brain function, behaviour and physiology, which will inform future microbiota-targeted interventions for brain disorders.
Collapse
Affiliation(s)
| | - Michael K Collins
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Gerard M Moloney
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Emily G Knox
- APC Microbiome Ireland, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland
| | - María R Aburto
- APC Microbiome Ireland, University College Cork, Ireland
| | | | - Shane J Morley
- APC Microbiome Ireland, University College Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland.
| |
Collapse
|
36
|
Pan L, Fu T, Cheng H, Mi J, Shang Q, Yu G. Polysaccharide from edible alga Gloiopeltis furcata attenuates intestinal mucosal damage by therapeutically remodeling the interactions between gut microbiota and mucin O-glycans. Carbohydr Polym 2022; 278:118921. [PMID: 34973740 DOI: 10.1016/j.carbpol.2021.118921] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/11/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
Gloiopeltis furcata is an edible alga that has long been consumed in China. However, the bioactive polysaccharides from G. furcata have been largely unexplored. Here, we show for the first time that a sulfated polysaccharide from G. furcata (SAO) could improve the integrity of the colonic epithelial layer and protect against dextran sulfate sodium-induced intestinal mucosal damage. Mechanistically, SAO attenuated colonic mucosal damage by therapeutically remodeling the interactions between gut microbiota and mucin O-glycans. Specifically, SAO increased the proportions of complex long-chain mucin O-glycans in the epithelial layer with two terminal N-acetylneuraminic acid residues and promoted the growth of probiotic bacteria including Roseburia spp. and Muribaculaceae. Altogether, our study demonstrates a novel application of SAO for the treatment of inflammatory bowel disease-associated mucosal damage and forms the basis to understand the therapeutic effects of natural polysaccharides from the perspective of symbiotic interactions between host mucin O-glycome and gut microbiome.
Collapse
Affiliation(s)
- Lin Pan
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Tianyu Fu
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Hao Cheng
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jianchen Mi
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Qingsen Shang
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Qingdao Marine Biomedical Research Institute, Qingdao 266071, China.
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China.
| |
Collapse
|
37
|
Islam MR, Arthur S, Haynes J, Butts MR, Nepal N, Sundaram U. The Role of Gut Microbiota and Metabolites in Obesity-Associated Chronic Gastrointestinal Disorders. Nutrients 2022; 14:624. [PMID: 35276983 PMCID: PMC8838694 DOI: 10.3390/nu14030624] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/13/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
The gut microbiota is a complex community of microorganisms that has become a new focus of attention due to its association with numerous human diseases. Research over the last few decades has shown that the gut microbiota plays a considerable role in regulating intestinal homeostasis, and disruption to the microbial community has been linked to chronic disease conditions such as inflammatory bowel disease (IBD), colorectal cancer (CRC), and obesity. Obesity has become a global pandemic, and its prevalence is increasing worldwide mostly in Western countries due to a sedentary lifestyle and consumption of high-fat/high-sugar diets. Obesity-mediated gut microbiota alterations have been associated with the development of IBD and IBD-induced CRC. This review highlights how obesity-associated dysbiosis can lead to the pathogenesis of IBD and CRC with a special focus on mechanisms of altered absorption of short-chain fatty acids (SCFAs).
Collapse
Affiliation(s)
| | | | | | | | | | - Uma Sundaram
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (M.R.I.); (S.A.); (J.H.); (M.R.B.); (N.N.)
