1
|
Trotta RJ, Swanson KC, Klotz JL, Harmon DL. Influence of postruminal casein infusion and exogenous glucagon-like peptide 2 administration on the jejunal mucosal transcriptome in cattle. PLoS One 2024; 19:e0308983. [PMID: 39146343 PMCID: PMC11326568 DOI: 10.1371/journal.pone.0308983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/03/2024] [Indexed: 08/17/2024] Open
Abstract
We previously demonstrated that postruminal casein infusion and exogenous glucagon-like peptide 2 (GLP-2) administration independently stimulated growth and carbohydrase activity of the pancreas and jejunal mucosa in cattle. The objective of the current study was to profile the jejunal mucosal transcriptome of cattle using next-generation RNA sequencing in response to postruminal casein infusion and exogenous GLP-2. Twenty-four Holstein steers [250 ± 23.1 kg body weight (BW)] received a continuous abomasal infusion of 3.94 g raw corn starch/kg of BW combined with either 0 or 1.30 g casein/kg of BW for 7 d. Steers received subcutaneous injections at 0800 and 2000 h to provide either 0 or 100 μg GLP-2/kg of BW per day. At the end of the 7-d treatment period, steers were slaughtered for collection of the jejunal mucosa. Total RNA was extracted from jejunal mucosal tissue, strand-specific cDNA libraries were prepared, and RNA sequencing was conducted to generate 150-bp paired-end reads at a depth of 40 M reads per sample. Differentially expressed genes (DEG), KEGG pathway enrichment, and gene ontology enrichment were determined based on the FDR-corrected P-value (padj). Exogenous GLP-2 administration upregulated (padj < 0.05) 667 genes and downregulated 1,101 genes of the jejunal mucosa. Sphingolipid metabolism, bile secretion, adherens junction, and galactose metabolism were among the top KEGG pathways enriched with upregulated DEG (padj < 0.05) in response to exogenous GLP-2 administration. The top gene ontologies enriched with upregulated DEG (padj < 0.05) in response to exogenous GLP-2 administration included nutrient metabolic processes, brush border and bicellular tight junction assembly, and enzyme and transporter activities. Exogenous GLP-2 administration increased or tended to increase (padj < 0.10) brush border carbohydrase (MGAM, LCT, TREH), hexose transporter (SLC5A1, SLC2A2), and associated transcription factor (HNF1, GATA4, KAT2B) mRNA expression of the jejunal mucosa. Gene ontologies and KEGG pathways that were downregulated (padj < 0.05) in response to exogenous GLP-2 were related to genetic information processing. Postruminal casein infusion downregulated (padj < 0.05) 7 jejunal mucosal genes that collectively did not result in enriched KEGG pathways or gene ontologies. This study highlights some of the transcriptional mechanisms associated with increased growth, starch assimilation capacity, and barrier function of the jejunal mucosa in response to exogenous GLP-2 administration.
Collapse
Affiliation(s)
- Ronald J. Trotta
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kendall C. Swanson
- Department of Animal Science, North Dakota State University, Fargo, North Dakota, United States of America
| | - James L. Klotz
- Forage-Animal Production Research Unit, United States Department of Agriculture, Agricultural Research Service, Lexington, Kentucky, United States of America
| | - David L. Harmon
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
2
|
Stroe MS, Huang MC, Annaert P, Leys K, Smits A, Allegaert K, Van Bockstal L, Valenzuela A, Ayuso M, Van Ginneken C, Van Cruchten S. Drug Disposition in Neonatal Göttingen Minipigs: Exploring Effects of Perinatal Asphyxia and Therapeutic Hypothermia. Drug Metab Dispos 2024; 52:824-835. [PMID: 38906699 DOI: 10.1124/dmd.124.001677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/25/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024] Open
Abstract
Asphyxiated neonates often undergo therapeutic hypothermia (TH) to reduce morbidity and mortality. Since both perinatal asphyxia (PA) and TH influence physiology, altered pharmacokinetics (PK) and pharmacodynamics (PD) are expected. Given that TH is the standard of care for PA with moderate to severe hypoxic-ischemic encephalopathy, disentangling the effect of PA versus TH on PK/PD is not possible in clinical settings. However, animal models can provide insights into this matter. The (neonatal) Göttingen Minipig, the recommended strain for nonclinical drug development, was selected as translational model. Four drugs-midazolam (MDZ), fentanyl (FNT), phenobarbital (PHB), and topiramate (TPM)-were intravenously administered under four conditions: control (C), therapeutic hypothermia (TH), hypoxia (H), and hypoxia plus TH (H+TH). Each group included six healthy male neonatal Göttingen Minipigs anesthetized for 24 hours. Blood samples were drawn at 0 (predose) and 0.5, 2, 2.5, 3, 4, 4.5, 6, 8, 12, and 24 hours post drug administration. Drug plasma concentrations were determined using validated bioanalytical assays. The PK parameters were estimated through compartmental and noncompartmental PK analysis. The study showed a statistically significant decrease in FNT clearance (CL; 66% decrease), with an approximately threefold longer half-life (t1/2) in the TH group. The H+TH group showed a 17% reduction in FNT CL, with a 62% longer t1/2 compared with the C group; however, it was not statistically significant. Although not statistically significant, trends toward lower CL and longer t1/2 were observed in the TH and H+TH groups for MDZ and PHB. Additionally, TPM demonstrated a 28% decrease in CL in the H group compared with controls. SIGNIFICANCE STATEMENT: The overarching goal of this study using the neonatal Göttingen Minipig model was to disentangle the effects of systemic hypoxia and TH on PK using four model drugs. Such insights can subsequently be used to inform and develop a physiologically based pharmacokinetic model, which is useful for drug exposure prediction in human neonates.
Collapse
Affiliation(s)
- Marina-Stefania Stroe
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Miao-Chan Huang
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Pieter Annaert
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Karen Leys
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Anne Smits
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Karel Allegaert
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Lieselotte Van Bockstal
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Allan Valenzuela
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Miriam Ayuso
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Chris Van Ginneken
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| | - Steven Van Cruchten
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium (M.S.-S., L.V.B., A.V., M.A., C.V.G., S.V.C.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (M.-C.H., P.A., K.L.); BioNotus GCV, Niel, Belgium (P.A.); Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Departments of Development and Regeneration (A.S., K.A.) and Pharmaceutical and Pharmacological Sciences (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands (K.A.)
| |
Collapse
|
3
|
Pálsson TG, Gilliam-Vigh H, Jensen BAH, Jeppesen PB, Lund AB, Knop FK, Nielsen CK. Targeting the GLP-2 receptor in the management of obesity. Peptides 2024; 177:171210. [PMID: 38579917 DOI: 10.1016/j.peptides.2024.171210] [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: 12/22/2023] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
Recent advancements in understanding glucagon-like peptide 2 (GLP-2) biology and pharmacology have sparked interest in targeting the GLP-2 receptor (GLP-2R) in the treatment of obesity. GLP-2 is a proglucagon-derived 33-amino acid peptide co-secreted from enteroendocrine L cells along with glucagon-like peptide 1 (GLP-1) and has a range of actions via the GLP-2R, which is particularly expressed in the gastrointestinal tract, the liver, adipose tissue, and the central nervous system (CNS). In humans, GLP-2 evidently induces intestinotrophic effects (i.e., induction of intestinal mucosal proliferation and improved gut barrier function) and promotes mesenteric blood flow. However, GLP-2 does not seem to have appetite or food intake-reducing effects in humans, but its gut barrier-promoting effect may be of interest in the context of obesity. Obesity is associated with reduced gut barrier function, increasing the translocation of proinflammatory gut content to the circulation. This phenomenon constitutes a strong driver of obesity-associated systemic low-grade inflammation, which in turn plays a major role in the development of most obesity-associated complications. Thus, the intestinotrophic and gut barrier-improving effect of GLP-2, which in obese rodent models shows strong anti-inflammatory potential, may, in combination with food intake-reducing strategies, e.g., GLP-1 receptor (GLP-1) agonism, be able to rectify core pathophysiological mechanism of obesity. Here, we provide an overview of GLP-2 physiology in the context of obesity pathophysiology and review the pharmacological potential of GLP-2R activation in the management of obesity and related comorbidities.
Collapse
Affiliation(s)
- Thorir G Pálsson
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Hannah Gilliam-Vigh
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Benjamin A H Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Palle B Jeppesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Intestinal Failure and Liver Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Asger B Lund
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark; Steno Diabetes Center Copenhagen, Copenhagen University Hospital, Herlev, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Copenhagen University Hospital, Herlev, Denmark
| | - Casper K Nielsen
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark.
| |
Collapse
|
4
|
Cheng SY, Jiang L, Wang Y, Cai W. Emerging role of regulated cell death in intestinal failure-associated liver disease. Hepatobiliary Pancreat Dis Int 2024; 23:228-233. [PMID: 36621400 DOI: 10.1016/j.hbpd.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/08/2022] [Indexed: 01/10/2023]
Abstract
Intestinal failure-associated liver disease (IFALD) is a common complication of long-term parenteral nutrition that is associated with significant morbidity and mortality. It is mainly characterized by cholestasis in children and steatohepatitis in adults. Unfortunately, there is no effective approach to prevent or reverse the disease. Regulated cell death (RCD) represents a fundamental biological paradigm that determines the outcome of a variety of liver diseases. Nowadays cell death is reclassified into several types, based on the mechanisms and morphological phenotypes. Emerging evidence has linked different modes of RCD, such as apoptosis, necroptosis, ferroptosis, and pyroptosis to the pathogenesis of liver diseases. Recent studies have shown that different modes of RCD are present in animal models and patients with IFALD. Understanding the pathogenic roles of cell death may help uncover the underlying mechanisms and develop novel therapeutic strategies in IFALD. In this review, we discuss the current knowledge on how RCD may link to the pathogenesis of IFALD. We highlight examples of cell death-targeted interventions aiming to attenuate the disease, and provide perspectives for future basic and translational research in the field.
