1
|
Covello C, Becherucci G, Di Vincenzo F, Del Gaudio A, Pizzoferrato M, Cammarota G, Gasbarrini A, Scaldaferri F, Mentella MC. Parenteral Nutrition, Inflammatory Bowel Disease, and Gut Barrier: An Intricate Plot. Nutrients 2024; 16:2288. [PMID: 39064731 PMCID: PMC11279609 DOI: 10.3390/nu16142288] [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: 05/26/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Malnutrition poses a critical challenge in inflammatory bowel disease, with the potential to detrimentally impact medical treatment, surgical outcomes, and general well-being. Parenteral nutrition is crucial in certain clinical scenarios, such as with patients suffering from short bowel syndrome, intestinal insufficiency, high-yielding gastrointestinal fistula, or complete small bowel obstruction, to effectively manage malnutrition. Nevertheless, research over the years has attempted to define the potential effects of parenteral nutrition on the intestinal barrier and the composition of the gut microbiota. In this narrative review, we have gathered and analyzed findings from both preclinical and clinical studies on this topic. Based on existing evidence, there is a clear correlation between short- and long-term parenteral nutrition and negative effects on the intestinal system. These include mucosal atrophic damage and immunological and neuroendocrine dysregulation, as well as alterations in gut barrier permeability and microbiota composition. However, the mechanistic role of these changes in inflammatory bowel disease remains unclear. Therefore, further research is necessary to effectively address the numerous gaps and unanswered questions pertaining to these issues.
Collapse
Affiliation(s)
- Carlo Covello
- Gastroenterology Department, Centro di Malattie dell’Apparato Digerente (CEMAD), Center for Diagnosis and Treatment of Digestive Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (F.D.V.); (A.D.G.); (A.G.)
| | - Guia Becherucci
- UOS Malattie Infiammatorie Croniche Intestinali, Centro di Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.B.); (F.S.)
| | - Federica Di Vincenzo
- Gastroenterology Department, Centro di Malattie dell’Apparato Digerente (CEMAD), Center for Diagnosis and Treatment of Digestive Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (F.D.V.); (A.D.G.); (A.G.)
| | - Angelo Del Gaudio
- Gastroenterology Department, Centro di Malattie dell’Apparato Digerente (CEMAD), Center for Diagnosis and Treatment of Digestive Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (F.D.V.); (A.D.G.); (A.G.)
| | - Marco Pizzoferrato
- UOC Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (G.C.)
| | - Giovanni Cammarota
- UOC Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (G.C.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Gastroenterology Department, Centro di Malattie dell’Apparato Digerente (CEMAD), Center for Diagnosis and Treatment of Digestive Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (F.D.V.); (A.D.G.); (A.G.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Franco Scaldaferri
- UOS Malattie Infiammatorie Croniche Intestinali, Centro di Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.B.); (F.S.)
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maria Chiara Mentella
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- UOC di Nutrizione Clinica, Dipartimento Scienze Mediche e Chirurgiche Addominali ed Endocrino-Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
2
|
Ossola M, Ferrocino I, Franciosa I, Aimasso U, Cravero L, Bonciolini A, Cardenia V, Merlo FD, Anrò M, Chiarotto A, Bosa C, Cocolin L, Bo S. Does Microbiome Matter in Chronic Intestinal Failure Due to Type 1 Short Bowel Syndrome in Adults? Nutrients 2024; 16:2282. [PMID: 39064725 PMCID: PMC11280028 DOI: 10.3390/nu16142282] [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: 06/08/2024] [Revised: 06/29/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
The exact microbiome composition and function of patients with Short Bowel Syndrome (SBS) and Chronic Intestinal Failure (CIF) are still unknown. Patients with type I SBS-CIF (end-jejunostomy/ileostomy) are little represented in available studies. The aim of this study is to evaluate the microbiome characteristics of adult type 1 SBS-CIF patients according to their clinical features. Fecal microbiota was studied by amplicon-based sequencing and volatile organic compounds (VOCs) were assessed by solid-phase microextraction and gas chromatography-mass spectrometry. A total of 44 adult type 1 SBS-CIF patients were enrolled. At the family level, Lactobacillaceae (38% of the relative frequency) and Streptococcaceae (24%) were predominant; at the genus level, Streptococcus (38% of the relative frequency) and Lactobacillus (24%) were the dominant amplicon sequence variants (ASVs). Patients with increased stomal output showed higher ASVs for Lactobacillus (Rho = +0.38; p = 0.010), which was confirmed after adjusting for small bowel length (OR = 1.04; 95% CI 1.01-1.07, p = 0.023). Hyperphagia was associated with higher concentrations of short-chain fatty acid (SCFA) esters, such as butanoic acid ethyl ester (p = 0.005) and hexanoic acid ethyl ester (p = 0.004). Dietary fiber intake was directly correlated with most VOCs. Hyperphagia was associated with dietary fiber, after adjusting for small bowel length (OR = 1.35; 95% CI 1.01-1.81; p = 0.040). In type 1 SBS-CIF patients, a greater frequency of Lactobacilli was associated with increased stomal outputs, while increased fiber intake and concentrations of SCFA esters were associated with hyperphagia. These results might have implications for clinical practice.
Collapse
Affiliation(s)
- Marta Ossola
- Dietetic and Clinical Nutrition Unit, Città della Salute e della Scienza Hospital, C.so Bramante 88, 10126 Torino, Italy
| | - Ilario Ferrocino
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Irene Franciosa
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Umberto Aimasso
- Dietetic and Clinical Nutrition Unit, Città della Salute e della Scienza Hospital, C.so Bramante 88, 10126 Torino, Italy
| | - Leila Cravero
- Department of Medical Science, University of Torino, C.so Dogliotti 14, 10126 Torino, Italy
| | - Ambra Bonciolini
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Vladimiro Cardenia
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Fabio Dario Merlo
- Dietetic and Clinical Nutrition Unit, Città della Salute e della Scienza Hospital, C.so Bramante 88, 10126 Torino, Italy
| | - Marta Anrò
- Dietetic and Clinical Nutrition Unit, Città della Salute e della Scienza Hospital, C.so Bramante 88, 10126 Torino, Italy
| | - Alessia Chiarotto
- Dietetic and Clinical Nutrition Unit, Città della Salute e della Scienza Hospital, C.so Bramante 88, 10126 Torino, Italy
| | - Clara Bosa
- Dietetic and Clinical Nutrition Unit, Città della Salute e della Scienza Hospital, C.so Bramante 88, 10126 Torino, Italy
| | - Luca Cocolin
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Simona Bo
- Dietetic and Clinical Nutrition Unit, Città della Salute e della Scienza Hospital, C.so Bramante 88, 10126 Torino, Italy
- Department of Medical Science, University of Torino, C.so Dogliotti 14, 10126 Torino, Italy
| |
Collapse
|
3
|
Pironi L, D'Amico F, Guidetti M, Brigidi P, Sasdelli AS, Turroni S. The gut microbiota in adults with chronic intestinal failure. Clin Nutr 2024; 43:1331-1342. [PMID: 38677044 DOI: 10.1016/j.clnu.2024.04.018] [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: 11/21/2023] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
OBJECTIVE Fecal microbiota was investigated in adult patients with chronic intestinal failure (CIF) due to short bowel syndrome (SBS) with jejunocolonic anastomosis (SBS-2). Few or no data are available on SBS with jejunostomy (SBS-1) and CIF due to intestinal dysmotility (DYS) or mucosal disease (MD). We profiled the fecal microbiota of various pathophysiological mechanisms of CIF. METHODS Cross-sectional study on 61 adults with CIF (SBS-1 30, SBS-2 17, DYS 8, MD 6). Fecal samples were collected and profiled by 16S rRNA amplicon sequencing. Healthy controls (HC) were selected from pre-existing cohorts, matched with patients by sex and age. RESULTS Compared to HC, SBS-1, SBS-2 and MD patients showed lower alpha diversity; no difference was found for DYS. In beta diversity analysis, SBS-1, SBS-2 and DYS groups segregated from HC and from each other. Taxonomically, the CIF groups differed from HC even at the phylum level. In particular, CIF patients' microbiota was dominated by Lactobacillaceae and Enterobacteriaceae, while depleted in typical health-associated taxa belonging to Lachnospiraceae and Ruminococcaceae. Notably, compositional peculiarities of the CIF groups emerged. Furthermore, in the SBS groups, the microbiota profile differed according to the amount of parenteral nutrition required and the duration of CIF. CONCLUSIONS CIF patients showed marked intestinal dysbiosis with microbial signatures specific to the pathophysiological mechanism of CIF as well as to the severity and duration of SBS.
Collapse
Affiliation(s)
- Loris Pironi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; Centre for Chronic Intestinal Failure, IRCCS AOUBO, Bologna, Italy.
| | - Federica D'Amico
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
4
|
Senanayake T, Makanyengo S, Hoedt EC, Goggins B, Smith SR, Keely S. Influence of the bile acid/microbiota axis in ileal surgery: a systematic review. Colorectal Dis 2024; 26:243-257. [PMID: 38177086 DOI: 10.1111/codi.16837] [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: 04/02/2023] [Revised: 09/21/2023] [Accepted: 11/09/2023] [Indexed: 01/06/2024]
Abstract
AIM The gastrointestinal bile acid (BA)/microbiota axis has emerged as a potential mediator of health and disease, particularly in relation to pathologies such as inflammatory bowel disease (IBD) and colorectal cancer. Whilst it presents an exciting new avenue for therapies, it has not yet been characterized in surgical resection of the ileum, where BA reabsorption occurs. The identification of BA/microbiota signatures may provide future therapies with perioperative personalized medicine. In this work we conduct a systematic review with the aim of investigating the microbiome and BA changes that are associated with resection of the ileum. METHOD The databases included were MEDLINE, EMBASE, Web of Science and Cochrane libraries. The outcomes of interest were faecal microbiome and BA signatures after ileal resection. RESULTS Of the initial 3106 articles, three studies met the inclusion/exclusion criteria for data extraction. A total of 257 patients (46% surgery, 54% nonsurgery controls) were included in the three studies. Two studies included patients with short bowel syndrome and the other included patients with IBD. Large-scale microbiota changes were reported. In general, alpha diversity had decreased amongst patients with ileal surgery. Phylum-level changes included decreased Bacteroidetes and increased Proteobacteria and Fusobacteria in patients with an intestinal resection. Surgery was associated with increased total faecal BAs, cholic acid and chenodeoxycholic acid. There were decreases in deoxycholic acid and glycine and taurine conjugated bile salts. Integrated BA and microbiota data identified correlations with several bacterial families and BA. CONCLUSION The BA/microbiota axis is still a novel area with minimal observational data in surgery. Further mechanistic research is necessary to further explore this and identify its role in improving perioperative outcomes.