| |
Collapse
|
38
|
Xiong W, Devkota L, Zhang B, Muir J, Dhital S. Intact cells: “Nutritional capsules” in plant foods. Compr Rev Food Sci Food Saf 2022; 21:1198-1217. [DOI: 10.1111/1541-4337.12904] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/23/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Weiyan Xiong
- Department of Chemical and Biological Engineering Monash University Clayton Campus, VIC 3800 Australia
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety South China University of Technology Guangzhou Guangdong P. R. China
| | - Lavaraj Devkota
- Department of Chemical and Biological Engineering Monash University Clayton Campus, VIC 3800 Australia
| | - Bin Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety South China University of Technology Guangzhou Guangdong P. R. China
| | - Jane Muir
- Department of Gastroenterology Central Clinical School, Monash University Melbourne Victoria Australia
| | - Sushil Dhital
- Department of Chemical and Biological Engineering Monash University Clayton Campus, VIC 3800 Australia
| |
Collapse
|
39
|
Gu X, Sim JX, Lee WL, Cui L, Chan YF, Chang ED, Teh YE, Zhang AN, Armas F, Chandra F, Chen H, Zhao S, Lee Z, Thompson JR, Ooi EE, Low JG, Alm EJ, Kalimuddin S. Gut Ruminococcaceae levels at baseline correlate with risk of antibiotic-associated diarrhea. iScience 2022; 25:103644. [PMID: 35005566 PMCID: PMC8718891 DOI: 10.1016/j.isci.2021.103644] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) affects a significant proportion of patients receiving antibiotics. We sought to understand if differences in the gut microbiome would influence the development of AAD. We administered a 3-day course of amoxicillin-clavulanate to 30 healthy adult volunteers, and analyzed their stool microbiome, using 16S rRNA gene sequencing, at baseline and up to 4 weeks post antibiotic administration. Lower levels of gut Ruminococcaceae were significantly and consistently observed from baseline until day 7 in participants who developed AAD. Overall, participants who developed AAD experienced a greater decrease in microbial diversity. The probability of AAD could be predicted based on qPCR-derived levels of Faecalibacterium prausnitzii at baseline. Our findings suggest that a lack of gut Ruminococcaceae influences development of AAD. Quantification of F. prausnitzii in stool prior to antibiotic administration may help identify patients at risk of AAD, and aid clinicians in devising individualized treatment regimens to minimize such adverse effects.
Collapse
Affiliation(s)
- Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Jean X.Y. Sim
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Liang Cui
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yvonne F.Z. Chan
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Ega Danu Chang
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yii Ean Teh
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - An-Ni Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Shijie Zhao
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Zhanyi Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Janelle R. Thompson
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Asian School of the Environment, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, Singapore 117549, Singapore
| | - Jenny G. Low
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
| | - Eric J. Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Building E25-321, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
40
|
Chen W, Liu D, Ren C, Su X, Wong CK, Yang R. A Special Network Comprised of Macrophages, Epithelial Cells, and Gut Microbiota for Gut Homeostasis. Cells 2022; 11:cells11020307. [PMID: 35053422 PMCID: PMC8774616 DOI: 10.3390/cells11020307] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
A number of gut epithelial cells derived immunological factors such as cytokines and chemokines, which are stimulated by the gut microbiota, can regulate host immune responses to maintain a well-balance between gut microbes and host immune system. Multiple specialized immune cell populations, such as macrophages, dendritic cells (DCs), innate lymphoid cells, and T regulatory (Treg) cells, can communicate with intestinal epithelial cells (IEC) and/or the gut microbiota bi-directionally. The gut microbiota contributes to the differentiation and function of resident macrophages. Situated at the interface between the gut commensals and macrophages, the gut epithelium is crucial for gut homeostasis in microbial recognition, signaling transformation, and immune interactions, apart from being a physical barrier. Thus, three distinct but interactive components—macrophages, microbiota, and IEC—can form a network for the delicate and dynamic regulation of intestinal homeostasis. In this review, we will discuss the crucial features of gut microbiota, macrophages, and IEC. We will also summarize recent advances in understanding the cooperative and dynamic interactions among the gut microbiota, gut macrophages, and IEC, which constitute a special network for gut homeostasis.
Collapse
Affiliation(s)
- Wei Chen
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Dan Liu
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Changhao Ren
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Xiaomin Su
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
| | - Chun-Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Rongcun Yang
- Department of Immunology, School of Medicine, Nankai University, Tianjin 300071, China; (W.C.); (D.L.); (C.R.); (X.S.)