Collapse
Affiliation(s)
- Si-Yang Cheng
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Lu Jiang
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Ying Wang
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Wei Cai
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China.
| |
Collapse
|
5
|
Cheng S, Wang Y, Zhao Y, Wang N, Yan J, Jiang L, Cai W. Targeting GPX4-mediated Ferroptosis Alleviates Liver Steatosis in a Rat Model of Total Parenteral Nutrition. J Pediatr Surg 2024; 59:981-991. [PMID: 37968154 DOI: 10.1016/j.jpedsurg.2023.10.048] [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: 07/17/2023] [Revised: 09/23/2023] [Accepted: 10/07/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Parenteral nutrition-associated liver disease (PNALD) is a common hepatobiliary complication resulting from long-term parenteral nutrition (PN) that is associated with significant morbidity and mortality. Ferroptosis plays a significant role in the pathogenesis of various liver diseases. This study aims to explore the role of ferroptosis in PNALD and to uncover its underlying mechanisms. METHODS Ferroptosis was evaluated in pediatric patients with PNALD and in rats administered with total parenteral nutrition (TPN) as an animal model of PNALD. In TPN-fed rats, we applied liproxstatin-1 (Lip-1) to inhibit ferroptosis for 7 days and assessed its impact on liver steatosis. We performed RNA-seq analysis to profile the alterations in miRNAs in livers from TPN-fed rats. The ferroptosis-promoting effects of miR-431 were evaluated in HepG2 cells and the direct targeting effects on glutathione peroxidase 4 (GPX4) were evaluated in HEK293T cells. RESULTS RNA-seq analysis and experimental validation suggested that ferroptosis was increased in the livers of pediatric patients and rats with PNALD. Inhibiting ferroptosis with Lip-1 attenuated liver steatosis by regulating PPARα expression. RNA-seq analysis uncovered miR-431 as the most upregulated miRNA in the livers of TPN-fed rats, showing a negative correlation with hepatic GPX4 expression. In vitro studies demonstrated that miR-431 promoted ferroptosis by directly binding to the 3'UTR of GPX4 mRNA, resulting in the suppression of its expression. CONCLUSIONS Our study demonstrates that TPN induces the upregulation of miR-431 in rats, leading to activation of ferroptosis through downregulation of GPX4. Inhibition of ferroptosis attenuates TPN-induced liver steatosis by regulating PPARα expression.
Collapse
Affiliation(s)
- Siyang Cheng
- Division of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Shanghai Institute for Pediatric Research, 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; Shanghai Institute for Pediatric Research, Shanghai, China
| | - Yuling Zhao
- Division of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China
| | - Nan Wang
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China
| | - Junkai Yan
- 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; Shanghai Institute for Pediatric Research, Shanghai, China
| | - Lu Jiang
- 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; Shanghai Institute for Pediatric Research, Shanghai, China.
| | - Wei Cai
- Division of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; 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; Shanghai Institute for Pediatric Research, Shanghai, China.
| |
Collapse
|
6
|
Takahashi T, Maejima T, Miyazaki D, Fukahori S, Hagiwara M. Teduglutide-induced acute gastric mucosal necrosis in short bowel syndrome with hepatorenal failure: Case report. Int J Surg Case Rep 2024; 117:109524. [PMID: 38493615 PMCID: PMC10958469 DOI: 10.1016/j.ijscr.2024.109524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
INTRODUCTION Short bowel syndrome (SBS) resulting from acute aortic dissection (AAD)-induced visceral malperfusions leads to chronic intestinal failure (CIF), necessitating patients to adopt home parenteral nutrition to prevent malabsorption. Teduglutide (TED), a glucagon-like peptide-2 analog, is a promising pharmacotherapy for intestinal rehabilitation that reduces parenteral support and improves the quality of life. Gastric mucosal necrosis, a rare gastrointestinal disorder, had never been observed as an adverse event relevant to this drug. We report a case of mucosal necrosis in the stomach after TED treatment for SBS-CIF with hepatorenal failure. PRESENTATION OF CASE A 68-year-old Japanese man who underwent massive intestinal resection for AAD experienced malnutrition and diarrhea caused by SBS-CIF. The patient received TED to improve intestinal absorption and entero-hepatic circulation besides controlling infectious diseases. Endoscopy showed mucosal hyperplasia in the stomach and duodenum 1.5 months after TED administration. The patient consented to enteral nutrition via a nasogastric tube because of anorexia. The nutritional status gradually improved after initiating enteral feeding. However, the patient experienced hematemesis 13 days after enteral feeding, and endoscopy revealed acute gastric mucosal necrosis, followed by fatal septic shock. DISCUSSION For patients with SBS, TED is expected to increase intestinal absorption through epithelial proliferation. When SBS is accompanied by multiple ischemic organ failure, TED therapeutic effects remain unclear as malnutrition-associated infectious diseases are refractory, and many underlying mechanisms can be involved. CONCLUSION TED administration should be deliberately considered for patients with SBS-CIF and multiple organ failure experiencing uncontrolled systemic infection.
Collapse
Affiliation(s)
- Tohru Takahashi
- Department of General Surgery, Sapporo Higashi Tokushukai Hospital, 3-1, North 33 East 14, East district, Sapporo, Hokkaido prefecture 065-0033, Japan.
| | - Taku Maejima
- Department of General Surgery, Sapporo Higashi Tokushukai Hospital, 3-1, North 33 East 14, East district, Sapporo, Hokkaido prefecture 065-0033, Japan
| | - Dai Miyazaki
- Department of General Surgery, Sapporo Higashi Tokushukai Hospital, 3-1, North 33 East 14, East district, Sapporo, Hokkaido prefecture 065-0033, Japan
| | - Susumu Fukahori
- Department of General Surgery, Sapporo Higashi Tokushukai Hospital, 3-1, North 33 East 14, East district, Sapporo, Hokkaido prefecture 065-0033, Japan
| | - Masahiro Hagiwara
- Department of General Surgery, Sapporo Higashi Tokushukai Hospital, 3-1, North 33 East 14, East district, Sapporo, Hokkaido prefecture 065-0033, Japan
| |
Collapse
|
7
|
Trotta RJ, Swanson KC, Klotz JL, Harmon DL. Postruminal Casein Infusion and Exogenous Glucagon-Like Peptide 2 Administration Differentially Stimulate Pancreatic α-Amylase and Small Intestinal α-Glucosidase Activity in Cattle. J Nutr 2023; 153:2854-2867. [PMID: 37573014 DOI: 10.1016/j.tjnut.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Increasing luminal carbohydrate flow decreases pancreatic α-amylase activity but can increase jejunal maltase activity, suggesting that regulation of carbohydrase activity is perhaps uncoordinated in response to luminal carbohydrate flow. Increasing luminal casein flow increases pancreatic α-amylase activity in cattle, and exogenous glucagon-like peptide 2 (GLP-2) has been shown to increase small intestinal α-glucosidase activity in nonruminants. OBJECTIVES The objective was to evaluate the effects of postruminal casein infusion, exogenous GLP-2, or their combination on endogenous pancreatic and small intestinal carbohydrase activity in cattle postruminally infused with starch. METHODS Holstein steers [n = 24; 250 ± 23 kg body weight (BW)] received a continuous abomasal infusion of 3.94 g raw corn starch/kg of BW combined with either 0 or 1.30 g casein/kg of BW. Steers received subcutaneous injections in 2 equal portions daily of excipient (0.5% bovine serum albumin) or 100 μg GLP-2/kg of BW per day. At the end of the 7-d treatment period, steers were slaughtered for tissue collection. Data were analyzed using the MIXED procedure of SAS version 9.4 (SAS Institute Inc.). RESULTS Postruminal casein infusion increased (P ≤ 0.03) pancreatic mass by 12.6%, total pancreatic α-amylase activity by 50%, and postruminal starch disappearance from 96.7% to 99.3%. Exogenous GLP-2 increased (P < 0.01) total small intestinal and mucosal mass by 1.2 kg and 896 g, respectively. Relative to control, GLP-2 and casein + GLP-2 increased (P = 0.04) total small intestinal α-glucosidase activity by 83.5%. Total small intestinal maltase, isomaltase, and glucoamylase activity was 90%, 100%, and 66.7% greater for GLP-2 and casein + GLP-2 steers compared with control. CONCLUSIONS Casein increased pancreatic α-amylase activity, GLP-2 increased small intestinal α-glucosidase activity, and the combination of casein and GLP-2 increased both pancreatic α-amylase activity and small intestinal α-glucosidase activity. This novel approach provides an in vivo model to evaluate effects of increasing endogenous carbohydrase activity on small intestinal starch digestion.
Collapse
Affiliation(s)
- Ronald J Trotta
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY, United States
| | - Kendall C Swanson
- Department of Animal Science, North Dakota State University, Fargo, ND, United States
| | - James L Klotz
- Forage-Animal Production Research Unit, USDA, ARS, Lexington, KY, United States
| | - David L Harmon
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
8
|
Xu J, Zhou Y, Cheng S, Zhao Y, Yan J, Wang Y, Cai W, Jiang L. Lactobacillus johnsonii Attenuates Liver Steatosis and Bile Acid Dysregulation in Parenteral Nutrition-Fed Rats. Metabolites 2023; 13:1043. [PMID: 37887368 PMCID: PMC10608838 DOI: 10.3390/metabo13101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Parenteral nutrition (PN), a vital therapy for patients with intestinal failure, can lead to the development of parenteral nutrition-associated liver disease (PNALD). In this study, we aimed to investigate the role of Lactobacillus johnsonii (L. johnsonii) in a rat model of PNALD. Total parenteral nutrition (TPN)-fed rats were used to assess the role of L. johnsonii in liver steatosis, bile acid metabolism, gut microbiota, and hepatocyte apoptosis. We observed a depletion of L. johnsonii that was negatively correlated with the accumulation of glycochenodeoxycholic acid (GCDCA), a known apoptosis inducer, in rats subjected to TPN. L. johnsonii attenuated TPN-induced liver steatosis by inhibiting fatty acid synthesis and promoting fatty acid oxidation. TPN resulted in a decrease in bile acid synthesis and biliary bile secretion, which were partially restored by L. johnsonii treatment. The gut microbial profile revealed depletion of pathogenic bacteria in L. johnsonii-treated rats. L. johnsonii treatment reduced both hepatic GCDCA levels and hepatocyte apoptosis compared with the TPN group. In vitro, L. johnsonii treatment inhibited GCDCA-induced hepatocyte apoptosis via its bile salt hydrolase (BSH) activity. Our findings suggest that L. johnsonii protects against liver steatosis, bile acid dysregulation, and hepatocyte apoptosis in TPN-fed rats.
Collapse
Affiliation(s)
- Juan Xu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
| | - Yongchang Zhou
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
| | - Siyang Cheng
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (S.C.); (Y.Z.)
| | - Yuling Zhao
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (S.C.); (Y.Z.)
| | - Junkai Yan
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Wei Cai
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (S.C.); (Y.Z.)
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Lu Jiang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; (J.X.); (J.Y.); (Y.W.)