Collapse
Affiliation(s)
- Tharindu Senanayake
- NHMRC Centre of Research Excellence in Digestive Health, New Lambton Heights, New South Wales, Australia
- Surgical and Perioperative Care Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, Immune Health Program, New Lambton Heights, New South Wales, Australia
| | - Samwel Makanyengo
- NHMRC Centre of Research Excellence in Digestive Health, New Lambton Heights, New South Wales, Australia
- Surgical and Perioperative Care Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, Immune Health Program, New Lambton Heights, New South Wales, Australia
| | - Emily C Hoedt
- NHMRC Centre of Research Excellence in Digestive Health, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, Immune Health Program, New Lambton Heights, New South Wales, Australia
| | - Bridie Goggins
- NHMRC Centre of Research Excellence in Digestive Health, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, Immune Health Program, New Lambton Heights, New South Wales, Australia
| | - Stephen R Smith
- Surgical and Perioperative Care Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Simon Keely
- NHMRC Centre of Research Excellence in Digestive Health, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, Immune Health Program, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
5
|
Wolfschluckner V, Obermüller B, Horvath A, Rodriguez-Blanco G, Fuchs P, Miekisch W, Mittl B, Flucher C, Till H, Singer G. Metabolomic Alterations of Volatile Organic Compounds and Bile Acids as Biomarkers of Microbial Shifts in a Murine Model of Short Bowel Syndrome. Nutrients 2023; 15:4949. [PMID: 38068807 PMCID: PMC10708115 DOI: 10.3390/nu15234949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Pediatric short bowel syndrome (SBS) is a rare condition characterized by a massive loss of the small intestine, leading to the inability to meet nutritional requirements without the use of parenteral or enteral supplementation. SBS causes profound alterations in the intestinal microbiome and metabolome. The aim of this study was a detailed assessment of the intestinal microbiome and metabolome in a murine model of SBS. We performed a 60% proximal small bowel resection versus a sham operation in C57BL/6 mice. Four weeks postoperatively, the microbial communities of different intestinal segments (jejunum, ileum, colon) and stool were assessed by 16S rRNA gene sequencing. Bile acids in serum and stool and volatile organic compounds (VOCs) in the fecal headspace were assessed using LC-MS and GC-MS techniques. The α-diversity of the different intestinal segments did not significantly differ between the two groups. β-diversity significantly differed between sham and SBS mice. While in the jejunum, Faecalibaculum was significantly increased in SBS animals, a significant reduction in Lactobacillus and Sporosarcina was detected in the ileum of SBS mice. In the colon of SBS mice, a significant decrease in Ruminococcaceae and a significant increase in Proteobacteria such as Faecalibaculum and Escherichia-Shigella were found. Serum levels of deoxycholic, taurocholic and taurochenodeoxycholic acids were significantly higher in the SBS group. Of the 29 VOCs tested, hexane, isoflurane and pentane were significantly higher in the SBS group, and pyrrole was significantly lower. We were able to show that SBS causes shifts in the murine intestinal microbiome and metabolome including serum BAs and fecal VOCs.
Collapse
Affiliation(s)
- Vanessa Wolfschluckner
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (V.W.); (B.M.); (C.F.); (H.T.); (G.S.)
| | - Beate Obermüller
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (V.W.); (B.M.); (C.F.); (H.T.); (G.S.)
| | - Angela Horvath
- Division of Gastroenterology and Hepatology, Medical University of Graz, 8036 Graz, Austria;
| | - Giovanny Rodriguez-Blanco
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria;
| | - Patricia Fuchs
- Department of Anaesthesiology, Intensive Care and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany; (P.F.); (W.M.)
| | - Wolfram Miekisch
- Department of Anaesthesiology, Intensive Care and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany; (P.F.); (W.M.)
| | - Barbara Mittl
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (V.W.); (B.M.); (C.F.); (H.T.); (G.S.)
| | - Christina Flucher
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (V.W.); (B.M.); (C.F.); (H.T.); (G.S.)
| | - Holger Till
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (V.W.); (B.M.); (C.F.); (H.T.); (G.S.)
| | - Georg Singer
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (V.W.); (B.M.); (C.F.); (H.T.); (G.S.)
| |
Collapse
|
6
|
Chowdhury F, Hill L, Shah N, Popov J, Cheveldayoff P, Pai N. Intestinal microbiome in short bowel syndrome: diagnostic and therapeutic opportunities. Curr Opin Gastroenterol 2023; 39:463-471. [PMID: 37751391 DOI: 10.1097/mog.0000000000000970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
PURPOSE OF REVIEW The intestinal microbiome plays a strong, complementary role in the development and integrity of the intestinal epithelium. This biology is crucial for intestinal adaptation, particularly after the mucosal insults that lead to short bowel syndrome (SBS). The purpose of this review is to discuss relationships between the intestinal microbiota and the physiology of intestinal adaptation. RECENT FINDINGS We will address interactions between the intestinal microbiome and nutritional metabolism, factors leading to dysbiosis in SBS, and common compositional differences of the gut microbiome in SBS patients as compared to healthy controls. We will also discuss novel opportunities to expand diagnostic and therapeutic interventions in this population, by using our knowledge of the microbiome to manipulate luminal bacteria and study their resultant metabolites. As microbial therapeutics advance across so many fields of medicine, this review is timely in its advocacy for ongoing research that focuses on the SBS population.Our review will discuss 4 key areas: 1) physiology of the intestinal microbiome in SBS, 2) clinical and therapeutic insults that lead to a state of dysbiosis, 3) currently available evidence on microbiome-based approaches to SBS management, and 4) opportunities and innovations to inspire future research. SUMMARY The clinical implications of this review are both current, and potential. Understanding how the microbiome impacts intestinal adaptation and host physiology may enhance our understanding of why we experience such clinical variability in SBS patients' outcomes. This review may also expand clinicians' understanding of what 'personalized medicine' can mean for this patient population, and how we may someday consider our nutritional, therapeutic, and prognostic recommendations based on our patients' host, and microbial physiology.
Collapse
Affiliation(s)
- Fariha Chowdhury
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
| | - Lee Hill
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
- Department of Pediatrics, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Nyah Shah
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
| | - Jelena Popov
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paige Cheveldayoff
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
- Centre for Metabolism, Obesity and Diabetes Research
| | - Nikhil Pai
- Department of Pediatrics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario
- Centre for Metabolism, Obesity and Diabetes Research
- Farncombe Family Digestive Health Research Institute, McMaster University
- Division of Pediatric Gastroenterology & Nutrition, McMaster Children's Hospital, Hamilton, Ontario, Canada
| |
Collapse
|
7
|
Jiang L, Xu J, Cheng SY, Wang Y, Cai W. The gut microbiome and intestinal failure-associated liver disease. Hepatobiliary Pancreat Dis Int 2023; 22:452-457. [PMID: 37453856 DOI: 10.1016/j.hbpd.2023.07.002] [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: 04/25/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Intestinal failure-associated liver disease (IFALD) is a common hepatobiliary complication resulting from long-term parenteral nutrition (PN) in patients with intestinal failure. The spectrum of IFALD ranges from cholestasis, steatosis, portal fibrosis, to cirrhosis. Development of IFALD is a multifactorial process, in which gut dysbiosis plays a critical role in its initiation and progression in conjunction with increased intestinal permeability, activation of hepatic immune responses, and administration of lipid emulsion. Gut microbiota manipulation including pre/probiotics, fecal microbiota transplantation, and antibiotics has been studied in IFALD with varying success. In this review, we summarize current knowledge on the taxonomic and functional changes of gut microbiota in preclinical and clinical studies of IFALD. We also review the function of microbial metabolites and associated signalings in the context of IFALD. By providing microbiota-targeted interventions aiming to optimize PN-induced liver injury, our review provides perspectives for future basic and translational investigations in the field.
Collapse
Affiliation(s)
- Lu Jiang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Juan Xu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Si-Yang Cheng
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Wei Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
8
|
Busing JD, Fouladi F, Bulik-Sullivan EC, Carroll IM, Fodor AA, Thomsen KF, Gulati AS, Nicholson MR. Gut Microbial Changes Following Fecal Microbiota Transplantation for D-Lactic Acidosis in Two Children. JPGN REPORTS 2023; 4:e319. [PMID: 37600604 PMCID: PMC10435018 DOI: 10.1097/pg9.0000000000000319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 03/05/2023] [Indexed: 08/22/2023]
Abstract
D-lactic acidosis (D-LA) is an uncommon complication of short bowel syndrome characterized by elevated plasma D-lactate and encephalopathy. Treatments include rehydration, dietary carbohydrate restriction, and antibiotics to alter the gut microbiota. Fecal microbiota transplantation (FMT) has recently been used in children to successfully treat D-LA. We compared the clinical course and then utilized metagenomic shotgun sequencing to describe changes in the composition and function of the intestinal microbiome following FMT in 2 patients with recurrent D-LA. FMT altered the composition of the fecal microbiota in these 2 patients with recurrent D-LA, though not necessarily in a consistent manner. Importantly, microbial metabolic pathways were also impacted by FMT, which may be critical for achieving desired clinical outcomes. While sample size limits the generalizability of our results, these findings set the stage for further understanding of the role of microbes in the pathogenesis of recurrent D-LA.
Collapse
Affiliation(s)
- Jordan D. Busing
- From the Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN
| | - Farnaz Fouladi
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC
| | | | - Ian M. Carroll
- Department of Nutrition, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Anthony A. Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC
| | - Kelly F. Thomsen
- From the Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN
| | - Ajay S. Gulati
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Maribeth R. Nicholson
- From the Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
9
|
Huang Y, Jiao J, Yao D, Guo F, Li Y. Altered fecal microbiome and metabolome profiles in rat models of short bowel syndrome. Front Microbiol 2023; 14:1185463. [PMID: 37362931 PMCID: PMC10289890 DOI: 10.3389/fmicb.2023.1185463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
INTRODUCTION Short bowel syndrome (SBS) is featured by impaired nutrients and fluids absorption due to massive small intestine resection. Gut dysbiosis has been implicated in SBS, this study aimed to characterize the metagenomic and metabolomic profiles of SBS and identify potential therapeutic targets. METHODS Fecal samples from SBS and Sham rats (n = 8 per group) were collected for high-throughput metagenomic sequencing. Fecal metabolomics was measured by untargeted liquid chromatography-mass spectrometry. RESULTS We found that the species-level α-diversity significantly decreased in SBS rats, accompanied by altered microbiome compositions. The beneficial anaerobes from Firmicutes and Bacteroidetes were depleted while microorganisms from Lactobacillus, Escherichia, Enterococcus, and Streptococcus were enriched in faces from SBS rats. LEfSe analysis identified 17 microbial species and 38 KEGG modules that were remarkably distinct between SBS and Sham rats. In total, 1,577 metabolites with known chemical identity were detected from all samples, among them, 276 metabolites were down-regulated and 224 metabolites were up-regulated in SBS group. The typical signatures of SBS fecal metabolome comprised reduced short-chain fatty acids and products of amino acid metabolism (indole derivatives and p-cresol), as well as altered bile acid spectrum. We revealed 215 robust associations between representative differentially abundant microbial species and metabolites, the species with the same changing trend tended to have a similar correlation with some certain metabolites. CONCLUSION The fecal microbiome and metabolome significantly altered in SBS. Our findings may lay the foundation for developing new strategies to facilitate intestinal adaptation in SBS patients.