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Correspondence:
| |
Collapse
|
41
|
Kang S, You HJ, Ju Y, Kim HJ, Jeong YJ, Johnston TV, Ji GE, Ku S, Park MS. Butyl-fructooligosaccharides modulate gut microbiota in healthy mice and ameliorate ulcerative colitis in a DSS-induced model. Food Funct 2022; 13:1834-1845. [DOI: 10.1039/d1fo03337a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Butyl-fructooligosaccharides (B-FOSs) are newly synthesized prebiotics composed of short-chain FOS (GF2, 1-kestose; GF3, nystose; GF4, fructofuranosyl-nystose; GF5, 1-F-(1-b-D-fructofuranosyl)-2-nystose) bound with one or two butyric groups by ester bonds. Previous in...
Collapse
|
42
|
Maioli TU, Borras-Nogues E, Torres L, Barbosa SC, Martins VD, Langella P, Azevedo VA, Chatel JM. Possible Benefits of Faecalibacterium prausnitzii for Obesity-Associated Gut Disorders. Front Pharmacol 2021; 12:740636. [PMID: 34925006 PMCID: PMC8677946 DOI: 10.3389/fphar.2021.740636] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
Metabolic disorders are an increasing concern in the industrialized world. Current research has shown a direct link between the composition of the gut microbiota and the pathogenesis of obesity and diabetes. In only a few weeks, an obesity-inducing diet can lead to increased gut permeability and microbial dysbiosis, which contributes to chronic inflammation in the gut and adipose tissues, and to the development of insulin resistance. In this review, we examine the interplay between gut inflammation, insulin resistance, and the gut microbiota, and discuss how some probiotic species can be used to modulate gut homeostasis. We focus primarily on Faecalibacterium prausnitzii, a highly abundant butyrate-producing bacterium that has been proposed both as a biomarker for the development of different gut pathologies and as a potential treatment due to its production of anti-inflammatory metabolites.
Collapse
Affiliation(s)
- Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Université Paris Saclay, INRAE, AgroParisTech, Micalis, Jouy-en-Josas, France
| | | | - Licia Torres
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sara Candida Barbosa
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vinicius Dantas Martins
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Philippe Langella
- Université Paris Saclay, INRAE, AgroParisTech, Micalis, Jouy-en-Josas, France
| | - Vasco Ariston Azevedo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jean-Marc Chatel
- Université Paris Saclay, INRAE, AgroParisTech, Micalis, Jouy-en-Josas, France
| |
Collapse
|
43
|
Martyniak A, Medyńska-Przęczek A, Wędrychowicz A, Skoczeń S, Tomasik PJ. Prebiotics, Probiotics, Synbiotics, Paraprobiotics and Postbiotic Compounds in IBD. Biomolecules 2021; 11:biom11121903. [PMID: 34944546 PMCID: PMC8699341 DOI: 10.3390/biom11121903] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing incidence of inflammatory bowel diseases (IBD) and the increasing severity of the course of these diseases create the need for developing new methods of therapy. The gut microbiome is extensively studied as a factor influencing the development and course of IBD. The composition of intestinal microbiota can be relatively easily modified by diet (i.e., prebiotics, mainly dietary fibers) and bacterial supplementation using beneficial bacteria strains called probiotics. Additionally, the effects of the improved microbiome could be enhanced or gained by using paraprobiotics (non-viable, inactivated bacteria or their components) and/or postbiotics (products of bacterial metabolism or equal synthetic products that beneficially modulate immunological response and inflammation). This study summarizes the recent works on prebiotics, probiotics, synbiotics (products merging pre- and probiotics), paraprobiotics and postbiotics in IBD.