- Shanghai Institute for Pediatric Research, Shanghai 200092, China;
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| |
Collapse
|
9
|
Stroe MS, Van Bockstal L, Valenzuela A, Ayuso M, Leys K, Annaert P, Carpentier S, Smits A, Allegaert K, Zeltner A, Mulder A, Van Ginneken C, Van Cruchten S. Development of a neonatal Göttingen Minipig model for dose precision in perinatal asphyxia: technical opportunities, challenges, and potential further steps. Front Pediatr 2023; 11:1163100. [PMID: 37215599 PMCID: PMC10195037 DOI: 10.3389/fped.2023.1163100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Animal models provide useful information on mechanisms in human disease conditions, but also on exploring (patho)physiological factors affecting pharmacokinetics, safety, and efficacy of drugs in development. Also, in pediatric patients, nonclinical data can be critical for better understanding the disease conditions and developing new drug therapies in this age category. For perinatal asphyxia (PA), a condition defined by oxygen deprivation in the perinatal period and possibly resulting in hypoxic ischemic encephalopathy (HIE) or even death, therapeutic hypothermia (TH) together with symptomatic drug therapy, is the standard approach to reduce death and permanent brain damage in these patients. The impact of the systemic hypoxia during PA and/or TH on drug disposition is largely unknown and an animal model can provide useful information on these covariates that cannot be assessed separately in patients. The conventional pig is proven to be a good translational model for PA, but pharmaceutical companies do not use it to develop new drug therapies. As the Göttingen Minipig is the commonly used pig strain in nonclinical drug development, the aim of this project was to develop this animal model for dose precision in PA. This experiment consisted of the instrumentation of 24 healthy male Göttingen Minipigs, within 24 h of partus, weighing approximately 600 g, to allow the mechanical ventilation and the multiple vascular catheters inserted for maintenance infusion, drug administration and blood sampling. After premedication and induction of anesthesia, an experimental protocol of hypoxia was performed, by decreasing the inspiratory oxygen fraction (FiO2) at 15%, using nitrogen gas. Blood gas analysis was used as an essential tool to evaluate oxygenation and to determine the duration of the systemic hypoxic insult to approximately 1 h. The human clinical situation was mimicked for the first 24 h after birth in case of PA, by administering four compounds (midazolam, phenobarbital, topiramate and fentanyl), frequently used in a neonatal intensive care unit (NICU). This project aimed to develop the first neonatal Göttingen Minipig model for dose precision in PA, allowing to separately study the effect of systemic hypoxia versus TH on drug disposition. Furthermore, this study showed that several techniques that were thought to be challenging or even impossible in these very small animals, such as endotracheal intubation and catheterization of several veins, are feasible by trained personnel. This is relevant information for laboratories using the neonatal Göttingen Minipig for other disease conditions or drug safety testing.
Collapse
Affiliation(s)
| | | | - Allan Valenzuela
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium
| | - Miriam Ayuso
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium
| | - Karen Leys
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
- BioNotus GCV, Niel, Belgium
| | | | - Anne Smits
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, Netherlands
| | | | - Antonius Mulder
- Neonatal Intensive Care Unit, Antwerp University Hospital, Antwerp, Belgium
| | - Chris Van Ginneken
- Comparative Perinatal Development, University of Antwerp, Antwerp, Belgium
| | | |
Collapse
|
10
|
You T, Tang J, Yin S, Jia G, Liu G, Tian G, Chen X, Cai J, Kang B, Zhao H. Effect of dietary licorice flavonoids powder on performance, intestinal immunity and health of weaned piglets. J Anim Physiol Anim Nutr (Berl) 2023; 107:147-156. [PMID: 35247278 DOI: 10.1111/jpn.13694] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/21/2021] [Accepted: 01/31/2022] [Indexed: 01/10/2023]
Abstract
Licorice flavonoids, a bioactive substance derived from glycyrrhiza, have been reported for many pharmacological properties and are beneficial to animal health. This study aimed to explore the effects of licorice flavonoids powder (LFP) on growth performance and intestinal health of piglets. A total of 96 weaned piglets were randomly assigned into four treatments and supplemented with 0, 50, 150 and 250 mg/kg LFP for 5 weeks. Dietary LFP supplementation tended to increase (p = 0.068) average daily gain (ADG) and reduce (p = 0.089) the feed intake/body gain (F/G) of piglets than that of the control group during 15-35 days; and concentrations of LFP supplementation reduced (p < 0.01) diarrhoea index during 14-35 days and 0-35 days. Piglets fed on diets supplied with LFP had a lower (p < 0.05) pH in caecum and colon. Dietary LFP supplementation increased (p < 0.01) the villi height and the ratio of villi height/crypt depth in duodenum, and reduced (p < 0.05) crypt depth in duodenum. Compared with the control group, 250 mg/kg LFP supplementation up-regulated (p < 0.05) the mRNA level of occludin (OCLN) in ileum. Meanwhile, dietary LFP supplementation down-regulated (p < 0.05) mRNA abundance of Interleukin (IL)-1β, IL-8 and induced nitrogen monoxide synthase (INOS) in duodenum. Dietary 150 mg/kg LFP supplementation down-regulated (p < 0.05) mRNA abundance of IL-1β and 250 mg/kg LFP up-regulated (p < 0.05) the expression of IL-10 in ileum. In summary, dietary LFP supplementation has a trend to improve the performance of weaning piglets, those improvements are accompanied by reduction in diarrhoea, enhancement of intestinal morphological structure, barrier function, immune function, and development. In general, 150 mg/kg LFP supplementation is more effective.
Collapse
Affiliation(s)
- Ting You
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Jiayong Tang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Shenggang Yin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Gang Jia
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Guangmang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Gang Tian
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Xiaoling Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Jingyi Cai
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| | - Bo Kang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hua Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, China
| |
Collapse
|
11
|
Kounatidis D, Vallianou NG, Tsilingiris D, Christodoulatos GS, Geladari E, Stratigou T, Karampela I, Dalamaga M. Therapeutic Potential of GLP-2 Analogs in Gastrointestinal Disorders: Current Knowledge, Nutritional Aspects, and Future Perspectives. Curr Nutr Rep 2022; 11:618-642. [PMID: 35933503 DOI: 10.1007/s13668-022-00433-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Although Glucagon-like peptide (GLP)-1 receptor agonists have been used for almost two decades in the treatment of diabetes mellitus type 2 and, lately, in obesity, recent years have seen an increasing interest in the pharmacological agonism of other proglucagon-derived peptides, including GLP-2. Herein, we aimed to review the available evidence on the effects of GLP-2 agonism from animal and clinical studies. Furthermore, we summarize the current clinical applications of GLP-2 agonists among patients with intestinal failure associated with short bowel syndrome (SBS-IF) as well as potential future expansion of their indications to other intestinal disorders. RECENT FINDINGS Evidence from preclinical studies has highlighted the cellular trophic and functional beneficial actions of GLP-2 on small intestinal and colonic mucosa. Subsequently, pharmacologic agonism of GLP-2 has gathered interest for the treatment of patients with conditions pertaining to the loss of intestinal anatomical and/or functional integrity to a degree requiring parenteral support, collectively referred to as intestinal failure. GLP-2 analogs positively influence nutrient absorption in animal models and humans, although continued therapy is likely needed for sustained effects. The degradation-resistant GLP-2-analog teduglutide has received approval for the treatment of SBS-IF, in which it may decisively reduce patient dependency on parenteral support and improve quality of life. Another two longer-acting analogs, glepaglutide and apraglutide, are currently undergoing phase III clinical trials. The use of GLP-2 analogs is effective in the management of SBS-IF and may show promise in the treatment of other severe gastrointestinal disorders associated with loss of effective intestinal resorptive surface area.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Departments of Internal Medicine and Endocrinology, Evangelismos General Hospital, 45-47 Ypsilantou Street, 10676, Athens, Greece
| | - Natalia G Vallianou
- Departments of Internal Medicine and Endocrinology, Evangelismos General Hospital, 45-47 Ypsilantou Street, 10676, Athens, Greece.
| | - Dimitrios Tsilingiris
- First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 17 St Thomas Street, 11527, Athens, Greece
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, Goudi, 11527, Athens, Greece
| | - Eleni Geladari
- Departments of Internal Medicine and Endocrinology, Evangelismos General Hospital, 45-47 Ypsilantou Street, 10676, Athens, Greece
| | - Theodora Stratigou
- Departments of Internal Medicine and Endocrinology, Evangelismos General Hospital, 45-47 Ypsilantou Street, 10676, Athens, Greece
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Street, Chaidari, 12462, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, Goudi, 11527, Athens, Greece.
| |
Collapse
|
12
|
Zhao X, Hui Q, Azevedo P, Nyachoti CM, O K, Yang C. Calcium-sensing receptor is not expressed in the absorptive enterocytes of weaned piglets. J Anim Sci 2022; 100:6549683. [PMID: 35294536 PMCID: PMC9030235 DOI: 10.1093/jas/skac085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/12/2022] [Indexed: 11/14/2022] Open
Abstract
The calcium-sensing receptor (CaSR) is a kokumi receptor that plays an essential role in nutrient sensing and animal physiology, growth, and development. Pig CaSR (pCaSR) was identified and characterized in the intestine. However, further research is still needed to confirm the expression of CaSR in the epithelial cells isolated from weaned piglets. In this study, primary enterocytes were isolated and characterized from the ileum of weaned piglets by the Weiser distended intestinal sac technique and fluorescence-activated cell sorting (FACS) based on sucrase-isomaltase (SI) as an enterocyte-specific marker. The expression of CaSR was investigated in both primary enterocytes and the intestinal porcine enterocyte cell line-j2 (IPEC-J2) by droplet digital polymerase chain reaction (ddPCR), immunofluorescence staining, and Western blotting. Results demonstrated that porcine enterocytes could be obtained using FACS with the SI as the enterocyte-specific marker and that pCaSR is not expressed in both porcine ileal enterocytes and IPEC-J2 cells, which specifically identified the expression of pCaSR in ileal enterocytes with sensitive and specific approaches.
Collapse
Affiliation(s)
- Xiaoya Zhao
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Qianru Hui
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Paula Azevedo
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | | | - Karmin O
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,CCARM, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Chengbo Yang
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
13
|
Zhou H, Sun J, Yu B, Liu Z, Chen H, He J, Mao X, Zheng P, Yu J, Luo J, Luo Y, Yan H, Ge L, Chen D. Gut microbiota absence and transplantation affect growth and intestinal functions: An investigation in a germ-free pig model. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:295-304. [PMID: 34258417 PMCID: PMC8245803 DOI: 10.1016/j.aninu.2020.11.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022]
Abstract
This study was conducted to investigate host–microbiota interactions and explore the effects of maternal gut microbiota transplantation on the growth and intestinal functions of newborns in a germ-free (GF) pig model. Twelve hysterectomy-derived GF Bama piglets were reared in 6 sterile isolators. Among them, 6 were considered as the GF group, and the other 6 were orally inoculated with healthy sow fecal suspension as fecal microbiota transplanted (FMT) group. Another 6 piglets from natural birth were regarded as the conventional (CV) group. The GF and FMT groups were hand-fed with Co60-γ-irradiated sterile milk powder, while the CV group was reared by lactating Bama sows. All groups were fed for 21 days. Then, all piglets and then were switched to sterile feed for another 21 days. Results showed that the growth performance, nutrient digestibility, and concentrations of short-chain fatty acids in the GF group decreased (P < 0.05). Meanwhile, the serum urea nitrogen concentration and digesta pH values in the GF group increased compared with those in the FMT and CV groups (P < 0.05). Compared with the CV group, the GF group demonstrated upregulation in the mRNA expression levels of intestinal barrier function-related genes in the small intestine (P < 0.05). In addition, the mRNA abundances of intestinal development and absorption-related genes in the small intestine and colon were higher in the GF group than in the CV and FMT groups (P < 0.05). The FMT group exhibited greater growth performance, lipase activity, and nutrient digestibility (P < 0.05), higher mRNA expression levels of intestinal development and barrier-related genes in the small intestine (P < 0.05), and lower mRNA abundances of pro-inflammatory factor in the colon and jejunum (P < 0.05) than the CV group. In conclusion, the absence of gut microbes impaired the growth and nutrient digestibility, and healthy sow gut microbiota transplantation increased the growth and nutrient digestibility and improved the intestinal development and barrier function of newborn piglets, indicating the importance of intestinal microbes for intestinal development and functions.