Collapse
Affiliation(s)
- Yuhua Huang
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jian Jiao
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Danhua Yao
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Feilong Guo
- Department of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Yousheng Li
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
The Role of a Colon-in-Continuity in Short Bowel Syndrome. Nutrients 2023; 15:nu15030628. [PMID: 36771335 PMCID: PMC9918966 DOI: 10.3390/nu15030628] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/28/2023] Open
Abstract
Short bowel syndrome (SBS) is a rare gastrointestinal condition that is defined as having less than 200 cm of remaining small intestine. SBS results from extensive surgical resection and is associated with a high risk for intestinal failure (IF) with a need for parenteral support (PS). Depending on the region of intestinal resection, three different main anatomy types can be distinguished from each other. In this review, we synthesize the current knowledge on the role of the colon in the setting of SBS-IF with a colon-in-continuity (SBS-IF-CiC), e.g., by enhancing the degree of intestinal adaptation, energy salvage, and the role of the microbiota. In addition, the effect of the disease-modifying treatment with glucagon-like peptide-2 (GLP-2) analogs in SBS-IF-CiC and how it differs from patients without a colon will be discussed. Overall, the findings explained in this review highlight the importance of preservation of the colon in SBS-IF.
Collapse
|
11
|
Berlin P, Barrantes I, Reiner J, Schröder E, Vollmar B, Bull J, Kreikemeyer B, Lamprecht G, Witte M. Dysbiosis and reduced small intestinal function are required to induce intestinal insufficiency in mice. Am J Physiol Gastrointest Liver Physiol 2023; 324:G10-G23. [PMID: 36346150 PMCID: PMC9799149 DOI: 10.1152/ajpgi.00201.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Extensive bowel resection can lead to short bowel syndrome and intestinal failure. Resection-induced dysbiosis may be related to the specific anatomic site of resection and influences the disease progression. Although patients with end-jejunostomy are at high risk for intestinal failure, preservation of the ileocecal valve and colon counteracts this risk. The present study investigated the role of the cecum in maintaining microbial homeostasis after different types of small bowel resection. Male C57BL6/J mice were anesthetized by intraperitoneal injection of ketamine-xylazine and received extended ileocecal resection (extended ICR), limited ileocecal resection (limited ICR), or mid-small bowel resection (SBR). Stool samples were collected before surgery and between postoperative days 2-7, for 16S rRNA gene sequencing. Only extended ICR, but neither limited ICR nor SBR, induced intestinal insufficiency. α-Diversity was reduced in both ICR variants but not after SBR. All resections resulted in an increase in Proteobacteria. Pathobionts, such as Clostridia, Shigella, and Enterococcus, increased after SBR while Muribaculaceae, Lactobacillus, and Lachnospiraceae decreased. Limited ICR resulted in an increase of members of the Clostridium sensu stricto group, Terrisporobacter and Enterococcus and a decrease of Muribaculaceae. The increase of Enterococcus was even more pronounced after extended ICR while Muribaculaceae and Akkermansia were dramatically reduced. Both ICR variants caused a decrease in steroid biosynthesis and glycosaminoglycan degradation-associated pathways, suggesting altered bile acid transformation and mucus utilization.NEW & NOTEWORTHY Resection-induced dysbiosis affects disease progression in patients with short bowel syndrome. Severe dysbiosis occurs after removal of the ileocecal valve, even in the absence of short bowel conditions, and is associated with the loss of Muribaculaceae and Akkermansia and an increase of Clostridium and Enterococcus. The preservation of the cecum should be considered in surgical therapy, and dysbiosis should be targeted based on its specific anatomical signature to improve postoperative bacterial colonization.
Collapse
Affiliation(s)
- Peggy Berlin
- 1Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, Rostock, Germany
| | - Israel Barrantes
- 2Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Johannes Reiner
- 1Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, Rostock, Germany
| | - Emma Schröder
- 1Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Vollmar
- 3Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Jana Bull
- 4Institute for Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Bernd Kreikemeyer
- 4Institute for Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Georg Lamprecht
- 1Division of Gastroenterology, Department of Medicine II, Rostock University Medical Center, Rostock, Germany
| | - Maria Witte
- 5Department of General, Visceral, Thoracic, Vascular and Transplant Surgery, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
12
|
Cerdó T, García-Santos JA, Rodríguez-Pöhnlein A, García-Ricobaraza M, Nieto-Ruíz A, G. Bermúdez M, Campoy C. Impact of Total Parenteral Nutrition on Gut Microbiota in Pediatric Population Suffering Intestinal Disorders. Nutrients 2022; 14:4691. [PMID: 36364953 PMCID: PMC9658482 DOI: 10.3390/nu14214691] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023] Open
Abstract
Parenteral nutrition (PN) is a life-saving therapy providing nutritional support in patients with digestive tract complications, particularly in preterm neonates due to their gut immaturity during the first postnatal weeks. Despite this, PN can also result in several gastrointestinal complications that are the cause or consequence of gut mucosal atrophy and gut microbiota dysbiosis, which may further aggravate gastrointestinal disorders. Consequently, the use of PN presents many unique challenges, notably in terms of the potential role of the gut microbiota on the functional and clinical outcomes associated with the long-term use of PN. In this review, we synthesize the current evidence on the effects of PN on gut microbiome in infants and children suffering from diverse gastrointestinal diseases, including necrotizing enterocolitis (NEC), short bowel syndrome (SBS) and subsequent intestinal failure, liver disease and inflammatory bowel disease (IBD). Moreover, we discuss the potential use of pre-, pro- and/or synbiotics as promising therapeutic strategies to reduce the risk of severe gastrointestinal disorders and mortality. The findings discussed here highlight the need for more well-designed studies, and harmonize the methods and its interpretation, which are critical to better understand the role of the gut microbiota in PN-related diseases and the development of efficient and personalized approaches based on pro- and/or prebiotics.
Collapse
Affiliation(s)
- Tomás Cerdó
- Maimonides Institute for Research in Biomedicine of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - José Antonio García-Santos
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Anna Rodríguez-Pöhnlein
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - María García-Ricobaraza
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Ana Nieto-Ruíz
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Mercedes G. Bermúdez
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
| | - Cristina Campoy
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria Ibs-GRANADA, Health Sciences Technological Park, 18012 Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada’s Node, Carlos III Health Institute, Avda. Monforte de Lemos 5, 28028 Madrid, Spain
| |
Collapse
|
13
|
Gerasimidis K, Gkikas K, Stewart C, Neelis E, Svolos V. Microbiome and paediatric gut diseases. Arch Dis Child 2022; 107:784-789. [PMID: 34716173 DOI: 10.1136/archdischild-2020-320875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/16/2021] [Indexed: 11/04/2022]
Abstract
In the human gut resides a vast community of microorganisms which perform critical functions for the maintenance of whole body homeostasis. Changes in the composition and function of this community, termed microbiome, are believed to provoke disease onset, including non-communicable diseases. In this review, we debate the current evidence on the role of the gut microbiome in the pathogenesis, outcomes and management of paediatric gut disease. We conclude that even though the gut microbiome is altered in paediatric inflammatory bowel disease, coeliac disease, intestinal failure, necrotising enterocolitis and irritable bowel syndrome, there are currently very few implications for unravelling disease pathogenesis or guiding clinical practice. In the future, the gut microbiome may aid in disease differential diagnosis and prediction of clinical outcomes, and comprise a target for therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Christopher Stewart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Esther Neelis
- Paediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Vaios Svolos
- Human Nutrition, University of Glasgow, Glasgow, UK
| |
Collapse
|
14
|
Bauer KC, York EM, Cirstea MS, Radisavljevic N, Petersen C, Huus KE, Brown EM, Bozorgmehr T, Berdún R, Bernier L, Lee AHY, Woodward SE, Krekhno Z, Han J, Hancock REW, Ayala V, MacVicar BA, Finlay BB. Gut microbes shape microglia and cognitive function during malnutrition. Glia 2022; 70:820-841. [PMID: 35019164 PMCID: PMC9305450 DOI: 10.1002/glia.24139] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/17/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022]
Abstract
Fecal-oral contamination promotes malnutrition pathology. Lasting consequences of early life malnutrition include cognitive impairment, but the underlying pathology and influence of gut microbes remain largely unknown. Here, we utilize an established murine model combining malnutrition and iterative exposure to fecal commensals (MAL-BG). The MAL-BG model was analyzed in comparison to malnourished (MAL mice) and healthy (CON mice) controls. Malnourished mice display poor spatial memory and learning plasticity, as well as altered microglia, non-neuronal CNS cells that regulate neuroimmune responses and brain plasticity. Chronic fecal-oral exposures shaped microglial morphology and transcriptional profile, promoting phagocytic features in MAL-BG mice. Unexpectedly, these changes occurred independently from significant cytokine-induced inflammation or blood-brain barrier (BBB) disruption, key gut-brain pathways. Metabolomic profiling of the MAL-BG cortex revealed altered polyunsaturated fatty acid (PUFA) profiles and systemic lipoxidative stress. In contrast, supplementation with an ω3 PUFA/antioxidant-associated diet (PAO) mitigated cognitive deficits within the MAL-BG model. These findings provide valued insight into the malnourished gut microbiota-brain axis, highlighting PUFA metabolism as a potential therapeutic target.