Collapse
Affiliation(s)
- Adrian Martyniak
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland;
| | - Aleksandra Medyńska-Przęczek
- Department of Paediatrics, Gastroenterology and Nutrition, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.-P.); (A.W.)
| | - Andrzej Wędrychowicz
- Department of Paediatrics, Gastroenterology and Nutrition, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.-P.); (A.W.)
| | - Szymon Skoczeń
- Department of Pediatric Oncology and Hematology, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland;
| | - Przemysław J. Tomasik
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland;
- Correspondence:
| |
Collapse
|
44
|
Hinrichsen F, Hamm J, Westermann M, Schröder L, Shima K, Mishra N, Walker A, Sommer N, Klischies K, Prasse D, Zimmermann J, Kaiser S, Bordoni D, Fazio A, Marinos G, Laue G, Imm S, Tremaroli V, Basic M, Häsler R, Schmitz RA, Krautwald S, Wolf A, Stecher B, Schmitt-Kopplin P, Kaleta C, Rupp J, Bäckhed F, Rosenstiel P, Sommer F. Microbial regulation of hexokinase 2 links mitochondrial metabolism and cell death in colitis. Cell Metab 2021; 33:2355-2366.e8. [PMID: 34847376 DOI: 10.1016/j.cmet.2021.11.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/07/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022]
Abstract
Hexokinases (HK) catalyze the first step of glycolysis limiting its pace. HK2 is highly expressed in gut epithelium, contributes to immune responses, and is upregulated during inflammation. We examined the microbial regulation of HK2 and its impact on inflammation using mice lacking HK2 in intestinal epithelial cells (Hk2ΔIEC). Hk2ΔIEC mice were less susceptible to acute colitis. Analyzing the epithelial transcriptome from Hk2ΔIEC mice during colitis and using HK2-deficient intestinal organoids and Caco-2 cells revealed reduced mitochondrial respiration and epithelial cell death in the absence of HK2. The microbiota strongly regulated HK2 expression and activity. The microbially derived short-chain fatty acid (SCFA) butyrate repressed HK2 expression via histone deacetylase 8 (HDAC8) and reduced mitochondrial respiration in wild-type but not in HK2-deficient Caco-2 cells. Butyrate supplementation protected wild-type but not Hk2ΔIEC mice from colitis. Our findings define a mechanism how butyrate promotes intestinal homeostasis and suggest targeted HK2-inhibition as therapeutic avenue for inflammation.
Collapse
Affiliation(s)
- Finn Hinrichsen
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Jacob Hamm
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Magdalena Westermann
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Lena Schröder
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany
| | - Neha Mishra
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Centre for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Nina Sommer
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Kenneth Klischies
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Daniela Prasse
- Institute of General Microbiology, University of Kiel, 24118 Kiel, Germany
| | | | - Sina Kaiser
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Dora Bordoni
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Antonella Fazio
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Georgios Marinos
- Institute of Experimental Medicine, University of Kiel, 24105 Kiel, Germany
| | - Georg Laue
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Simon Imm
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany; Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Ruth A Schmitz
- Institute of General Microbiology, University of Kiel, 24118 Kiel, Germany
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Andrea Wolf
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany; German Center for Infection Research (DZIF), partner site LMU Munich, Munich Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Centre for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Christoph Kaleta
- Institute of Experimental Medicine, University of Kiel, 24105 Kiel, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany.
| |
Collapse
|
45
|
Xu E, Chen C, Fu J, Zhu L, Shu J, Jin M, Wang Y, Zong X. Dietary fatty acids in gut health: Absorption, metabolism and function. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:1337-1344. [PMID: 34786506 PMCID: PMC8570925 DOI: 10.1016/j.aninu.2021.09.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 12/23/2022]
Abstract
In biological responses, fatty acids (FA) are absorbed and metabolized in the form of substrates for energy production. The molecular structures (number of double bonds and chain length) and composition of dietary FA impact digestion, absorption and metabolism, and the biological roles of FA. Recently, increasing evidence indicates that FA are essentially utilized as an energy source and are signaling molecules that exert physiological activity of gut microbiota and immune responses. In addition, FA could serve as natural ligands for orphan G protein-coupled receptors (GPCR), also called free fatty acid receptors (FFAR), which intertwine metabolic and immune systems via multiple mechanisms. The present review explores the recent findings on FA absorption and its impact on gut health, particularly addressing the mechanism by which dietary FA potentially influences intestinal microbiota and epithelial functions. Also, this work attempts to uncover research ideas for devising future strategies for manipulating the composition of dietary FA to regulate gut health and support a normal immune system for metabolic and immune disorders.