Collapse
Affiliation(s)
- Hua Zhou
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jing Sun
- Key Laboratory of Pig Industry Sciences, Rongchang, Chongqing 402460, China.,Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China
| | - Bing Yu
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zuohua Liu
- Key Laboratory of Pig Industry Sciences, Rongchang, Chongqing 402460, China.,Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China
| | - Hong Chen
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China
| | - Jun He
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiangbing Mao
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Ping Zheng
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jie Yu
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Junqiu Luo
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yuheng Luo
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Hui Yan
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Liangpeng Ge
- Key Laboratory of Pig Industry Sciences, Rongchang, Chongqing 402460, China.,Chongqing Academy of Animal Sciences, Rongchang, Chongqing 402460, China
| | - Daiwen Chen
- Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan 611130, China.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
14
|
Billeschou A, Hunt JE, Ghimire A, Holst JJ, Kissow H. Intestinal Adaptation upon Chemotherapy-Induced Intestinal Injury in Mice Depends on GLP-2 Receptor Activation. Biomedicines 2021; 9:biomedicines9010046. [PMID: 33430185 PMCID: PMC7825593 DOI: 10.3390/biomedicines9010046] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/16/2020] [Accepted: 01/04/2021] [Indexed: 01/15/2023] Open
Abstract
Intestinal adaptation is an important response and a natural repair mechanism in acute intestinal injury and is critical for recovery. Glucagon-like peptide 2 (GLP-2) has been demonstrated to enhance mucosal repair following intestinal damage. In this study, we aimed to investigate the role of GLP-2 receptor activation on intestinal protection and adaptation upon chemotherapy-induced intestinal injury. The injury was induced with a single injection of 5-fluorouracil in female GLP-2 receptor knockout (GLP-2R(-/-)) mice and their wild type (WT) littermates. The mice were euthanized in the acute or the recovery phase of the injury; the small intestines were analysed for weight changes, morphology, histology, inflammation, apoptosis and proliferation. In the acute phase, only inflammation was slightly increased in the GLP-2R(-/-) mice compared to WT. In the recovery phase, we observed the natural compensatory response with an increase in small intestinal weight, crypt depth and villus height in WT mice, and this was absent in the GLP-2R(-/-) mice. Both genotypes responded with hyperproliferation. From this, we concluded that GLP-2R signalling does not have a major impact on acute intestinal injury but is pivotal for the adaptive response in the small intestine.
Collapse
Affiliation(s)
- Anna Billeschou
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; (A.B.); (J.E.H.); (A.G.); (J.J.H.)
| | - Jenna Elizabeth Hunt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; (A.B.); (J.E.H.); (A.G.); (J.J.H.)
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Aruna Ghimire
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; (A.B.); (J.E.H.); (A.G.); (J.J.H.)
| | - Jens J. Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; (A.B.); (J.E.H.); (A.G.); (J.J.H.)
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; (A.B.); (J.E.H.); (A.G.); (J.J.H.)
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
15
|
Qi KK, Wu J, Wen Jun Z, Bo D, Xu ZW. Catch-up growth in intrauterine growth-restricted piglets associated with the restore of pancreatic and intestinal functions via porcine glucagon-like peptide-2 microspheres. Arch Anim Nutr 2020; 74:462-475. [PMID: 33076701 DOI: 10.1080/1745039x.2020.1833598] [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] [Indexed: 02/04/2023]
Abstract
Intrauterine growth restriction (IUGR) results in abnormal morphology and gastrointestinal function, such as reduced villi height and crypt depth, thinner mucosa and muscle layers, and reduced brush border enzyme activities, delayed gastric emptying, increased stress response. As a gastrointestinal growth factor, the manner by which the porcine glucagon-like peptide-2 (pGLP-2) microsphere administration restored the gastrointestinal function and growth performance of IUGR piglets was investigated. Fourteen newborn Duroc × (Yorkshire × Landrace) IUGR piglets (0.92 ± 0.113 kg) were assigned into the IUGR (negative control group) and pGLP-2 microsphere groups. The piglets in group pGLP-2 were intraperitoneally administered with 100 mg pGLP-2 microspheres on day 1 after birth. From days 15 to 26 of trial, the body weight of the pGLP-2 group was significantly higher than that of the control. IUGR piglets of group pGLP-2 showed a significantly increased pancreas weight, serum insulin content and activity of lipase and amylase. Injection of pGLP-2 microspheres restored the intestinal absorptive capacity by significantly increasing the mRNA expression of the sodium-glucose cotransporter 1 in the jejunum and the peptide transporter 1 in the jejunum. It also restored the redox balance by increasing the catalase mRNA expression and decreasing the heat shock protein 70 mRNA expression. In addition, this improvement was associated with the significant increase in gut diameter, length and weight. Therefore, it was concluded that the injection of pGLP-2 microspheres was a suitable therapeutic strategy for compensatory growth in low birth weight IUGR piglets.
Collapse
Affiliation(s)
- Ke Ke Qi
- Institute of Animal Science, Zhejiang Academy of Agricultural Sciences , Hangzhou, China
| | - Jie Wu
- Institute of Animal Science, Zhejiang Academy of Agricultural Sciences , Hangzhou, China
| | - Zhou Wen Jun
- Institute of Animal Science, Zhejiang Academy of Agricultural Sciences , Hangzhou, China
| | - Deng Bo
- Institute of Animal Science, Zhejiang Academy of Agricultural Sciences , Hangzhou, China
| | - Zi Wei Xu
- Institute of Animal Science, Zhejiang Academy of Agricultural Sciences , Hangzhou, China
| |
Collapse
|
16
|
Burrin D, Sangild PT, Stoll B, Thymann T, Buddington R, Marini J, Olutoye O, Shulman RJ. Translational Advances in Pediatric Nutrition and Gastroenterology: New Insights from Pig Models. Annu Rev Anim Biosci 2020; 8:321-354. [PMID: 32069436 DOI: 10.1146/annurev-animal-020518-115142] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pigs are increasingly important animals for modeling human pediatric nutrition and gastroenterology and complementing mechanistic studies in rodents. The comparative advantages in size and physiology of the neonatal pig have led to new translational and clinically relevant models of important diseases of the gastrointestinal tract and liver in premature infants. Studies in pigs have established the essential roles of prematurity, microbial colonization, and enteral nutrition in the pathogenesis of necrotizing enterocolitis. Studies in neonatal pigs have demonstrated the intestinal trophic effects of akey gut hormone, glucagon-like peptide 2 (GLP-2), and its role in the intestinal adaptation process and efficacy in the treatment of short bowel syndrome. Further, pigs have been instrumental in elucidating the physiology of parenteral nutrition-associated liver disease and the means by which phytosterols, fibroblast growth factor 19, and a new generation of lipid emulsions may modify disease. The premature pig will continue to be a valuable model in the development of optimal infant diets (donor human milk, colostrum), specific milk bioactives (arginine, growth factors), gut microbiota modifiers (pre-, pro-, and antibiotics), pharmaceutical drugs (GLP-2 analogs, FXR agonists), and novel diagnostic tools (near-infrared spectroscopy) to prevent and treat these pediatric diseases.
Collapse
Affiliation(s)
- Douglas Burrin
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Barbara Stoll
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Randal Buddington
- College of Nursing, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Juan Marini
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA; .,Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Robert J Shulman
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
17
|
Hunt JE, Billeschou A, Windeløv JA, Hartmann B, Ullmer C, Holst JJ, Kissow H. Pharmacological activation of TGR5 promotes intestinal growth via a GLP-2-dependent pathway in mice. Am J Physiol Gastrointest Liver Physiol 2020; 318:G980-G987. [PMID: 32308039 DOI: 10.1152/ajpgi.00062.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glucagon-like peptide (GLP)-1 and -2-secreting L cells have been shown to express the bile acid receptor Takeda G protein-receptor-5 (TGR5) and increase secretion upon receptor activation. Previous studies have explored GLP-1 secretion following acute TGR5 activation, but chronic activation and GLP-2 responses have not been characterized. In this study, we aimed to investigate the consequences of pharmacological TGR5 receptor activation on L cell hormone production in vivo using the specific TGR5 agonist RO5527239 and the GLP-2 receptor knockout mouse. Here, we show that 1) TGR5 receptor activation led to increased GLP-1 and GLP-2 content in the colon, which 2) was associated with an increased small intestinal weight that 3) was GLP-2 dependent. Additionally, we report that TGR5-mediated gallbladder filling occurred independently of GLP-2 signaling. In conclusion, we demonstrate that pharmacological TGR5 receptor activation stimulates L cells, triggering GLP-2-dependent intestinal adaption in mice.NEW & NOTEWORTHY Using the specific Takeda G protein-receptor-5 (TGR5) agonist RO5527239 and GLP-2 receptor knockout mice, we show that activation of TGR5 led to the increase in colonic GLP-1 and GLP-2 concomitant with a GLP-2 dependent growth response in the proximal portion of the small intestine.
Collapse
Affiliation(s)
- Jenna Elizabeth Hunt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordic Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Billeschou
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordic Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johanne Agerlin Windeløv
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordic Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordic Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoph Ullmer
- Roche Pharmaceutical Research and Early Development, F. Hoffmann-La Roche Limited, Basel, Switzerland
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordic Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordic Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
A Transcriptome Analysis Identifies Biological Pathways and Candidate Genes for Feed Efficiency in DLY Pigs. Genes (Basel) 2019; 10:genes10090725. [PMID: 31540540 PMCID: PMC6771153 DOI: 10.3390/genes10090725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/08/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022] Open
Abstract
Feed cost accounts for approximately 65–75% of overall commercial pork production costs. Therefore, improving the feed efficiency of pig production is important. In this study, 12 individuals with either extremely high (HE) or low (LE) feed efficiency were selected from 225 Duroc × (Landrace × Yorkshire) (DLY) pigs. After the pigs were slaughtered, we collected small intestine mucosal tissue. Next, RNA sequencing (RNA-seq) analysis was used to reveal the presence and quantity of genes expressed between these extremely HE- and LE-groups. We found 433 significantly differentially expressed genes (DEGs) between the HE- and LE-groups. Of these, 389 and 44 DEGs were upregulated and downregulated in the HE-group, respectively. An enrichment analysis showed that the DEGs were mainly enriched in functions related to apical plasma membrane composition, transporter activity, transport process and hormone regulation of digestion and absorption. Protein network interaction and gene function analyses revealed that SLC2A2 was an important candidate gene for FE in pigs, which may give us a deeper understanding of the mechanism of feed efficiency. Furthermore, some significant DEGs identified in the current study could be incorporated into artificial selection programs for increased feeding efficiency in pigs.