Collapse
Affiliation(s)
- Kylynda C. Bauer
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Elisa M. York
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Mihai S. Cirstea
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Nina Radisavljevic
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Biochemistry and Molecular Biology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Charisse Petersen
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
| | - Kelsey E. Huus
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Eric M. Brown
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | | | - Rebeca Berdún
- Institut de Recerca Biomèdica de Lleida (IRB‐Lleida)LleidaSpain
- Department of Experimental MedicineUniversitat de Lleida (UdL)LleidaSpain
| | - Louis‐Philippe Bernier
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Amy H. Y. Lee
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Sarah E. Woodward
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Zakhar Krekhno
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
| | - Jun Han
- The Metabolomics Innovation CentreUniversity of VictoriaVictoriaCanada
| | - Robert E. W. Hancock
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Victoria Ayala
- Institut de Recerca Biomèdica de Lleida (IRB‐Lleida)LleidaSpain
- Department of Experimental MedicineUniversitat de Lleida (UdL)LleidaSpain
| | - Brian A. MacVicar
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Barton Brett Finlay
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
- Biochemistry and Molecular Biology DepartmentUniversity of British ColumbiaVancouverCanada
| |
Collapse
|
15
|
Hartmann P. Editorial: The Microbiome in Hepatobiliary and Intestinal Disease. Front Physiol 2022; 13:893074. [PMID: 35492588 PMCID: PMC9044070 DOI: 10.3389/fphys.2022.893074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Phillipp Hartmann
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Division of Gastroenterology, Hepatology and Nutrition, Rady Children’s Hospital San Diego, San Diego, CA, United States
- *Correspondence: Phillipp Hartmann,
| |
Collapse
|
16
|
Shaughnessy MP, Park CJ, Salvi PS, Cowles RA. Jejunoileal mucosal growth in mice with a limited microbiome. PLoS One 2022; 17:e0266251. [PMID: 35349599 PMCID: PMC8963542 DOI: 10.1371/journal.pone.0266251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/15/2022] [Indexed: 11/18/2022] Open
Abstract
Previous work demonstrated enhanced enterocyte proliferation and mucosal growth in gnotobiotic mice, suggesting that intestinal flora participate in mucosal homeostasis. Furthermore, broad-spectrum enteral antibiotics are known to induce near germ-free (GF) conditions in mice with conventional flora (CONV). We hypothesized that inducing near GF conditions with broad-spectrum enteral antibiotics would cause ordered small intestinal mucosal growth in CONV mice but would have no effect in GF mice with no inherent microbiome. C57BL/6J CONV and GF mice received either an antibiotic solution (Ampicillin, Ciprofloxacin, Metronidazole, Vancomycin, Meropenem) or a vehicle alone. After treatment, small intestinal villus height (VH), crypt depth (CD), mucosal surface area (MSA), crypt proliferation index (CPI), apoptosis, and villus and crypt cell types were assessed. Antibiotic-treated CONV (Abx-CONV) mice had taller villi, deeper crypts, increased CPI, increased apoptosis, and greater MSA compared to vehicle-treated CONV mice. Minor differences were noted in enterocyte and enterochromaffin cell proportions between groups, but goblet and Paneth cell proportions were unchanged in Abx-CONV mice compared to vehicle-treated CONV mice (p>0.05). Antibiotics caused no significant changes in VH or MSA in GF mice when compared to vehicle-treated GF mice (p>0.05). Enteral administration of broad-spectrum antibiotics to mice with a conventional microbiome stimulates ordered small intestinal mucosal growth. Mucosal growth was not seen in germ-free mice treated with antibiotics, implying that intestinal mucosal growth is associated with change in the microbiome in this model.
Collapse
Affiliation(s)
- Matthew P. Shaughnessy
- Division of Pediatric Surgery, Department of Surgery, Yale University, New Haven, Connecticut, United States of America
| | - Christine J. Park
- Division of Pediatric Surgery, Department of Surgery, Yale University, New Haven, Connecticut, United States of America
| | - Pooja S. Salvi
- Division of Pediatric Surgery, Department of Surgery, Yale University, New Haven, Connecticut, United States of America
| | - Robert A. Cowles
- Division of Pediatric Surgery, Department of Surgery, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
17
|
Jiang L, Wang Y, Xiao Y, Wang Y, Yan J, Schnabl B, Cai W. Role of the Gut Microbiota in Parenteral Nutrition-Associated Liver Disease: From Current Knowledge to Future Opportunities. J Nutr 2022; 152:377-385. [PMID: 34734271 DOI: 10.1093/jn/nxab380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/02/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022] Open
Abstract
Parenteral nutrition-associated liver disease (PNALD) refers to a spectrum of conditions that can develop cholestasis, steatosis, fibrosis, and cirrhosis in the setting of parenteral nutrition (PN) use. Patient risk factors include short bowel syndrome, bacterial overgrowth and translocation, disturbance of hepatobiliary circulation, and lack of enteral feeding. A growing body of evidence suggests an intricate linkage between the gut microbiota and the pathogenesis of PNALD. In this review, we highlight current knowledge on the taxonomic and functional changes in the gut microbiota that might serve as noninvasive biomarkers. We also discuss the function of microbial metabolites and associated signaling pathways in the pathogenesis of PNALD. By providing the perspectives of microbiota-host interactions in PNALD for basic and translational research and summarizing current limitations of microbiota-based approaches, this review paves the path for developing novel and precise microbiota-based therapies in PNALD.
Collapse
Affiliation(s)
- Lu Jiang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yongtao Xiao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yong Wang
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junkai Yan
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.,Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| | - Wei Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Boutte HJ, Chen J, Wylie TN, Wylie KM, Xie Y, Geisman M, Prabu A, Gazit V, Tarr PI, Levin MS, Warner BW, Davidson NO, Rubin DC. Fecal microbiome and bile acid metabolome in adult short bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2022; 322:G154-G168. [PMID: 34816756 PMCID: PMC8793869 DOI: 10.1152/ajpgi.00091.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Loss of functional small bowel surface area causes short bowel syndrome (SBS), intestinal failure, and parenteral nutrition (PN) dependence. The gut adaptive response following resection may be difficult to predict, and it may take up to 2 yr to determine which patients will wean from PN. Here, we examined features of gut microbiota and bile acid (BA) metabolism in determining adaptation and ability to wean from PN. Stool and sera were collected from healthy controls and from patients with SBS (n = 52) with ileostomy, jejunostomy, ileocolonic, and jejunocolonic anastomoses fed with PN plus enteral nutrition or who were exclusively enterally fed. We undertook 16S rRNA gene sequencing, BA profiling, and 7α-hydroxy-4-cholesten-3-one (C4) quantitation with LC-MS/MS and serum amino acid analyses. Patients with SBS exhibited altered gut microbiota with reduced gut microbial diversity compared with healthy controls. We observed differences in the microbiomes of patients with SBS with ileostomy versus jejunostomy, jejunocolonic versus ileocolonic anastomoses, and PN dependence compared with those who weaned from PN. Stool and serum BA composition and C4 concentrations were also altered in patients with SBS, reflecting adaptive changes in enterohepatic BA cycling. Stools from patients who were weaned from PN were enriched in secondary BAs including deoxycholic acid and lithocholic aicd. Shifts in gut microbiota and BA metabolites may generate a favorable luminal environment in select patients with SBS, promoting the ability to wean from PN. Proadaptive microbial species and select BA may provide novel targets for patient-specific therapies for SBS.NEW & NOTEWORTHY Loss of intestinal surface area causes short bowel syndrome, intestinal failure, and parenteral nutrition dependence. We analyzed the gut microbiota and bile acid metabolome of a large cohort of short bowel syndrome adult patients with different postsurgical anatomies. We report a novel analysis of the microbiome of patients with ileostomy and jejunostomy. Enrichment of specific microbial and bile acid species may be associated with the ability to wean from parenteral nutrition.
Collapse
Affiliation(s)
- Harold J. Boutte
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jacqueline Chen
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Todd N. Wylie
- 2Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri,3McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Kristine M. Wylie
- 2Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri,3McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Yan Xie
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mackenzie Geisman
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Anirudh Prabu
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Vered Gazit
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Phillip I. Tarr
- 2Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri,4Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri
| | - Marc S. Levin
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri,7Veterans Administration Saint Louis Health Care System, St. Louis, Missouri
| | - Brad W. Warner
- 5Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Nicholas O. Davidson
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri,6Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| | - Deborah C. Rubin
- 1Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri,6Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
19
|
Wang Y, Zheng L, Zhou Z, Yao D, Huang Y, Liu B, Duan Y, Li Y. Review article: insights into the bile acid-gut microbiota axis in intestinal failure-associated liver disease-redefining the treatment approach. Aliment Pharmacol Ther 2022; 55:49-63. [PMID: 34713470 DOI: 10.1111/apt.16676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/04/2021] [Accepted: 10/15/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Intestinal failure-associated liver disease (IFALD) increases mortality of patients with intestinal failure (IF), but lacks effective prevention or treatment approaches. Bile acids, gut microbiota and the host have close and complex interactions, which play a central role in modulating host immune and metabolic homeostasis. Increasing evidence suggests that derangement of the bile acid-gut microbiota (BA-GM) axis contributes to the development of IFALD. AIMS To review the BA-GM axis in the pathogenesis and clinical applications of IFALD, and to explore future directions for effective disease management. METHODS We conducted a literature search on bile acid and gut microbiota in IF and liver diseases. RESULTS The BA-GM axis demonstrates a unique IF signature manifesting as an increase in primary-to-secondary bile acids ratio, disturbed enterohepatic circulation, blunted bile acid signalling pathways, gut microbial dysbiosis, and altered microbial metabolic outputs. Bile acids and gut microbiota shape the compositional and functional alterations of each other in IF; collaboratively, they promote immune dysfunction and metabolic aberration in the liver. Diagnostic markers and treatments targeting the BA-GM axis showed promising potential in the management of IFALD. CONCLUSIONS Bile acids and gut microbiota play a central role in the development of IFALD and make attractive biomarkers as well as therapeutic targets. A multitarget, individualised therapy aiming at different parts of the BA-GM axis may provide optimal clinical benefits and requires future investigation.
Collapse
Affiliation(s)
- Yaoxuan Wang
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Lei Zheng
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Zhiyuan Zhou
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Danhua Yao
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Yuhua Huang
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Bin Liu
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Yantao Duan
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Yousheng Li
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| |
Collapse
|
20
|
Josyabhatla R, Imseis EM. Pediatric intestinal failure and the microbiome. Semin Perinatol 2021; 45:151453. [PMID: 34332780 DOI: 10.1016/j.semperi.2021.151453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Neonatal intestinal failure is a complex medical condition that is associated with the need for long term parenteral nutrition and its associated complications. The microbiome in this diseased state is different from what is now understood to be a healthy microbiome. The effect of this dysbiotic microbiome on the complications of intestinal failure are only starting to be understood. The ability to modulate the microbiome with enteral/parenteral nutrients, as well probiotics to a healthier state, is an exciting opportunity that holds promise.