Collapse
Affiliation(s)
- E. Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Aniaml Science, Guizhou University, 550025 Guiyang, China
| | - Chao Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Aniaml Science, Guizhou University, 550025 Guiyang, China
| | - Jie Fu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Luoyi Zhu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Junlan Shu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Aniaml Science, Guizhou University, 550025 Guiyang, China
| | - Mingliang Jin
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| | - Xin Zong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China
| |
Collapse
|
46
|
Lee JG, Lee J, Lee AR, Jo SV, Park CH, Han DS, Eun CS. Impact of short-chain fatty acid supplementation on gut inflammation and microbiota composition in a murine colitis model. J Nutr Biochem 2021; 101:108926. [PMID: 34848335 DOI: 10.1016/j.jnutbio.2021.108926] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/15/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023]
Abstract
Short-chain fatty acids (SCFAs) play a pivotal role in maintaining intestinal homeostasis. We aimed to investigate the effects of SCFA supplementation on gut inflammation and microbiota composition in a murine colitis model. Mice were fed with sodium butyrate or a mixture of SCFAs in the drinking water for 2 weeks, followed by 2% dextran sulfate sodium (DSS) for 7 d. After euthanasia, mouse colons were extracted to examine histological findings. Flow cytometry of the mouse colon tissues was performed to assess T cell differentiation. Changes in gut microbiota were assessed by high-throughput sequencing of the mouse feces. There were no significant differences in weight change, colonic length, or histologic inflammation score between the DSS, butyrate, and SCFA mix groups. However, flow cytometry revealed that both the expression of CD4+Foxp3+ regulatory T cells and of IL-17-producing T cells were increased in the butyrate and SCFA mix groups. Microbial compositions of the butyrate and SCFA mix groups were significantly different from those of the control and DSS groups in principal coordinate analysis. Relative abundances of the phyla Verrucomicrobia and Proteobacteria, species Akkermansia muciniphila and Escherichia fergusonii were increased in the butyrate and SCFA mix groups. Genera Roseburia and Lactobacillus showed a negative correlation with the degree of colitis, whereas genera Escherichia and Mucispirillum showed a positive correlation. SCFA supplementation did not result in a significant reduction in colon inflammation, but it promoted both regulatory T cell and IL-17-producing T cell expression, and increased both protective and aggressive gut microbiota.
Collapse
Affiliation(s)
- Jae Gon Lee
- Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea
| | - Jiyoung Lee
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - A-Reum Lee
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Su Vin Jo
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Chan Hyuk Park
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Dong Soo Han
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Chang Soo Eun
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea.
| |
Collapse
|
47
|
Siddiqui MT, Cresci GAM. The Immunomodulatory Functions of Butyrate. J Inflamm Res 2021; 14:6025-6041. [PMID: 34819742 PMCID: PMC8608412 DOI: 10.2147/jir.s300989] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal (GI) system contains many different types of immune cells, making it a key immune organ system in the human body. In the last decade, our knowledge has substantially expanded regarding our understanding of the gut microbiome and its complex interaction with the gut immune system. Short chain fatty acids (SCFA), and specifically butyrate, play an important role in mediating the effects of the gut microbiome on local and systemic immunity. Gut microbial alterations and depletion of luminal butyrate have been well documented in the literature for a number of systemic and GI inflammatory disorders. Although a substantial knowledge gap exists requiring the need for further investigations to determine cause and effect, there is heightened interest in developing immunomodulatory therapies by means of reprogramming of gut microbiome or by supplementing its beneficial metabolites, such as butyrate. In the current review, we discuss the role of endogenous butyrate in the inflammatory response and maintaining immune homeostasis within the intestine. We also present the experimental models and human studies which explore therapeutic potential of butyrate supplementation in inflammatory conditions associated with butyrate depletion.