Collapse
|
19
|
Lin S, Stoll B, Robinson J, Pastor JJ, Marini JC, Ipharraguerre IR, Hartmann B, Holst JJ, Cruz S, Lau P, Olutoye O, Fang Z, Burrin DG. Differential action of TGR5 agonists on GLP-2 secretion and promotion of intestinal adaptation in a piglet short bowel model. Am J Physiol Gastrointest Liver Physiol 2019; 316:G641-G652. [PMID: 30920308 PMCID: PMC6580240 DOI: 10.1152/ajpgi.00360.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/14/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
Abstract
Enteroendocrine L cells and glucagon-like peptide 2 (GLP-2) secretion are activated in the intestinal adaptation process following bowel resection in patients with short bowel syndrome. We hypothesized that enteral activation of Takeda G protein-coupled receptor 5 (TGR5), expressed in enteroendocrine L cells, could augment endogenous GLP-2 secretion and the intestinal adaptation response. Our aim was to assess the efficacy of different TGR5 agonists to stimulate GLP-2 secretion and intestinal adaptation in a piglet short-bowel model. In study 1, parenterally fed neonatal pigs (n = 6/group) were gavaged with vehicle, olive extract (OE; 10 or 50 mg/kg), or ursolic acid (UA; 10 mg/kg), and plasma GLP-2 was measured for 6 h. In study 2, neonatal pigs (n = 6-8/group) were subjected to transection or 80% mid-small intestine resection and, after 2 days, assigned to treatments for 10 days as follows: 1) transection + vehicle (sham), 2) resection + vehicle (SBS), 3) resection + 30 mg UA (SBS + UA), and 4) resection + 180 mg/kg OE (SBS + OE). We measured plasma GLP-2, intestinal histology, cell proliferation, and gene expression, as well as whole body citrulline-arginine kinetics and bile acid profiles. In study 1, GLP-2 secretion was increased by UA and tended to be increased by OE. In study 2, SBS alone, but not additional treatment with either TGR5 agonist, resulted in increased mucosal thickness and crypt cell proliferation in remnant jejunum and ileum sections. SBS increased biliary and ileal concentration of bile acids and expression of inflammatory and farnesoid X receptor target genes, but these measures were suppressed by UA treatment. In conclusion, UA is an effective oral GLP-2 secretagogue in parenterally fed pigs but is not capable of augmenting GLP-2 secretion or the intestinal adaptation response after massive small bowel resection. NEW & NOTEWORTHY Therapeutic activation of endogenous glucagon-like peptide 2 (GLP-2) secretion is a promising strategy to improve intestinal adaptation in patients with short bowel syndrome. This study in neonatal pigs showed that oral supplementation with a selective Takeda G protein-coupled receptor 5 (TGR5) agonist is an effective approach to increase GLP-2 secretion. The results warrant further study to establish a more potent oral TGR5 agonist that can effectively improve intestinal adaptation in pediatric patients with SBS.
Collapse
Affiliation(s)
- Sen Lin
- Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu, Sichuan , People's Republic of China
| | - Barbara Stoll
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
| | - Jason Robinson
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
| | | | - Juan C Marini
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Ignacio R Ipharraguerre
- Lucta S.A., Montornès del Vallès, Spain
- Institute of Human Nutrition and Food Science, University of Kiel , Kiel , Germany
| | - Bolette Hartmann
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Jens J Holst
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Stephanie Cruz
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Texas Children's Hospital , Houston, Texas
| | - Patricio Lau
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Texas Children's Hospital , Houston, Texas
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Texas Children's Hospital , Houston, Texas
| | - Zhengfeng Fang
- Institute of Animal Nutrition, Sichuan Agricultural University , Chengdu, Sichuan , People's Republic of China
| | - Douglas G Burrin
- US Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center , Houston, Texas
- Section of Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
20
|
Nerup N, Ambrus R, Lindhe J, Achiam MP, Jeppesen PB, Svendsen LB. The effect of glucagon‐like peptide‐1 and glucagon‐like peptide‐2 on microcirculation: A systematic review. Microcirculation 2019; 26:e12367. [DOI: 10.1111/micc.12367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/28/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Nikolaj Nerup
- Department of Surgical GastroenterologyRigshospitaletCopenhagen University Hospital Copenhagen Ø Denmark
| | - Rikard Ambrus
- Department of Surgical GastroenterologyRigshospitaletCopenhagen University Hospital Copenhagen Ø Denmark
| | - Joanna Lindhe
- Department of Surgical GastroenterologyRigshospitaletCopenhagen University Hospital Copenhagen Ø Denmark
| | - Michael P. Achiam
- Department of Surgical GastroenterologyRigshospitaletCopenhagen University Hospital Copenhagen Ø Denmark
| | - Palle B. Jeppesen
- Department of Medical GastroenterologyRigshospitaletCopenhagen University Hospital Copenhagen Ø Denmark
| | - Lars B. Svendsen
- Department of Surgical GastroenterologyRigshospitaletCopenhagen University Hospital Copenhagen Ø Denmark
| |
Collapse
|
21
|
Ebbesen MS, Kissow H, Hartmann B, Grell K, Gørløv JS, Kielsen K, Holst JJ, Müller K. Glucagon-Like Peptide-1 Is a Marker of Systemic Inflammation in Patients Treated with High-Dose Chemotherapy and Autologous Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1085-1091. [PMID: 30731250 DOI: 10.1016/j.bbmt.2019.01.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022]
Abstract
Autologous stem cell transplantation (ASCT) is challenged by side effects that may be propagated by chemotherapy-induced mucositis, resulting in bacterial translocation and systemic inflammation. Because gastrointestinal damage appears as an early event in this cascade of reactions, we hypothesized that markers reflecting damage to the intestinal barrier could serve as early predictive markers of toxicity. Glucagon-like peptide-1 (GLP-1), a well-known regulator of blood glucose, has been found to promote intestinal growth and repair in animal studies. We investigated fasting GLP-1 plasma levels in 66 adults undergoing ASCT for lymphoma and multiple myeloma. GLP-1 increased significantly after chemotherapy, reaching peak levels at day +7 post-transplant (median, 8 pmol/L [interquartile range, 4 to 12] before conditioning versus 10 pmol/L [interquartile range, 6 to 17] at day +7; P = .007). The magnitude of the GLP-1 increase was related to the intensity of conditioning. GLP-1 at the day of transplantation (day 0) was positively associated with peak C-reactive protein (CRP) levels (46 mg/L per GLP-1 doubling, P < .001) and increase in days with fever (32% per GLP-1 doubling, P = .0058). Patients with GLP-1 above the median at day 0 had higher CRP levels from days +3 to +10 post-transplant than patients with lower GLP-1 (P ≤ .041) with peak values of 238 versus 129 mg/L, respectively. This study, which represents the first clinical investigation of fasting GLP-1 in relation to high-dose chemotherapy, provides evidence that GLP-1 plays a role in regulation of mucosal defenses. Fasting GLP-1 levels may serve as an early predictor of systemic inflammation and fever in patients receiving high-dose chemotherapy.
Collapse
Affiliation(s)
- Maria Schou Ebbesen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark.
| | - Hannelouise Kissow
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kathrine Grell
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Section of Biostatistics, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Katrine Kielsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Institute for Inflammation Research, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Institute for Inflammation Research, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
22
|
Early Serum Gut Hormone Concentrations Associated With Time to Full Enteral Feedings in Preterm Infants. J Pediatr Gastroenterol Nutr 2018; 67:97-102. [PMID: 29620597 DOI: 10.1097/mpg.0000000000001987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES The primary objective of this study was to evaluate early postnatal serum gut hormone concentrations in preterm infants as predictors of time to full enteral feedings. The secondary objective was to identify infant characteristics and nutritional factors that modulate serum gut hormone concentrations and time to full enteral feedings. METHODS Sixty-four preterm infants less than 30 weeks of gestation were included in this retrospective cohort study. Serum gut hormone concentrations at postnatal days 0 and 7 were measured using enzyme-linked immunosorbent assays. Linear regression and mediation analyses were performed. RESULTS Median (interquartile range) serum concentrations of glucose-dependent insulinotropic peptide (GIP) and peptide YY (PYY) on postnatal day 7 were 31.3 pg/mL (18.2, 52.3) and 1181.7 pg/mL (859.0, 1650.2), respectively. GIP and PYY concentrations on day 7 were associated with days to full enteral feedings after adjustment for confounders (β = -1.1, P = 0.03; and β = -0.002, P = 0.02, respectively). Nutritional intake was correlated with serum concentrations of GIP and PYY on postnatal day 7 and time to full enteral feedings. Mediation analysis revealed that the effect of serum gut hormone concentrations on time to full enteral feedings was not fully explained by nutritional intake. Intrauterine growth restriction, mechanical ventilation on postnatal day 7, and patent ductus arteriosus treated with indomethacin were associated with longer time to full enteral feedings. CONCLUSIONS Serum concentrations of GIP and PYY on postnatal 7 are independently associated with time to full enteral feedings. The link between serum gut hormone concentrations and time to full enteral feedings is not fully mediated by nutritional factors, suggesting an independent mechanism underlying the influence of gut hormones on feeding tolerance and time to full enteral feedings.
Collapse
|
23
|
Brubaker PL. Glucagon‐like Peptide‐2 and the Regulation of Intestinal Growth and Function. Compr Physiol 2018; 8:1185-1210. [DOI: 10.1002/cphy.c170055] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Hytting-Andreasen R, Balk-Møller E, Hartmann B, Pedersen J, Windeløv JA, Holst JJ, Kissow H. Endogenous glucagon-like peptide- 1 and 2 are essential for regeneration after acute intestinal injury in mice. PLoS One 2018; 13:e0198046. [PMID: 29864142 PMCID: PMC5986149 DOI: 10.1371/journal.pone.0198046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/12/2018] [Indexed: 12/15/2022] Open
Abstract
Objective Mucositis is a side effect of chemotherapy seen in the digestive tract, with symptoms including pain, diarrhoea, inflammation and ulcerations. Our aim was to investigate whether endogenous glucagon-like peptide -1 and -2 (GLP-1 and GLP-2) are implicated in intestinal healing after chemotherapy-induced mucositis. Design We used a transgenic mouse model Tg(GCG.DTR)(Tg) expressing the human diphtheria toxin receptor in the proglucagon-producing cells. Injections with diphtheria toxin ablated the GLP-1 and GLP-2 producing L-cells in Tg mice with no effect in wild-type (WT) mice. Mice were injected with 5-fluorouracil or saline and received vehicle, exendin-4, teduglutide (gly2-GLP-2), or exendin-4/teduglutide in combination. The endpoints were body weight change, small intestinal weight, morphology, histological scoring of mucositis and myeloperoxidase levels. Results Ablation of L-cells led to impaired GLP-2 secretion; increased loss of body weight; lower small intestinal weight; lower crypt depth, villus height and mucosal area; and increased the mucositis severity score in mice given 5-fluorouracil. WT mice showed compensatory hyperproliferation as a sign of regeneration in the recovery phase. Co-treatment with exendin-4 and teduglutide rescued the body weight of the Tg mice and led to a hyperproliferation in the small intestine, whereas single treatment was less effective. Conclusion The ablation of L-cells leads to severe mucositis and insufficient intestinal healing, shown by severe body weight loss and lack of compensatory hyperproliferation in the recovery phase. Co-treatment with exendin-4 and teduglutide could prevent this. Because both peptides were needed, we can conclude that both GLP-1 and GLP-2 are essential for intestinal healing in mice.