Collapse
Affiliation(s)
- Rohit Josyabhatla
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, University of Texas Health Science Center at Houston, TX, USA
| | - Essam M Imseis
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, University of Texas Health Science Center at Houston, TX, USA.
| |
Collapse
|
21
|
Phyo LY, Singkhamanan K, Laochareonsuk W, Surachat K, Phutong N, Boonsanit K, Chiengkriwate P, Sangkhathat S. Fecal microbiome alterations in pediatric patients with short bowel syndrome receiving a rotating cycle of gastrointestinal prophylactic antibiotics. Pediatr Surg Int 2021; 37:1371-1381. [PMID: 34156541 DOI: 10.1007/s00383-021-04948-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pediatric patients with short bowel syndrome (SBS) are at risk of developing small intestinal bacterial overgrowth (SIBO). Prevention of SIBO using cyclic enteric antibiotics has been implemented to control the balance in microbial ecosystems, although its effectiveness has not been well studied. PURPOSE This study aimed to explore the change in the gut microbial composition in SBS patients during cyclic antibiotic phases and antibiotic-free period, and to compare the microbiota composition between healthy controls and SBS patients. METHOD SBS patients taking oral metronidazole alternating with trimethoprim-sulfamethoxazole (TMP-SMT) and antibiotic-free conditions as a '10-day cyclic protocol' were involved in fecal microbiome study using Illumina 16S sequencing. RESULTS When healthy control possessed the majority of Bacteroidetes spp. (54%) and Firmicutes spp. (33%), the microbial composition in SBS patients especially Firmicutes spp. and Proteobacteria spp. was prominently changed in each phase of treatment. In antibiotic-free period, SBS patients displayed 49% Firmicutes and 36% Proteobacteria. However, higher Proteobacteria than Firmicutes were detected at the commencement of metronidazole (58% versus 33%). Similarly, 56% Proteobacteria and 27% Firmicutes were found during TMP-SMT. Escherichia coli increased prominently during the antibiotic periods. CONCLUSION Prophylactic antibiotics change the gut microbiota composition in an unfavorable direction, especially when repeatedly used for a long period. This practice should be reconsidered. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
- Le Yi Phyo
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
| | - Kamonnut Singkhamanan
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
| | - Wison Laochareonsuk
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
| | - Komwit Surachat
- Information and Communication Technology, Faculty of Science, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
| | - Napasorn Phutong
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
| | - Kaimook Boonsanit
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
| | - Piyawan Chiengkriwate
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
| | - Surasak Sangkhathat
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand.
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand.
| |
Collapse
|
22
|
Therapeutic potential of an intestinotrophic hormone, glucagon-like peptide 2, for treatment of type 2 short bowel syndrome rats with intestinal bacterial and fungal dysbiosis. BMC Infect Dis 2021; 21:583. [PMID: 34134659 PMCID: PMC8207711 DOI: 10.1186/s12879-021-06270-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/02/2021] [Indexed: 12/21/2022] Open
Abstract
Background Previous studies showed that type 2 short bowel syndrome (SBS) rats were accompanied by severe intestinal bacterial dysbiosis. Limited data are available for intestinal fungal dysbiosis. Moreover, no effective therapeutic drugs are available for these microbiota dysbiosis. The aims of our study were to investigate the therapeutic potential of glucagon-like peptide 2 (GLP-2) for these microbiota dysbiosis in type 2 SBS rats. Methods 8-week-old male SD rats which underwent 80% small bowel resection, ileocecum resection, partial colon resection and jejunocolostomy, were treated with saline (SBS group, n = 5) or GLP-2 (GLP2.SBS group, n = 5). The Sham group rats which underwent transection and re-anastomosis were given a saline placebo (Sham group, n = 5). 16S rRNA and ITS sequencing were applied to evaluate the colonic bacterial and fungal composition at 22 days after surgery, respectively. Results The relative abundance of Actinobacteria, Firmicutes and proinflammatory Proteobacteria increased significantly in SBS group rats, while the relative abundance of Bacteroidetes, Verrucomicrobia and Tenericutes decreased remarkably. GLP-2 treatment significantly decreased Proteus and increased Clostridium relative to the saline treated SBS rats. The diversity of intestinal fungi was significantly increased in SBS rats, accompanied with some fungi abnormally increased and some resident fungi (e.g., Penicillium) significantly decreased. GLP-2 treatment significantly decreased Debaryomyces and Meyerozyma, and increased Penicillium. Moreover, GLP-2 partially restored the bacteria-fungi interkingdom interaction network of SBS rats. Conclusion Our study confirms the bacterial and fungal dysbiosis in type 2 SBS rats, and GLP-2 partially ameliorated these microbiota dysbiosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06270-w.
Collapse
|
23
|
Neelis EG, de Koning BAE, Hulst JM, Papadopoulou R, Kerbiriou C, Rings EHHM, Wijnen RMH, Nichols B, Gerasimidis K. Gut microbiota and its diet-related activity in children with intestinal failure receiving long-term parenteral nutrition. JPEN J Parenter Enteral Nutr 2021; 46:693-708. [PMID: 33982321 PMCID: PMC9255855 DOI: 10.1002/jpen.2188] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background This study characterized gut microbiota and its diet‐related activity in children with intestinal failure (IF) receiving parenteral nutrition (PN) compared with those of healthy controls (HC) and in relation to disease characteristics. Methods The fecal microbiota and short‐chain fatty acids (SCFAs) were measured in 15 IF patients (n = 68) and 25 HC (n = 25). Results Patients with IF had a lower bacterial load (P = .003), diversity (P < .001), evenness (P < .001) and richness (P = 0.006) than HC. Patients with surgical IF had lower diversity (P < .039) than those with functional IF. Propionic acid and butyric acid (p < .001) were lower and d‐lactate and l‐lactate were higher (p < 0.001) in IF patients than in HC. The energy supplied by PN (%PN) was negatively associated with microbiota diversity and SCFA profile. IF patients had more Escherichia‐Shigella (P = .006), Cronobacter (P = .001), and Staphylococcus (Operational Taxonomic Unit 14, P < .001) and less Faecalibacterium (P < 0.001) and Ruminococcus 1 and 2 (P < .001). Duration of PN (P = .005), %PN (P = .005), and fiber intake (P = .011) were predictive of microbiota structure. Higher intake of enteral nutrition was associated with microbiota structure and function closer to those of HC. Conclusions Microbiota composition and its diet‐related function are altered in IF, with depletion of beneficial SCFAs and species and supraphysiological increase of potentially harmful pathobionts. The influence of this compositional and functional microbial dysbiosis on patients’ outcomes and management warrants further exploration.
Collapse
Affiliation(s)
- Esther G Neelis
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Barbara A E de Koning
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Jessie M Hulst
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rodanthi Papadopoulou
- Human Nutrition, School of Medicine, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Caroline Kerbiriou
- Human Nutrition, School of Medicine, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Edmond H H M Rings
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatric Gastroenterology, Leiden University Medical Center-Willem Alexander Children's Hospital, Leiden, The Netherlands
| | - René M H Wijnen
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ben Nichols
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Konstantinos Gerasimidis
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Fousekis FS, Mitselos IV, Christodoulou DK. New insights into intestinal failure-associated liver disease in adults: A comprehensive review of the literature. Saudi J Gastroenterol 2021; 27:3-12. [PMID: 33642350 PMCID: PMC8083246 DOI: 10.4103/sjg.sjg_551_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intestinal failure-associated liver disease (IFALD) remains one of the most common and serious complications of parenteral nutrition (PN), causing a wide spectrum of hepatic manifestations from steatosis and mild cholestasis to portal hypertension and end-stage liver failure. The prevalence of IFALD depends on the diagnostic criteria and ranges from 4.3% to 65%. Moreover, many factors are shown to contribute to its development, including nutrient deficiencies, toxicity of PN, infections, and alterations of bile acid metabolism and gut microbiota. Prevention and management of IFALD aim at ameliorating or eliminating the risk factors associated with IFALD. The use of PN formulations with a lower ratio omega-6-to-omega-3 polyunsaturated fatty acids, cycle PN, optimization of enteral stimulation and prevention and early treatment of infections constitute the main therapeutic targets. However, failure of improvement and severe IFALD with end-stage liver failure should be considered as the indications of intestinal transplantation. The aim of this review is to provide an update of the epidemiology, pathophysiology, and diagnosis of IFALD in the adult population as well as to present a clinical approach of the therapeutic strategies of IFALD and present novel therapeutic targets.
Collapse
Affiliation(s)
- Fotios S. Fousekis
- Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Ioannis V. Mitselos
- Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Dimitrios K. Christodoulou
- Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, Greece,Address for correspondence: Prof. Dimitrios K. Christodoulou, Professor of Gastroenterology, Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, POBox 1186, Ioannina 45110, Greece. E-mail:
| |
Collapse
|
25
|
Höllwarth ME, Solari V. Nutritional and pharmacological strategy in children with short bowel syndrome. Pediatr Surg Int 2021; 37:1-15. [PMID: 33392698 DOI: 10.1007/s00383-020-04781-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Short bowel syndrome in neonates is a severe and life-threatening disease after a major loss of small bowel with or without large bowel. Intestinal adaptation, by which the organism tries to restore digestive and absorptive capacities, is entirely dependent on stimulation of the active enterocytes by enteral nutrition. This review summarizes recent knowledge about the pathophysiologic consequences after the loss of different intestinal parts and outlines the options for enteral nutrition and pharmacological therapies to support the adaptation process.
Collapse
Affiliation(s)
- Michael E Höllwarth
- Univ. Clinic of Pediatric and Adolescent Surgery, Medical University, Graz, Austria.
| | - Valeria Solari
- Department of Pediatric Surgery, Klinik Donaustadt, 1220, Vienna, Austria
| |
Collapse
|
26
|
Andersen S, Banks M, Bauer J. Nutrition Support and the Gastrointestinal Microbiota: A Systematic Review. J Acad Nutr Diet 2020; 120:1498-1516. [PMID: 32682806 DOI: 10.1016/j.jand.2020.04.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Low microbial diversity or altered microbiota composition is associated with many disease states. In the treatment of many conditions, enteral (EN) or parenteral (PN) nutrition is frequently required. OBJECTIVE This systematic review aimed to identify and evaluate the evidence of the effect of EN vs PN on the gastrointestinal microbiota. METHOD A comprehensive systematic literature search of 5 databases was completed to review studies published until February 2020. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were utilized in completion of the review with the Academy of Nutrition and Dietetics quality criteria checklist and Grading of Recommendations Assessment, Development and Evaluation to evaluate the included studies. The review was registered on PROSPERO (CRD42018091328). Studies were eligible for inclusion if participants were older than 3 years, patients received either EN, PN or both, with some patients receiving each mode of nutrition support. The main outcome was any assessment of the gastrointestinal microbiota, including diversity or taxa abundance. RESULTS Eleven articles (n = 367 patients) met the inclusion criteria and were evaluated. Seven studies (n = 237) reported greater abundance of Proteobacteria with the provision of PN compared to EN; 6 studies (n = 172) reported lower Firmicutes and 5 studies (n = 155) lower Bacteroidetes. In 7 studies (n = 282), microbial diversity was lower with provision of PN than EN. The Grading of Recommendations Assessment, Development and Evaluation certainty of evidence was very low. CONCLUSIONS Provision of PN may lead to greater abundance of Proteobacteria and reduced microbial diversity; however, there is limited literature on this topic and additional research is warranted to improve understanding of the impact of EN vs PN on the microbiota.