Collapse
Affiliation(s)
- Mohamed Tausif Siddiqui
- Department of Gastroenterology, Hepatology and Human Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Gail A M Cresci
- Department of Gastroenterology, Hepatology and Human Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Pediatric Gastroenterology, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
48
|
Lluansí A, Llirós M, Oliver L, Bahí A, Elias-Masiques N, Gonzalez M, Benejam P, Cueva E, Termes M, Ramió-Pujol S, Malagón M, Amoedo J, Serrano M, Busquets D, Torreabla L, Sabat M, Buxó M, Cambra M, Serra-Pagès M, Delgado-Aros S, García-Gil LJ, Elias I, Aldeguer X. In vitro Prebiotic Effect of Bread-Making Process in Inflammatory Bowel Disease Microbiome. Front Microbiol 2021; 12:716307. [PMID: 34707578 PMCID: PMC8543021 DOI: 10.3389/fmicb.2021.716307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD), including its two main categories (Crohn's disease and ulcerative colitis), has been linked both to gut microbiota and to diet. Bread is a daily food that has a potential capacity as a prebiotic. Our aim was to evaluate different bread-making processes and their effect on fecal colonic microbiota in IBD patients. The microbial composition of several sourdoughs and dough samples was analyzed by high-throughput sequencing of 16S and 18S rRNA genes. Three types of bread, which followed different bread-making processes, were in vitro digested and incubated with feces from IBD patients. Changes in gut microbiota were assessed by a quantitative polymerase chain reaction using specific bacterial sequence targets. Short-chain fatty acid production was also analyzed by gas chromatography. Lactobacillus sanfranciscensis was the dominant lactic acid bacteria species found in sourdough and bread doughs prepared using sourdough, whereas Saccharomyces cerevisiae was the most dominant yeast in all groups, especially in bread doughs before baking. Differences in microbial composition in raw bread doughs were more related to the type of dough and elaboration than to fermentation time lengths. The analysis of in vitro fecal incubations with bread conditions revealed an increase in most bacterial groups analyzed and short-chain fatty acid production, both in Crohn's disease and ulcerative colitis samples. Most remarkable increases in short-chain fatty acid production mirrored higher abundances of Roseburia species. The potential prebiotic properties observed were mainly obtained when using a high quantity of bread, regardless of bread type. Overall, this study highlights the bacterial dynamics within the bread-making process and the potential prebiotic effect in IBD patients.
Collapse
Affiliation(s)
- Aleix Lluansí
- Digestive Diseases and Microbiota Group, Institut d'Investigació Biomèdica de Girona, Salt, Spain
| | - Marc Llirós
- Digestive Diseases and Microbiota Group, Institut d'Investigació Biomèdica de Girona, Salt, Spain
| | | | - Anna Bahí
- Digestive Diseases and Microbiota Group, Institut d'Investigació Biomèdica de Girona, Salt, Spain
| | | | | | | | | | | | | | | | | | | | - David Busquets
- Digestive Service, Hospital Universitari de Girona Dr. Josep Trueta, Girona, Spain
| | - Leyanira Torreabla
- Digestive Service, Hospital Universitari de Girona Dr. Josep Trueta, Girona, Spain
| | - Miriam Sabat
- Digestive Service, Hospital Universitari de Girona Dr. Josep Trueta, Girona, Spain
| | - Maria Buxó
- Digestive Diseases and Microbiota Group, Institut d'Investigació Biomèdica de Girona, Salt, Spain
| | - Maria Cambra
- Digestive Diseases and Microbiota Group, Institut d'Investigació Biomèdica de Girona, Salt, Spain
| | | | | | | | | | - Xavier Aldeguer
- Digestive Diseases and Microbiota Group, Institut d'Investigació Biomèdica de Girona, Salt, Spain.,GoodGut S.L., Girona, Spain.,Digestive Service, Hospital Universitari de Girona Dr. Josep Trueta, Girona, Spain
| |
Collapse
|
49
|