Collapse
Affiliation(s)
- Rasmus Hytting-Andreasen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- NNF Center of Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Balk-Møller
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- NNF Center of Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- NNF Center of Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- NNF Center of Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Johanne Agerlin Windeløv
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- NNF Center of Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- NNF Center of Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- NNF Center of Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
25
|
Villalona G, Price A, Blomenkamp K, Manithody C, Saxena S, Ratchford T, Westrich M, Kakarla V, Pochampally S, Phillips W, Heafner N, Korremla N, Greenspon J, Guzman MA, Kumar Jain A. No Gut No Gain! Enteral Bile Acid Treatment Preserves Gut Growth but Not Parenteral Nutrition-Associated Liver Injury in a Novel Extensive Short Bowel Animal Model. JPEN J Parenter Enteral Nutr 2018; 42:1238-1251. [PMID: 29701901 DOI: 10.1002/jpen.1167] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/27/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Parenteral nutrition (PN) provides nutrition intravenously; however, this life-saving therapy is associated with significant liver disease. Recent evidence indicates improvement in PN-associated injury in animals with intact gut treated with enteral bile acid (BA), chenodeoxycholic acid (CDCA), and a gut farnesoid X receptor (FXR) agonist, which drives the gut-liver cross talk (GLCT). We hypothesized that similar improvement could be translated in animals with short bowel syndrome (SBS). METHODS Using piglets, we developed a novel 90% gut-resected SBS model. Fifteen SBS piglets receiving PN were given CDCA or control (vehicle control) for 2 weeks. Tissue and serum were analyzed posteuthanasia. RESULTS CDCA increased gut FXR (quantitative polymerase chain reaction; P = .008), but not downstream FXR targets. No difference in gut fibroblast growth factor 19 (FGF19; P = .28) or hepatic FXR (P = .75), FGF19 (P = .86), FGFR4 (P = .53), or Cholesterol 7 α-hydroxylase (P = .61) was noted. PN resulted in cholestasis; however, no improvement was noted with CDCA. Hepatic fibrosis or immunostaining for Ki67, CD3, or Cytokeratin 7 was not different with CDCA. PN resulted in gut atrophy. CDCA preserved (P = .04 vs control) gut mass and villous/crypt ratio. The median (interquartile range) for gut mass for control was 0.28 (0.17-0.34) and for CDCA was 0.33 (0.26-0.46). CONCLUSIONS We note that, unlike in animals with intact gut, in an SBS animal model there is inadequate CDCA-induced activation of gut-derived signaling to cause liver improvement. Thus, it appears that activation of GLCT is critically dependent on the presence of adequate gut. This is clinically relevant because it suggests that BA therapy may not be as effective for patients with SBS.
Collapse
Affiliation(s)
- Gustavo Villalona
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Amber Price
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Keith Blomenkamp
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | | | - Saurabh Saxena
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Thomas Ratchford
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Matthew Westrich
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Vindhya Kakarla
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Shruthika Pochampally
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - William Phillips
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Nicole Heafner
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Niraja Korremla
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Jose Greenspon
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Miguel A Guzman
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ajay Kumar Jain
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
26
|
MohanKumar K, Namachivayam K, Ho TT, Torres BA, Ohls RK, Maheshwari A. Cytokines and growth factors in the developing intestine and during necrotizing enterocolitis. Semin Perinatol 2017; 41:52-60. [PMID: 27832931 PMCID: PMC5334139 DOI: 10.1053/j.semperi.2016.09.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytokines and growth factors play diverse roles in the uninflamed fetal/neonatal intestinal mucosa and in the development of inflammatory bowel injury during necrotizing enterocolitis (NEC). During gestational development and the early neonatal period, the fetal/premature intestine is exposed to high levels of many "inflammatory" cytokines and growth factors, first via swallowed amniotic fluid in utero and then, after birth, in colostrum and mother's milk. This article reviews the dual, seemingly counter-intuitive roles of cytokines, where these agents play a "trophic" role and promote maturation of the uninflamed mucosa, but can also cause inflammation and promote intestinal injury during NEC.
Collapse
Affiliation(s)
| | | | - Thao T.B. Ho
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL
| | | | - Robin K. Ohls
- Department of Pediatrics, University of New Mexico, Albuquerque, NM
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL; Departments of Molecular Medicine, Morsani College of Medicine, Tampa, FL; Department of Community and Family Health, College of Public Health, University of South Florida, 1 Tampa General Circle, Suite F170, Tampa, FL.
| |
Collapse
|
27
|
Interdependency of EGF and GLP-2 Signaling in Attenuating Mucosal Atrophy in a Mouse Model of Parenteral Nutrition. Cell Mol Gastroenterol Hepatol 2017; 3:447-468. [PMID: 28462383 PMCID: PMC5403977 DOI: 10.1016/j.jcmgh.2016.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/31/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Total parenteral nutrition (TPN), a crucial treatment for patients who cannot receive enteral nutrition, is associated with mucosal atrophy, barrier dysfunction, and infectious complications. Glucagon-like peptide-2 (GLP-2) and epidermal growth factor (EGF) improve intestinal epithelial cell (IEC) responses and attenuate mucosal atrophy in several TPN models. However, it remains unclear whether these 2 factors use distinct or overlapping signaling pathways to improve IEC responses. We investigated the interaction of GLP-2 and EGF signaling in a mouse TPN model and in patients deprived of enteral nutrition. METHODS Adult C57BL/6J, IEC-Egfrknock out (KO) and IEC-pik3r1KO mice receiving TPN or enteral nutrition were treated with EGF or GLP-2 alone or in combination with reciprocal receptor inhibitors, GLP-2(3-33) or gefitinib. Jejunum was collected and mucosal atrophy and IEC responses were assessed by histologic, gene, and protein expression analyses. In patients undergoing planned looped ileostomies, fed and unfed ileum was analyzed. RESULTS Enteral nutrient deprivation reduced endogenous EGF and GLP-2 signaling in mice and human beings. In the mouse TPN model, exogenous EGF or GLP-2 attenuated mucosal atrophy and restored IEC proliferation. The beneficial effects of EGF and GLP-2 were decreased upon Gefitinib treatment and in TPN-treated IEC-EgfrKO mice, showing epidermal growth factor-receptor dependency for these IEC responses. By contrast, in TPN-treated IEC-pi3kr1KO mice, the beneficial actions of EGF were lost, although GLP-2 still attenuated mucosal atrophy. CONCLUSIONS Upon enteral nutrient deprivation, exogenous GLP-2 and EGF show strong interdependency for improving IEC responses. Understanding the differential requirements for phosphatidylinositol 3-kinase/phosphoAKT (Ser473) signaling may help improve future therapies to prevent mucosal atrophy.
Collapse
Key Words
- EGF
- EGF, epidermal growth factor
- EGFR
- EGFR, epidermal growth factor receptor
- GLP-2
- GLP-2 (3-33), glucagon-like peptide 2 antagonist
- GLP-2, glucagon-like peptide 2
- GLP2R, glucagon-like peptide 2 receptor
- IEC, intestinal epithelial cell
- IGF-1, insulin-like growth factor 1
- ISC, intestinal stem cell
- IV, intravenous
- KO, knock out
- Lgr5, leucine-rich repeat-containing G-protein–coupled receptor 5
- Mucosal Atrophy
- PCNA, proliferating cell nuclear antigen
- PCR, polymerase chain reaction
- PI3K
- PI3K, phosphatidylinositol 3-kinase
- PI3KR1, phosphatidylinositol 3-kinase p85a
- SBS, short-bowel syndrome
- TNF, tumor necrosis factor
- TPN, total parenteral nutrition
- TUNEL, terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling
- Total Parenteral Nutrition
- WT, wild-type
- bp, base pair
- mRNA, messenger RNA
Collapse
|
28
|
Lee J, Koehler J, Yusta B, Bahrami J, Matthews D, Rafii M, Pencharz PB, Drucker DJ. Enteroendocrine-derived glucagon-like peptide-2 controls intestinal amino acid transport. Mol Metab 2017; 6:245-255. [PMID: 28271031 PMCID: PMC5324020 DOI: 10.1016/j.molmet.2017.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/11/2017] [Indexed: 12/28/2022] Open
Abstract
Objective Glucagon-like peptide-2 (GLP-2) is co-secreted with GLP-1 from gut endocrine cells, and both peptides act as growth factors to expand the surface area of the mucosal epithelium. Notably, GLP-2 also enhances glucose and lipid transport in enterocytes; however, its actions on control of amino acid (AA) transport remain unclear. Here we examined the mechanisms linking gain and loss of GLP-2 receptor (GLP-2R) signaling to control of intestinal amino acid absorption in mice. Methods Absorption, transport, and clearance of essential AAs, specifically lysine, were measured in vivo by Liquid Chromatography triple quadrupole Mass Spectrometry (LC-MS/MS) and ex vivo with Ussing chambers using intestinal preparations from Glp2r+/+ and Glp2r−/− mice. Immunoblotting determined jejunal levels of protein components of signaling pathways (PI3K-AKT, and mTORC1-pS6-p4E-BP1) following administration of GLP-2, protein gavage, and rapamycin to fasted Glp2r+/+ and Glp2r−/− mice. Expression of AA transporters from full thickness jejunum and 4F2hc from brush border membrane vesicles (BBMVs) was measured by real-time PCR and immunoblotting, respectively. Results Acute administration of GLP-2 increased basal AA absorption in vivo and augmented basal lysine transport ex vivo. GLP-2-stimulated lysine transport was attenuated by co-incubation with wortmannin, rapamycin, or tetrodotoxin ex vivo. Phosphorylation of mTORC1 effector proteins S6 and 4E-BP1 was significantly increased in wild-type mice in response to GLP-2 alone, or when co-administered with protein gavage, and abolished following oral gavage of rapamycin. In contrast, activation of GLP-1R signaling did not enhance S6 phosphorylation. Disruption of GLP-2 action in Glp2r−/− mice reduced lysine transport ex vivo and attenuated the phosphorylation of S6 and 4E-BP1 in response to oral protein. Moreover, the expression of cationic AA transporter slc7a9 in response to refeeding, and the abundance of 4F2hc in BBMVs following protein gavage, was significantly attenuated in Glp2r−/− mice. Conclusions These findings reveal an important role for GLP-2R signaling in the physiological and pharmacological control of enteral amino acid sensing and assimilation, defining an enteroendocrine cell-enterocyte axis for optimal energy absorption. GLP-2 promotes intestinal amino acid absorption in vivo. Intestinal amino acid absorption is reduced in Glp2r−/− mice. GLP-2 stimulates amino acid transport independently of blood flow. GLP-2, but not GLP-1, activates the mTORC1 signaling pathway. Amino acid transport by GLP-2 requires the enteric nervous system and mTORC1.