Collapse
|
27
|
Qian T, Zhu H, Zhu L, Chen C, Shen C, Zhang R. Small intestinal microbiota composition and the prognosis of infants with ileostomy resulting from distinct primary diseases. BMC Gastroenterol 2020; 20:224. [PMID: 32660555 PMCID: PMC7359560 DOI: 10.1186/s12876-020-01366-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 07/02/2020] [Indexed: 11/10/2022] Open
Abstract
Background Studies of microbiota composition of infants with small intestinal ostomy due to various etiologies are limited. Here, we characterized the intestinal microbiota of neonates with ileostomy resulting from distinct primary diseases. Methods Fifteen patients with necrotizing enterocolitis, eight patients with meconium peritonitis, and seven patients with Hirschsprung’s disease were included in the study. The small intestinal microbiota composition in infants with ileostomy caused by these diseases was studied. Results Microbial diversity in neonatal ileostomy fluid was generally low, and was dominated by members of the Proteobacteria and Firmicutes phyla. At the genus level, the most abundant were Klebsiella, Escherichia-Shigella, Streptococcus, Clostridium sensu stricto 1, Enterococcus, and Lactobacillus. Streptococcus and Veillonella are related to carbohydrate metabolism and immunity, and breastfeeding can increase the proportion of these potentially beneficial bacteria. The proportion of Bifidobacterium in the breastfeeding group was higher than that in the non-breastfeeding group, and incidence of colitis and sepsis was reduced in the breastfeeding group. The proportion of Bifidobacterium increased and incidence of colitis and sepsis decreased in the breastfeeding group compared with the non- breastfeeding group, but there was no significant difference. The increase in body weight in the breastfeeding group was observed to be higher than in the non-breastfeeding group. Conclusions Excessive Klebsiella and Escherichia-Shigella and low abundance of Streptococcus, Veillonella and Faecalibacterium suggests that the small intestinal microbiota is in an unhealthy state after ileostomy. However, Streptococcus, Faecalibacterium, and Veillonella species were frequently present, suggesting that expansion of these bacteria might assist the development of the immune system after surgery.
Collapse
Affiliation(s)
- Tian Qian
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Rd, Shanghai, 201102, P. R. China.,Department of Clinical Nutrition, Children's Hospital of Fudan University, 399 Wanyuan Rd, Shanghai, 201102, P. R. China
| | - Haitao Zhu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wanyuan Rd, Shanghai, 201102, P. R. China
| | - Li Zhu
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Rd, Shanghai, 201102, P. R. China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Rd, Shanghai, 201102, P. R. China
| | - Chun Shen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wanyuan Rd, Shanghai, 201102, P. R. China.
| | - Rong Zhang
- Department of Neonatology, Children's Hospital of Fudan University, 399 Wanyuan Rd, Shanghai, 201102, P. R. China.
| |
Collapse
|
28
|
Zhang T, Wang Y, Yan W, Lu L, Tao Y, Jia J, Cai W. Microbial alteration of small bowel stoma effluents and colonic feces in infants with short bowel syndrome. J Pediatr Surg 2020; 55:1366-1372. [PMID: 31493882 DOI: 10.1016/j.jpedsurg.2019.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/22/2019] [Accepted: 08/09/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIM Studies about differences in microbial communities between the small intestine and colon in infants with short bowel syndrome (SBS) are rare. We aimed to characterize the bacterial diversity of small bowel stoma effluents and feces of SBS infants. METHODS Seven SBS infants were enrolled in this study and provided two samples (one from the stoma and the other from the anus) each. Eleven age-matched healthy controls were recruited to provide one fecal sample each. 16S rRNA gene MiSeq sequencing was conducted to characterize the microbiota diversity and composition. RESULTS The bacterial diversity of the stoma effluents was significantly higher than that in the feces of SBS infants. Proteobacteria dominated in both the stoma effluents and colonic. Acinetobacter (P = 0.004), Klebsiella (P = 0.015), Citrobacter (P = 0.019), and Lactobacillus (P = 0.030) were more abundant in stoma effluents compared to feces of SBS patients, while Bacteroidetes, Bifidobacterium and Veillonella were less abundant in stoma effluents. Significantly higher levels of Proteobacteria, Enterococcus and lower levels of Blautia, Collinsella, Faecalibacterium, Veillonella were present in the fecal samples of SBS patients than those in the healthy controls. Kyoto Encyclopedia of Genes and Genomes pathways related to metabolism and membrane function were depleted in SBS patients. CONCLUSIONS The predominant intestinal bacterial groups were different in SBS children before and after the fistula closure. Fecal samples of SBS patients featured overabundant Proteobacteria and less SCFA producing bacteria. Depleted functional profiles of the microbiome were found in fecal samples of SBS patients. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Ying Wang
- Division of Pediatric GI and Nutrition, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Weihui Yan
- Division of Pediatric GI and Nutrition, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Lina Lu
- Division of Pediatric GI and Nutrition, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yijing Tao
- Division of Pediatric GI and Nutrition, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Jie Jia
- Department of Nutrition, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| | - Wei Cai
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.
| |
Collapse
|
29
|
Piper HG, Coughlin LA, Nguyen V, Channabasappa N, Koh AY. A comparison of small bowel and fecal microbiota in children with short bowel syndrome. J Pediatr Surg 2020; 55:878-882. [PMID: 32063370 DOI: 10.1016/j.jpedsurg.2020.01.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 01/25/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Babies with short bowel syndrome (SBS) have small intestinal microbial disturbances that impact gut function. Characterizing the small bowel microbiota is challenging, and the utility of sampling stool is unclear. This study compares the microbiota from fecal samples and the small bowel. METHODS Stool samples were collected (2016-2017) from infants with SBS and colon in continuity (COLON) or SBS with small bowel ostomy (sbSTOMA). The abundance and quantity of major bacterial genera was compared between groups and to healthy controls using 16S rRNA sequencing and qPCR. Kruskall-Wallis test was used for analysis with P values <0.05 considered significant. RESULTS Samples (n = 41) were collected from 15 SBS infants (<2 years) (9 sbSTOMA, 6 COLON) and 3 healthy infants. Demographics and small intestinal length did not differ between sbSTOMA and COLON infants. The microbiota of SBS groups differed significantly from healthy controls. Fecal samples contained higher quantities of bacteria, but there were no significant differences between sbSTOMA and COLON groups in the abundance of facultative or obligate anaerobes, anti-inflammatory Clostridia, Enterobacteriaceae, or Bifidobacterium. CONCLUSION Infants with SBS have disturbances to their intestinal microbiota. Sampling small intestinal effluent is challenging. Stool samples may provide a window into the more proximal microbial community. TYPE OF STUDY Diagnostic. LEVEL OF EVIDENCE Level II.
Collapse
Affiliation(s)
- Hannah G Piper
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| | - Laura A Coughlin
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Van Nguyen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nandini Channabasappa
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew Y Koh
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
30
|
Zhang T, Liu Y, Yan JK, Cai W. Early downregulation of P-glycoprotein facilitates bacterial attachment to intestinal epithelial cells and thereby triggers barrier dysfunction in a rodent model of total parenteral nutrition. FASEB J 2020; 34:4670-4683. [PMID: 32027421 DOI: 10.1096/fj.201902513r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/08/2020] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
Intestinal barrier dysfunction is a major complication of total parenteral nutrition (TPN). Our preliminary study revealed that intestinal P-glycoprotein (P-gp) was significantly downregulated under TPN treatment followed by disruption of barrier function, and thus the significance of early downregulation of P-gp needs to be addressed. Herein, we report a pivotal role of P-gp in the development of intestinal barrier dysfunction under TPN. Functional suppression of P-gp may facilitate bacterial attachment to intestinal epithelial cells (IECs) and thereby induce degradation of tight junctions to trigger barrier dysfunction. By using a rat model of TPN, we found early downregulation of P-gp function in ileum after 3-day TPN, followed by disruption of barrier function after 7-day TPN. By using Escherichia coli (E. coli) k88 and DH5α as type strains, we found significantly increased bacterial attachment to IECs in TPN group compared to sham. By using Caco-2 cells as an IEC model in vitro, we found that functional suppression of P-gp remarkably facilitated bacterial attachment to Caco-2 cells, leading to subsequent disruption of intestinal barrier function. Of note, Occludin was significantly downregulated by bacterial attachment when P-gp was functionally suppressed. Mechanistically, changes on Occludin were attributed to enhanced protein degradation instead of suppressed protein translation. Despite the half-life of Occludin protein being unchanged by DH5α treatment alone, it was decreased by about 40% when P-gp was simultaneously suppressed. Taken together, our findings revealed that early downregulation of intestinal P-gp under TPN may be a potential therapeutic target to prevent the development of barrier dysfunction.
Collapse
Affiliation(s)
- Tian Zhang
- School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Liu
- School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun-Kai Yan
- School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai Institute for Pediatric Research, Shanghai, China
| | - Wei Cai
- School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai Institute for Pediatric Research, Shanghai, China
| |
Collapse
|
31
|
Severe Intestinal Dysbiosis in Rat Models of Short Bowel Syndrome with Ileocecal Resection. Dig Dis Sci 2020; 65:431-441. [PMID: 31441001 DOI: 10.1007/s10620-019-05802-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Short bowel syndrome (SBS) resulting from extensive intestinal resection is thought to significantly affect gut microbiota. Data are limited on the signatures of the intestinal microbiome in SBS with different anatomical types. AIMS The aim of our investigation was to characterize the composition and function of gut microbiota in SBS with or without ileocecal resection (ICR). METHODS Six-week-old male Sprague-Dawley rats underwent 75% small bowel resection (SBR) with the ileocecal junction intact (SBR group, jejunoileal anastomosis, n = 10) or removed (ICR group, jejunocolic anastomosis, n = 10), or sham surgery (sham group, n = 10). Colonic contents of the rats were collected 28 days after operation, and 16S rRNA gene sequencing was performed on the MiSeq Illumina platform to analyze bacterial composition. RESULTS Overall structures of the gut microbiome differed significantly among the three groups. The bacterial α-diversity of the ICR group was remarkably lower than that of the sham group. ICR rats were enriched with Lactobacillus and opportunistic pathogens from Proteobacteria but depleted of commensal genera belonging to the Lachnospiraceae, Ruminococcaceae and Erysipelotrichaceae families. Genera from the Bacteroidales S24-7 group, Porphyromonadaceae, Prevotellaceae, Rikenellaceae and Christensenellaceae were prevalent in SBR rats. Functional pathways of branched-chain and aromatic amino acid biosynthesis, lipopolysaccharide biosynthesis and infectious diseases were abundant in the ICR group, while SBR rats featured pathways of C5 branched dibasic acid metabolism, biotin metabolism and one carbon pool folate. CONCLUSIONS ICR causes dramatically more severe intestinal dysbiosis than SBR only in SBS rat models, resulting in altered functional profiles of the gut microbiome.