Efficacy of a Preparation Based on Calcium Butyrate, Bifidobacterium bifidum, Bifidobacterium lactis, and Fructooligosaccharides in the Prevention of Relapse in Ulcerative Colitis: A Prospective Observational Study. J Clin Med 2021; 10:jcm10214961. [PMID: 34768480 PMCID: PMC8585056 DOI: 10.3390/jcm10214961] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/11/2021] [Accepted: 10/25/2021] [Indexed: 12/28/2022] Open
Abstract
Several compounds based on short chain fatty acids and/or probiotics/prebiotics have shown promising results in the therapy of ulcerative colitis (UC), possibly due to its key role in restoring gut homeostasis as well as intestinal barrier integrity. Here, we investigated the efficacy of a patented preparation based on calcium butyrate, Bifidobacterium bifidum, Bifidobacterium lactis, and fructooligosaccharides (FEEDColon®, Princeps, Cuneo, Italy) in maintaining remission and improving subjective symptoms and inflammatory indices in patients with UC receiving 5-ASA therapy. A total of 42 patients were prospectively recruited and randomized in 21 patients receiving combination therapy with mesalamine (5-ASA) plus FEEDColon® and 21 patients treated with standard 5-ASA therapy. Patients were assessed at baseline, at 6-month, and 12-month follow-up (FU). Therapeutic success (defined as Mayo partial score ≤ 2 and faecal calprotectin (FC) < 250 µg/g at 12-month FU) was reached by 32 (76%) patients: 20 (95%) among those treated with 5-ASA + FeedColon®, and 12 (57%) among those treated with 5-ASA only (p = 0.009). Consistently, patients treated with combination therapy improved subjective symptoms (quality of life, abdominal pain, and stool consistency) and reduced FC values, while those treated with 5-ASA alone, improved neither subjective symptoms nor FC during the FU. In conclusion, FEEDColon® supplementation appears to be a valid add-on therapy for the maintenance of remission in patients with UC. Further multicentre, placebo-controlled, double-blind clinical trials are needed to validate our results on larger cohorts of patients with UC.
Collapse
|
50
|
Schroeter CB, Huntemann N, Bock S, Nelke C, Kremer D, Pfeffer K, Meuth SG, Ruck T. Crosstalk of Microorganisms and Immune Responses in Autoimmune Neuroinflammation: A Focus on Regulatory T Cells. Front Immunol 2021; 12:747143. [PMID: 34691057 PMCID: PMC8529161 DOI: 10.3389/fimmu.2021.747143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) are the major determinant of peripheral immune tolerance. Many Treg subsets have been described, however thymus-derived and peripherally induced Tregs remain the most important subpopulations. In multiple sclerosis, a prototypical autoimmune disorder of the central nervous system, Treg dysfunction is a pathogenic hallmark. In contrast, induction of Treg proliferation and enhancement of their function are central immune evasion mechanisms of infectious pathogens. In accordance, Treg expansion is compartmentalized to tissues with high viral replication and prolonged in chronic infections. In friend retrovirus infection, Treg expansion is mainly based on excessive interleukin-2 production by infected effector T cells. Moreover, pathogens seem also to enhance Treg functions as shown in human immunodeficiency virus infection, where Tregs express higher levels of effector molecules such as cytotoxic T-lymphocyte-associated protein 4, CD39 and cAMP and show increased suppressive capacity. Thus, insights into the molecular mechanisms by which intracellular pathogens alter Treg functions might aid to find new therapeutic approaches to target central nervous system autoimmunity. In this review, we summarize the current knowledge of the role of pathogens for Treg function in the context of autoimmune neuroinflammation. We discuss the mechanistic implications for future therapies and provide an outlook for new research directions.
Collapse
Affiliation(s)
- Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefanie Bock
- Department of Neurology With Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|