Collapse
Key Words
- 4E-BP1, eukaryotic translation initiation factor 4E (eIF4e)-binding protein 1
- AA, amino acid
- Amino acid absorption
- BBMV, brush border membrane vesicles
- EAA, essential amino acid
- EECs, enteroendocrine cells
- ENS, enteric nervous system
- GLP-1
- GLP-1, Glucagon-like peptide-1
- GLP-2
- GLP-2, glucagon-like peptide-2
- GLP-2R, GLP-2 receptor
- Gut peptides
- LC-MS/MS, liquid chromatography triple quadrupole mass spectrometry
- PGDP, proglucagon-derived peptides
- Rapamycin
- S6K1, 70 kDa ribosomal protein S6 kinase 1
- mTORC1, mechanistic target of rapamycin complex 1
Collapse
Affiliation(s)
- Jennifer Lee
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jacqueline Koehler
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Bernardo Yusta
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jasmine Bahrami
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Dianne Matthews
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Mahroukh Rafii
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Paul B Pencharz
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Daniel J Drucker
- Department of Medicine, University of Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
29
|
Baldassano S, Amato A, Mulè F. Influence of glucagon-like peptide 2 on energy homeostasis. Peptides 2016; 86:1-5. [PMID: 27664588 DOI: 10.1016/j.peptides.2016.09.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 02/06/2023]
Abstract
Glucagon like peptide-2 (GLP-2) is a gastrointestinal hormone released from enteroendocrine L-type cells together with glucagon like peptide-1 in response to dietary nutrients. GLP-2 acts through a specific receptor, the GLP-2 receptor, mainly located in the gut and in the brain. Classically, GLP-2 is considered a trophic hormone involved in the maintenance of intestinal epithelial morphology and function. This role has been targeted for therapies promoting repair and adaptive growth of the intestinal mucosa. Recently, GLP-2 has been shown to exert beneficial effects on glucose metabolism specially in conditions related to increased uptake of energy, such as obesity. Several actions of GLP-2 are related to a positive energy balance: GLP-2 increases not only the absorptive surface, but also expression and activity of epithelial brush-border nutrient transporters and digestive enzymes, intestinal blood flow, postprandial chylomicron secretion and it inhibits gastrointestinal motility, providing the opportunity to increase absorption of nutrients. Other actions, including anorexigenic effects, appear in opposition to the energy intake. In this review, we discuss the GLP-2 functions related to energy homeostasis. GLP-2 could be considered an hormone causing positive energy balance, which, however has the role to mitigate the metabolic dysfunctions associated with hyper-adiposity.
Collapse
Affiliation(s)
- Sara Baldassano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy
| | - Antonella Amato
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy
| | - Flavia Mulè
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, 90128, Italy.
| |
Collapse
|
30
|
Lim DW, Diané A, Muto M, Vine DF, Nation PN, Wizzard PR, Sigalet DL, Bigam DL, Pencharz PB, Turner JM, Wales PW. Differential Effects on Intestinal Adaptation Following Exogenous Glucagon-Like Peptide 2 Therapy With and Without Enteral Nutrition in Neonatal Short Bowel Syndrome. JPEN J Parenter Enteral Nutr 2016; 41:156-170. [DOI: 10.1177/0148607116665812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- David W. Lim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Abdoulaye Diané
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Mitsuru Muto
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Donna F. Vine
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Patrick N. Nation
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Pamela R. Wizzard
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - David L. Sigalet
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - David L. Bigam
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Paul B. Pencharz
- Department of Nutritional Sciences and Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Justine M. Turner
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Paul W. Wales
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, Hospital for Sick Children & University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Zhang Z, Wu X, Cao L, Zhong Z, Zhou Y. Generation of glucagon-like peptide-2-expressing Saccharomyces cerevisiae and its improvement of the intestinal health of weaned rats. Microb Biotechnol 2016; 9:846-857. [PMID: 27641625 PMCID: PMC5072200 DOI: 10.1111/1751-7915.12412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/15/2016] [Accepted: 08/20/2016] [Indexed: 02/05/2023] Open
Abstract
We aimed to assess the feasibility of enhancing the intestinal development of weaned rats using glucagon-like peptide-2 (GLP-2)-expressing Saccharomyces cerevisiae (S. cerevisiae). GLP-2-expressing S. cerevisiae (GLP2-SC) was generated using a recombinant approach. The diet of weaned rats was supplemented with the GLP2-SC strain. The average daily gain (ADG), the intestinal morphology and the activities of the digestive enzymes in the jejunum were tested to assess the influence of the GLP2-SC strain on intestinal development. The proliferation of rat enterocytes was also assessed in vitro. The study revealed that the ADG of the weaned rats that received GLP2-SC was significantly greater than that of the controls fed a basal diet (Control) and S. cerevisiae harbouring an empty vector (EV-SC) (P < 0.05) but was equivalent to that of positive control rats fed recombinant human GLP-2 (rh-GLP2) (P > 0.05). Furthermore, GLP2-SC significantly increased villous height (P < 0.01) and digestive enzyme activity (P < 0.05) in the jejunum. Immunohistochemistry analysis further affirmed that enterocyte proliferation was stimulated in rats fed the GLP2-SC strain, as indicated by the greater number of enterocytes stained with proliferative cell nuclear antigen (P < 0.05). In vitro, the proliferation of rat enterocytes was also stimulated by GLP-2 expressed by the GLP2-SC strain (P < 0.01). Herein, the combination of the GLP-2 approach and probiotic delivery constitute a possible dietary supplement for animals after weaning.
Collapse
Affiliation(s)
- Zhongwei Zhang
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaodong Wu
- Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, 610041, China
| | - Lili Cao
- Medical School, Chengdu University, Chengdu, Sichuan, 610041, China
| | - Zhengdong Zhong
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610041, China
| | - Yan Zhou
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
32
|
Thymann T. Endocrine regulation of gut maturation in early life in pigs. Domest Anim Endocrinol 2016; 56 Suppl:S90-3. [PMID: 27345327 DOI: 10.1016/j.domaniend.2016.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 01/17/2023]
Abstract
After birth, the newborn must adapt to the acute challenges of circulatory changes, active respiration, thermoregulation, microbial colonization, and enteral nutrition. Whereas these processes normally occur without clinical complications in neonates born at term, birth at a preterm state of gestation is associated with high morbidity and mortality. In commercial pig production, perinatal mortality is higher than in any other mammalian species. Asphyxia, hypothermia, hypoglycemia, sepsis, and gut dysmotility, represent some of the most common findings. The intestine is a particularly sensitive organ after birth, as it must adapt acutely to enteral nutrition and microbial colonization. Likewise, during the weaning phase, the intestine must adapt to new diet types. Both critical phases are associated with high morbidity. This review focuses on the endocrine changes occurring around birth and weaning. There are a number of endocrine adaptations in late gestation and early postnatal life that are under influence of development stage and environmental factors such as diet. The review discusses general endocrine changes in perinatal life but specifically focuses on the role of glucagon-like peptide-2. This gut-derived hormone plays a key role in development and function of the intestine in early life.
Collapse
Affiliation(s)
- T Thymann
- Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, Department of Clinical Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, DK-1870, Denmark.
| |
Collapse
|
33
|
Diao H, Gao Z, Yu B, Zheng P, He J, Yu J, Huang Z, Chen D, Mao X. Effects of benzoic acid (VevoVitall®) on the performance and jejunal digestive physiology in young pigs. J Anim Sci Biotechnol 2016; 7:32. [PMID: 27239300 PMCID: PMC4884408 DOI: 10.1186/s40104-016-0091-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 05/11/2016] [Indexed: 12/03/2022] Open
Abstract
Background As a organic acid, benzoic acid has become one of the most important alternatives for antibiotics, and its beneficial effect on performance in animals has been proven for a decade. However, knowledge of the effects of benzoic acid on jejunal digestive physiology, especially the antioxidant capacity and mucosal glucagon-like peptide 2 (GLP-2) concentrations is lacking. Methods A total of 20 barrows [Duroc × (Yorkshire × Landrace)] with an average body weight (BW) of 18.75 ± 0.2 kg were used in a 14-d trial to determine the potential mechanisms of benzoic acid supplementation on the performance, nutrient digestibility and jejunal digestive physiology in young pigs. All pigs were randomly allotted to 1 of 2 diets supplemented with 0 or 5000 mg/kg benzoic acid. Results Relative to the control, benzoic acid supplementation increased the average daily feed intake (ADFI), and average daily gain (ADG) in young pigs (P < 0.05), improved the apparent total tract digestibility of dry matter (DM), crude protein (CP), ether extract (EE), gross energy (GE) and crude ash (P < 0.05), and enhanced the activities of trypsin, lipase and amylase in the jejunum (P < 0.05). Similarly, relative to the control, supplementing benzoic acid in the diet resulted in a trend to reduce the pH values of the digesta (P = 0.06), decreased crypt depth and increased the villus height to crypt depth ratio (P < 0.05) in the jejunum of pigs. Finally, benzoic acid supplementation increased the mRNA expression and concentration of glucagon-like peptide 2 and the activities of glutathione peroxidase and superoxide dismutase in the jejunal mucosa of young pigs (P < 0.05). Conclusions In conclusion, supplementation with 5000 mg/kg benzoic acid improved the performance of young pigs through promoting nutrient digestion, improving jejunal antioxidant capacity, and maintaining the jejunal morphology in young pigs.
Collapse
Affiliation(s)
- Hui Diao
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Zengbing Gao
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Bing Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Ping Zheng
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Jun He
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Jie Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Zhiqing Huang
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Daiwen Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| | - Xiangbing Mao
- Animal Nutrition Institute, Sichuan Agricultural University, Xinkang Road 46#, Ya'an, Sichuan Province 625014 People's Republic of China
| |
Collapse
|
34
|
Deng QH, Jia G, Zhao H, Chen ZL, Chen XL, Liu GM, Wang KN. The prolonged effect of glucagon-like peptide 2 pretreatment on growth performance and intestinal development of weaned piglets. J Anim Sci Biotechnol 2016; 7:28. [PMID: 27148449 PMCID: PMC4855712 DOI: 10.1186/s40104-016-0087-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 04/19/2016] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Glucagon-like peptide 2 (GLP-2) is a potent epithelium-specific intestinal growth factor. The aim of this study was to demonstrate the prolonged effect of GLP-2 on the growth performance of weaned piglets. Forty piglets weaned at the age of 28 d with an average BW of 6.8 ± 0.4 kg were assigned to four treatments: (i) non-challenged control; (ii) LPS-challenged control; (iii) LPS + low GLP-2; and (iv) LPS + high GLP-2. Piglets in groups (i), (ii), and (iv) were s.c. injected with PBS supplemented with human [Gly2]GLP-21-34 at doses of 0, 2 and 10 nmol/kg BW per day for seven consecutive days. BW, gain:feed ratio (G:F), and plasma GLP-2 levels were determined on d 0, 7, and 14 after weaning. Piglets were challenged with i.p. administration of Escherichia coli lipopolysaccharide (LPS) at a dose of 100 μg/kg on d 14 to induce intestinal damage. Twenty-four hours later, intestinal tract samples were collected to assess intestinal morphology and quantify enzyme activity. RESULTS Plasma GLP-2 levels decreased after weaning, but in the high GLP-2 group, plasma GLP-2 was maintained on d 7 and even increased to a level higher than the preweaning level on d 14 (P < 0.05). High GLP-2 treatment significantly increased the duodenal, jejunal and ileal weight, as well as the gross weight of the small intestine (SI), and the SI weight index (P < 0.05). LPS caused villous atrophy and disrupted intestinal morphology in the duodenum, jejunum and ileum. GLP-2 also significantly increased the villus height and the villus height/crypt depth ratio (VCR) of the duodenum, jejunum, and ileum (P < 0.05). Histological examination revealed that in GLP-2-treated groups, the integrity of the villus was maintained, and the villus was protected against LPS-induced damage. GLP-2 significantly increased the activity of alkaline phosphatase (AKP), γ-glutamyltranspeptidase (γ-GT), and pancreatic lipase in the duodenum and jejunum (P < 0.05). GLP-2 treatment also significantly increased the average daily gain (ADG) and G:F of piglets at 0 to 7, 7 to 14, as well as 0 to14 d (P < 0.05), resulting in a significant increase of final BW in high GLP-2 pigs (P = 0.016). CONCLUSIONS Exogenous GLP-2 improved the growth of weaned piglets and protected them against LPS-induced intestinal damage. These effects may be due to the ability of GLP-2 to promote the secretion of endogenous GLP-2 to stimulate the small intestinal development.