Collapse
|
32
|
Fecal dysbiosis in infants with cystic fibrosis is associated with early linear growth failure. Nat Med 2020; 26:215-221. [PMID: 31959989 PMCID: PMC7018602 DOI: 10.1038/s41591-019-0714-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022]
Abstract
Most infants with cystic fibrosis (CF) have pancreatic exocrine insufficiency that results in nutrient malabsorption and requires oral pancreatic enzyme replacement. Newborn screening for CF has enabled earlier diagnosis, nutritional intervention, and enzyme replacement for these infants, allowing most infants with CF to achieve their weight goals by 12 months of age1. Nevertheless, most infants with CF continue to have poor linear growth during their first year of life1. Although this early linear growth failure is associated with worse long-term respiratory function and survival2,3, the determinants of stature in infants with CF have not been defined. Several characteristics of the CF gastrointestinal (GI) tract, including inflammation, maldigestion and malabsorption, could promote intestinal dysbiosis4,5. As GI microbiome activities are known to affect endocrine functions6,7, the intestinal microbiome of infants with CF might also impact growth. We identified an early, progressive fecal dysbiosis that distinguished infants with CF and low length from infants with CF and normal length. This dysbiosis included altered abundances of taxa that perform functions important for GI health, nutrient harvest, and growth hormone signaling, including decreased Bacteroidetes and increased Proteobacteria. Thus, the GI microbiota represent a potential therapeutic target to correct linear growth defects among infants with CF.
Collapse
|
33
|
de Laffolie J, Sheridan D, Reinshagen K, Wessel L, Zimmermann C, Stricker S, Lerch MM, Weigel M, Hain T, Domann E, Rudloff S, Nichols BL, Naim HY, Zimmer KP. Digestive enzyme expression in the large intestine of children with short bowel syndrome in a late stage of adaptation. FASEB J 2020; 34:3983-3995. [PMID: 31957074 DOI: 10.1096/fj.201901758rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND AIMS Intestinal adaptation in short bowel syndrome (SBS) includes morphologic processes and functional mechanisms. This study investigated whether digestive enzyme expression in the duodenum and colon is upregulated in SBS patients. METHOD Sucrase-isomaltase (SI), lactase-phlorizin hydrolase (LPH), and neutral Aminopeptidase N (ApN) were analyzed in duodenal and colonic biopsies from nine SBS patients in a late stage of adaptation as well as healthy and disease controls by immunoelectron microscopy (IEM), Western blots, and enzyme activities. Furthermore, proliferation rates and intestinal microbiota were analyzed in the mucosal specimen. RESULTS We found significantly increased amounts of SI, LPH, and ApN in colonocytes in most SBS patients with large variation and strongest effect for SI and ApN. Digestive enzyme expression was only partially elevated in duodenal enterocytes due to a low proliferation level measured by Ki-67 staining. Microbiome analysis revealed high amounts of Lactobacillus resp. low amounts of Proteobacteria in SBS patients with preservation of colon and ileocecal valve. Colonic expression was associated with a better clinical course in single cases. CONCLUSION In SBS patients disaccharidases and peptidases can be upregulated in the colon. Stimulation of this colonic intestinalization process by drugs, nutrients, and pre- or probiotics might offer better therapeutic approaches.
Collapse
Affiliation(s)
- Jan de Laffolie
- Department of Paediatrics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Diana Sheridan
- Department of Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, UKE: University Hospital Eppendorf, Altona Children's Hospital, Hamburg, Germany
| | - Lucas Wessel
- Department of Pediatric Surgery, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | | | - Sebastian Stricker
- Department of Paediatrics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Markus M Lerch
- Department of Internal Medicine A, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Markus Weigel
- Institute of Medical Microbiology, Justus-Liebig-University Giessen, Giessen, Germany.,German Centre for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Justus Liebig University Giessen, Giessen, Germany
| | - Torsten Hain
- Institute of Medical Microbiology, Justus-Liebig-University Giessen, Giessen, Germany.,German Centre for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Justus Liebig University Giessen, Giessen, Germany
| | - Eugen Domann
- Institute of Medical Microbiology, Justus-Liebig-University Giessen, Giessen, Germany.,German Centre for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Justus Liebig University Giessen, Giessen, Germany
| | - Silvia Rudloff
- Department of Paediatrics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Buford L Nichols
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Hassan Y Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Klaus-Peter Zimmer
- Department of Paediatrics, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
34
|
Alomari M, Nusairat L, Al Momani L, Chadalavada P, Covut F, Olayan M, Young M, Romero-Marrero C. Effects of Probiotics on Intestinal Failure-Associated Liver Disease in Adult Patients Receiving Prolonged Parenteral Support: A Tertiary Care Center Experience. Nutr Clin Pract 2019; 35:454-463. [PMID: 31721285 DOI: 10.1002/ncp.10437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND It has been hypothesized that dysbiosis plays a significant role in the pathogenesis of intestinal failure-associated liver disease (IFALD). Therefore, we aimed to investigate the effect of probiotics on IFALD in patients receiving parenteral support, namely home parenteral nutrition (HPN) and home intravenous fluids (HIVFs). METHODS We retrospectively reviewed charts of patients with intestinal failure who received HPN or HIVF for >2 weeks at our tertiary center between January 2005 and August 2016. We excluded patients <18 years of age, patients with other causes of liver disease, patients who used probiotics for <30 days, patients with <6 months' follow-up, and those who had long-term antibiotic use (>30 days). Bivariable and multivariable logistic regression analyses were used in this study. RESULTS A total of 282 patients who received parenteral support were included. Eighty-five percent of our sample received PN. A total of 78 (27.7%) patients used probiotics. The prevalence of IFALD in patients who used probiotics was 35.9% vs 54.4% in patients who did not use probiotics, P = .005. In multivariable analysis, only small-bowel length of 10-90 cm and HPN use showed a significant impact on IFALD, odds ratio (OR) = 4.394 (95% confidence interval [CI], 1.635-11.814; P = .003) and OR = 4.502 (95% CI 1.412-14.351; P = .011), respectively. CONCLUSION Our study revealed that the prevalence of IFALD was comparable among the probiotic users and nonusers. Only small bowel length of 1090 cm and HPN use showed a significant impact on IFALD.
Collapse
Affiliation(s)
- Mohammad Alomari
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Leen Nusairat
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Laith Al Momani
- Department of Internal Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | | | - Fahrettin Covut
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - May Olayan
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Mark Young
- Department of Gastroenterology and Hepatology, East Tennessee State University, Johnson City, Tennessee, USA
| | - Carlos Romero-Marrero
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Dowhaniuk JK, Szamosi J, Chorlton S, Owens J, Mileski H, Clause R, Pernica JM, Bowdish DME, Surette MG, Ratcliffe EM. Starving the Gut: A Deficit of Butyrate in the Intestinal Ecosystem of Children With Intestinal Failure. JPEN J Parenter Enteral Nutr 2019; 44:1112-1123. [DOI: 10.1002/jpen.1715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/25/2019] [Accepted: 09/08/2019] [Indexed: 12/14/2022]
Affiliation(s)
| | - Jake Szamosi
- Department of Medicine and Biochemistry and Biomedical SciencesMcMaster University Hamilton Ontario Canada
| | - Sam Chorlton
- Department of Undergraduate MedicineMcMaster University Hamilton Ontario Canada
| | - Jillian Owens
- Division of Pediatric Gastroenterology and NutritionMcMaster Children's Hospital Hamilton Ontario Canada
| | - Heather Mileski
- Division of Pediatric Gastroenterology and NutritionMcMaster Children's Hospital Hamilton Ontario Canada
| | - Rose‐Frances Clause
- Division of Pediatric Gastroenterology and NutritionMcMaster Children's Hospital Hamilton Ontario Canada
| | | | - Dawn M. E. Bowdish
- Department of Pathology & Molecular MedicineMcMaster University Hamilton Ontario Canada
| | - Michael G. Surette
- Department of Medicine and Biochemistry and Biomedical SciencesMcMaster University Hamilton Ontario Canada
| | | |
Collapse
|
36
|
Fecal Microbiomes in Premature Infants With and Without Parenteral Nutrition-Associated Cholestasis. J Pediatr Gastroenterol Nutr 2019; 69:224-230. [PMID: 31058777 DOI: 10.1097/mpg.0000000000002352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Premature infants often require parenteral nutrition (PN) until they reach enteral autonomy which puts them at risk of developing PN-associated cholestasis (PNAC). We sought to compare longitudinal changes in fecal microbiomes of premature infants who developed PNAC versus those who did not despite being on similar PN doses. METHODS Stool samples from premature infants (gestational age <30 weeks) who developed direct bilirubin ≥1.5 mg/dL while receiving PN were classified as precholestasis, cholestasis, or postcholestasis based on bilirubin levels at the time of sample acquisition and were compared to matched control groups 1, 2, and 3, respectively. RESULTS A total of 102 fecal samples from 8 cases and 10 controls were analyzed. Precholestasis samples were more abundant in phylum Firmicutes and genus Staphylococcus, whereas control 1 was more abundant in phylum Proteobacteria and genus Escherichia-Shigella. Nonmetric multidimensional scaling ordination plots based on the taxonomic composition of early fecal samples revealed significant separation between cases and controls. On indicator species analysis, genus Bacilli was more prevalent in samples from the precholestasis group, whereas genus Escherichia-Shigella was more prevalent in control 1. With feeding advances, weaning of PN and resolution of PNAC, most differences in microbiota resolved with the exception of control 3 group being more diverse compared to the postcholestasis group. CONCLUSIONS Premature neonates who develop PNAC, compared to those who do not, show significantly different fecal microbiomes preceding the biochemical detection of cholestasis.
Collapse
|
37
|
Longitudinal changes in the gut microbiome of infants on total parenteral nutrition. Pediatr Res 2019; 86:107-114. [PMID: 30965357 PMCID: PMC6594895 DOI: 10.1038/s41390-019-0391-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/13/2019] [Accepted: 04/01/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Animal studies suggest that total parenteral nutrition (TPN) may alter bacterial colonization of the intestinal tract and contribute to complications. Progressive changes in gut microbiome of infants receiving TPN are not well understood. METHODS Infants with and without TPN/soy lipid were enrolled in a prospective, longitudinal study. Weekly fecal samples were obtained for the first 4 weeks of life. High throughput pyrosequencing of 16S rDNA was used for compositional analysis of the gut microbiome. RESULTS 47 infants were eligible for analyses, 25 infants received TPN, and 22 infants did not (control). Although similar between TPN and control groups in the first week, fecal bacterial alpha diversity was significantly lower in the TPN group compared to controls at week 4 (Shannon index 1.0 vs 1.5, P-value = 0.03). The TPN group had significantly lower Bacteroidetes and higher Verrucomicrobia abundance compared to controls (P-values < 0.05), and these differences became more pronounced over time. At the genus level, TPN was associated with lower abundance of Bacteroides and Bifidobacterium in all weeks. CONCLUSIONS TPN is associated with significant loss of biodiversity and alterations in the pattern of gut microbial colonization of infants over time. TPN-associated dysbiosis may predispose infants to adverse NICU outcomes.
Collapse
|
38
|
Budinska E, Gojda J, Heczkova M, Bratova M, Dankova H, Wohl P, Bastova H, Lanska V, Kostovcik M, Dastych M, Senkyrik M, Krizova J, Mraz M, Hradecky J, Hajslova J, Lenicek M, Podzimkova K, Chalupsky K, Sedlacek R, Cahova M. Microbiome and Metabolome Profiles Associated With Different Types of Short Bowel Syndrome: Implications for Treatment. JPEN J Parenter Enteral Nutr 2019; 44:105-118. [PMID: 31032975 DOI: 10.1002/jpen.1595] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/23/2019] [Accepted: 04/08/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND The gut microbiome and metabolome may significantly influence clinical outcomes in patients with short bowel syndrome (SBS). The study aimed to describe specific metagenomic/metabolomics profiles of different SBS types and to identify possible therapeutic targets. METHODS Fecal microbiome (FM), volatile organic compounds (VOCs), and bile acid (BA) spectrum were analyzed in parenteral nutrition (PN)-dependent SBS I, SBS II, and PN-independent (non-PN) SBS patients. RESULTS FM in SBS I, SBS II, and non-PN SBS shared characteristic features (depletion of beneficial anaerobes, high abundance of Lactobacilaceae and Enterobacteriaceae). SBS I patients were characterized by the abundance of oxygen-tolerant microrganisms and depletion of strict anaerobes. Non-PN SBS subjects showed markers of partial FM normalization. FM dysbiosis was translated into VOC and BA profiles characteristic for each SBS cohort. A typical signature of all SBS patients comprised high saturated aldehydes and medium-chain fatty acids and reduced short-chain fatty acid (SCFA) content. Particularly, SBS I and II exhibited low protein metabolism intermediate (indole, p-cresol) content despite the hypothetical presence of relevant metabolism pathways. Distinctive non-PN SBS marker was high phenol content. SBS patients' BA fecal spectrum was enriched by chenodeoxycholic and deoxycholic acids and depleted of lithocholic acid. CONCLUSIONS Environmental conditions in SBS gut significantly affect FM composition and metabolic activity. The common feature of diverse SBS subjects is the altered VOC/BA profile and the lack of important products of microbial metabolism. Strategies oriented on the microbiome/metabolome reconstitution and targeted delivery of key compounds may represent a promising therapeutic strategy in SBS patients.
Collapse
Affiliation(s)
- Eva Budinska
- RECETOX, Faculty of Science Masaryk University, Brno, Czech Republic
| | - Jan Gojda
- 2nd Department of Internal Medicine, Kralovske Vinohrady University Hospital and 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marie Heczkova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Miriam Bratova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Helena Dankova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Wohl
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Hana Bastova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vera Lanska
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Kostovcik
- BIOCEV, Institute of Microbiology, AS CR, Vestec, Czech Republic
| | - Milan Dastych
- Department of Gastroenterology and Internal Medicine, University Hospital Brno, Brno, Czech Republic
| | - Michal Senkyrik
- Department of Gastroenterology and Internal Medicine, University Hospital Brno, Brno, Czech Republic
| | - Jarmila Krizova
- 3rd Department of Medicine, Department of Endocrinology and Metabolism, General University Hospital and 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Milos Mraz
- 2nd Department of Internal Medicine, Kralovske Vinohrady University Hospital and 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jaromir Hradecky
- University of Chemistry and Technology, Prague, Czech Republic
- Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Prague, Czech Republic
| | - Jana Hajslova
- University of Chemistry and Technology, Prague, Czech Republic
| | - Martin Lenicek
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kateřina Podzimkova
- Czech Centre for Phenogenomics, Vestec, Czech Republic
- Laboratory of Transgenic Diseases, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Karel Chalupsky
- Czech Centre for Phenogenomics, Vestec, Czech Republic
- Laboratory of Transgenic Diseases, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Vestec, Czech Republic
- Laboratory of Transgenic Diseases, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Monika Cahova
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
39
|
Lauro A, Lacaille F. Short bowel syndrome in children and adults: from rehabilitation to transplantation. Expert Rev Gastroenterol Hepatol 2019; 13:55-70. [PMID: 30791840 DOI: 10.1080/17474124.2019.1541736] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Short bowel syndrome (SBS) is a dramatic clinical condition in both children and adults; the residual bowel length is not sufficient to avoid intestinal failure, with subsequent malnutrition and growth retardation, and intravenous support is required to provide the nutrients normally coming from the intestine. Apart from the primary disease, the medical status can be worsened by complications of intestinal failure: if there are irreversible, the prognosis is poor unless a successful intestinal rehabilitation is achieved. Areas covered: The rescue of the remnant small bowel requires a multidisciplinary expertise to achieve digestive autonomy. The use of intestinal trophic factors has shown encouraging results in improving the intestinal adaptation process. Whenever the residual bowel length is inadequate, in a well-selected population weaning parenteral nutrition (PN) off could be attempted by surgery through lengthening procedures. A further subset of patients, with total and irreversible intestinal failure and severe complications on PN, may have an indication to intestinal transplantation. This procedure is still affected by poor long-term results. Expert commentary: Novel approaches developed through a multidisciplinary team work, such as manipulation of microbiota or tissue bioengineering, should be added to current therapies to treat successfully SBS.
Collapse
Affiliation(s)
- Augusto Lauro
- a Emergency Surgery Department , St. Orsola University Hospital , Bologna , Italy
| | - Florence Lacaille
- b Gastroenterology Hepatology Nutrition Unit , Hôpital Necker-Enfants Malades , Paris , France
| |
Collapse
|
40
|
Role of the Gut⁻Liver Axis in Driving Parenteral Nutrition-Associated Injury. CHILDREN-BASEL 2018; 5:children5100136. [PMID: 30257520 PMCID: PMC6210809 DOI: 10.3390/children5100136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
For decades, parenteral nutrition (PN) has been a successful method for intravenous delivery of nutrition and remains an essential therapy for individuals with intolerance of enteral feedings or impaired gut function. Although the benefits of PN are evident, its use does not come without a significant risk of complications. For instance, parenteral nutrition-associated liver disease (PNALD)—a well-described cholestatic liver injury—and atrophic changes in the gut have both been described in patients receiving PN. Although several mechanisms for these changes have been postulated, data have revealed that the introduction of enteral nutrition may mitigate this injury. This observation has led to the hypothesis that gut-derived signals, originating in response to the presence of luminal contents, may contribute to a decrease in damage to the liver and gut. This review seeks to present the current knowledge regarding the modulation of what is known as the “gut–liver axis” and the gut-derived signals which play a role in PN-associated injury.
Collapse
|
41
|
Neelis E, Koning B, Rings E, Wijnen R, Nichols B, Hulst J, Gerasimidis K. The Gut Microbiome in Patients with Intestinal Failure: Current Evidence and Implications for Clinical Practice. JPEN J Parenter Enteral Nutr 2018; 43:194-205. [DOI: 10.1002/jpen.1423] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Esther Neelis
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Barbara Koning
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Edmond Rings
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
- Paediatric GastroenterologyLeiden University Medical Center–Willem Alexander Children's Hospital Leiden the Netherlands
| | - René Wijnen
- Paediatric SurgeryErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Ben Nichols
- Human NutritionSchool of MedicineCollege of MedicineVeterinary and Life SciencesUniversity of Glasgow Glasgow United Kingdom
| | - Jessie Hulst
- Department of Paediatric GastroenterologyErasmus MC–Sophia Children's Hospital Rotterdam the Netherlands
| | - Konstantinos Gerasimidis
- Human NutritionSchool of MedicineCollege of MedicineVeterinary and Life SciencesUniversity of Glasgow Glasgow United Kingdom
| |
Collapse
|
42
|
Parenteral Nutrition-Associated Liver Disease: The Role of the Gut Microbiota. Nutrients 2017; 9:nu9090987. [PMID: 28880224 PMCID: PMC5622747 DOI: 10.3390/nu9090987] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023] Open
Abstract
Parenteral nutrition (PN) provides life-saving nutritional support in situations where caloric supply via the enteral route cannot cover the necessary needs of the organism. However, it does have serious adverse effects, including parenteral nutrition-associated liver disease (PNALD). The development of liver injury associated with PN is multifactorial, including non-specific intestine inflammation, compromised intestinal permeability, and barrier function associated with increased bacterial translocation, primary and secondary cholangitis, cholelithiasis, short bowel syndrome, disturbance of hepatobiliary circulation, lack of enteral nutrition, shortage of some nutrients (proteins, essential fatty acids, choline, glycine, taurine, carnitine, etc.), and toxicity of components within the nutrition mixture itself (glucose, phytosterols, manganese, aluminium, etc.). Recently, an increasing number of studies have provided evidence that some of these factors are directly or indirectly associated with microbial dysbiosis in the intestine. In this review, we focus on PN-induced changes in the taxonomic and functional composition of the microbiome. We also discuss immune cell and microbial crosstalk during parenteral nutrition, and the implications for the onset and progression of PNALD. Finally, we provide an overview of recent advances in the therapeutic utilisation of pro- and prebiotics for the mitigation of PN-associated liver complications.
Collapse
|