Collapse
Affiliation(s)
- Qiu Hong Deng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130 China
| | - Gang Jia
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130 China
| | - Hua Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130 China
| | - Zheng Li Chen
- College of Animal Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130 China
| | - Xiao Ling Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130 China
| | - Guang Mang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130 China
| | - Kang Ning Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan 611130 China
| |
Collapse
|
35
|
Hansen CF, Thymann T, Andersen AD, Holst JJ, Hartmann B, Hilsted L, Langhorn L, Jelsing J, Sangild PT. Rapid gut growth but persistent delay in digestive function in the postnatal period of preterm pigs. Am J Physiol Gastrointest Liver Physiol 2016; 310:G550-60. [PMID: 26822913 PMCID: PMC4836131 DOI: 10.1152/ajpgi.00221.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/22/2016] [Indexed: 01/31/2023]
Abstract
Preterm infants often tolerate full enteral nutrition a few weeks after birth but it is not known how this is related to gut maturation. Using pigs as models, we hypothesized that intestinal structure and digestive function are similar in preterm and term individuals at 3-4 wk after birth and that early enteral nutrition promotes maturation. Preterm or term cesarean-delivered pigs were fed total parenteral nutrition, or partial enteral nutrition [Enteral (Ent), 16-64 ml·kg(-1)·day(-1) of bovine colostrum] for 5 days, followed by full enteral milk feeding until day 26 The intestine was collected for histological and biochemical analyses at days 0, 5, and 26 (n = 8-12 in each of 10 treatment groups). Intestinal weight (relative to body weight) was reduced in preterm pigs at 0-5 days but ENT feeding stimulated the mucosal volume and peptidase activities. Relative to term pigs, mucosal volume remained reduced in preterm pigs until 26 days although plasma glucagon-like peptide 2 (GLP-2) and glucose-dependent insulin-trophic peptide (GIP) levels were increased. Preterm pigs also showed reduced hexose absorptive capacity and brush-border enzyme (sucrase, maltase) activities at 26 days, relative to term pigs. Intestinal structure shows a remarkable growth adaptation in the first week after preterm birth, especially with enteral nutrition, whereas some digestive functions remain immature until at least 3-4 wk. It is important to identify feeding regimens that stimulate intestinal maturation in the postnatal period of preterm infants because some intestinal functions may show long-term developmental delay.
Collapse
Affiliation(s)
- Carl Frederik Hansen
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | - Thomas Thymann
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | | | - Jens Juul Holst
- 2Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Bolette Hartmann
- 2Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Linda Hilsted
- 4Department of Clinical Biochemistry, Copenhagen University Hospital, Cophenhagen Denmark; and
| | - Louise Langhorn
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark;
| | | | - Per Torp Sangild
- 1Comparative Pediatrics and Nutrition, University of Copenhagen, Copenhagen, Denmark; ,5Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Denmark
| |
Collapse
|
36
|
Lim DW, Wales PW, Turner JM, Bigam DL, Brubaker PL. On the horizon: trophic peptide growth factors as therapy for neonatal short bowel syndrome. Expert Opin Ther Targets 2016; 20:819-30. [DOI: 10.1517/14728222.2016.1146695] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- David W. Lim
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Paul W. Wales
- Department of Surgery, University of Toronto & Hospital for Sick Children, Toronto, ON, Canada
| | - Justine M. Turner
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - David L. Bigam
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Patricia L. Brubaker
- Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
GLP-2 Prevents Intestinal Mucosal Atrophy and Improves Tissue Antioxidant Capacity in a Mouse Model of Total Parenteral Nutrition. Nutrients 2016; 8:nu8010033. [PMID: 26761030 PMCID: PMC4728647 DOI: 10.3390/nu8010033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 01/04/2023] Open
Abstract
We investigated the effects of exogenous glucagon-like peptide-2 (GLP-2) on mucosal atrophy and intestinal antioxidant capacity in a mouse model of total parenteral nutrition (TPN). Male mice (6–8 weeks old) were divided into three groups (n = 8 for each group): a control group fed a standard laboratory chow diet, and experimental TPN (received standard TPN solution) and TPN + GLP-2 groups (received TPN supplemented with 60 µg/day of GLP-2 for 5 days). Mice in the TPN group had lower body weight and reduced intestinal length, villus height, and crypt depth compared to the control group (all p < 0.05). GLP-2 supplementation increased all parameters compared to TPN only (all p < 0.05). Intestinal total superoxide dismutase activity and reduced-glutathione level in the TPN + GLP-2 group were also higher relative to the TPN group (all p < 0.05). GLP-2 administration significantly upregulated proliferating cell nuclear antigen expression and increased glucose-regulated protein (GRP78) abundance. Compared with the control and TPN + GLP-2 groups, intestinal cleaved caspase-3 was increased in the TPN group (all p < 0.05). This study shows GLP-2 reduces TPN-associated intestinal atrophy and improves tissue antioxidant capacity. This effect may be dependent on enhanced epithelial cell proliferation, reduced apoptosis, and upregulated GRP78 expression.
Collapse
|
38
|
Porcine models of digestive disease: the future of large animal translational research. Transl Res 2015; 166:12-27. [PMID: 25655839 PMCID: PMC4458388 DOI: 10.1016/j.trsl.2015.01.004] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/03/2015] [Accepted: 01/07/2015] [Indexed: 12/14/2022]
Abstract
There is increasing interest in nonrodent translational models for the study of human disease. The pig, in particular, serves as a useful animal model for the study of pathophysiological conditions relevant to the human intestine. This review assesses currently used porcine models of gastrointestinal physiology and disease and provides a rationale for the use of these models for future translational studies. The pig has proven its utility for the study of fundamental disease conditions such as ischemia-reperfusion injury, stress-induced intestinal dysfunction, and short bowel syndrome. Pigs have also shown great promise for the study of intestinal barrier function, surgical tissue manipulation and intervention, as well as biomaterial implantation and tissue transplantation. Advantages of pig models highlighted by these studies include the physiological similarity to human intestine and mechanisms of human disease. Emerging future directions for porcine models of human disease include the fields of transgenics and stem cell biology, with exciting implications for regenerative medicine.
Collapse
|
39
|
PEGylated porcine glucagon-like peptide-2 improved the intestinal digestive function and prevented inflammation of weaning piglets challenged with LPS. Animal 2015; 9:1481-9. [PMID: 25963800 DOI: 10.1017/s1751731115000749] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This study was conducted to determine the effects on intestinal function, anti-inflammatory role and possible mechanism of polyethylene glycosylated (PEGylated) porcine glucagon-like peptide-2 (pGLP-2), a long-acting form of pGLP-2, in weaning piglets challenged with Escherichia coli lipopolysaccharide (LPS). We divided 18 weaned piglets on day 21 into three groups (control, LPS and LPS+PEG-pGLP-2; n=6). The piglets from the LPS+PEG-pGLP-2 group were injected with PEG-pGLP-2 at 10 nmol/kg BW from 5 to 7 days of the trials daily. On 8th day, the piglets in the LPS and LPS+PEG-pGLP-2 groups were intraperitoneally administered with 100 µg LPS/kg. The control group was administered with the same volume of saline solution. The piglets were then sacrificed on day 28. Afterwards, serum, duodenum, jejunum and ileum samples were collected for analysis of structural and functional endpoints. LPS+PEG-pGLP-2 treatment increased (P<0.05) lactase activities in the duodenum and the jejunum compared with LPS treatment. LPS+PEG-pGLP-2 treatment also significantly increased sucrase activity in the jejunum compared with LPS treatment. Furthermore, LPS treatment increased (P<0.05) the mRNA expression levels of interleukin (IL)-8, tumour necrosis factor-α (TNF-α) and IL-10 in the ileum compared with the control treatment. By contrast, LPS+PEG-pGLP-2 treatment decreased (P<0.05) the mRNA expression levels of IL-8, IL-10 and TNF-α in the ileum compared with the LPS treatment. LPS treatment also increased (P<0.05) the mRNA expression level of GLP-2 receptor (GLP-2R) and the percentage of GLP-2R-positive cells in the ileum; by comparison, these results were (P<0.05) reduced by LPS+PEG-pGLP-2 treatment. Moreover, LPS+PEG-pGLP-2 treatment increased (P<0.05) the content of serum keratinocyte growth factor compared with the control group and the LPS group. The protective effects of PEG-pGLP-2 on intestinal digestive function were associated with the release of GLP-2R mediator (keratinocyte growth factor) and the decrease in the expressions of intestinal pro-inflammatory cytokines.
Collapse
|
40
|
Bedford A, Chen T, Huynh E, Zhu C, Medeiros S, Wey D, de Lange C, Li J. Epidermal growth factor containing culture supernatant enhances intestine development of early-weaned pigs in vivo: potential mechanisms involved. J Biotechnol 2015; 196-197:9-19. [PMID: 25615942 DOI: 10.1016/j.jbiotec.2015.01.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/31/2014] [Accepted: 01/05/2015] [Indexed: 12/17/2022]
Abstract
We have previously generated epidermal factor expressing Lactococcus lactis (EGF-LL) using a bioengineering approach, and shown that EGF-LL fermentation supernatant enhanced newly weaned pigs growth. The objective of the current study was to further understand the mechanisms behind this improved performance. Sixty-four piglets were weaned at 3 weeks of age and then fed ad libitum according to a 2-phase feeding program. Four pens with 8 pigs per pen were assigned to each of two treatments for 3 weeks: (1) EGF containing supernatant from EGF-LL culture (SuperEGF) or (2) blank M17GE media (Control). Consistent with previous findings, SuperEGF pigs had an increased average daily gain during week 3 post-weaning (433.4 ± 10.86 vs 388.7 ± 7.76 g; P<0.05) and overall gain:feed ratio (0.757 ± 0.03 vs 0.677 ± 0.01 kg/kg, P < 0.05). Moreover, jejunal structure development was enhanced, and inflammation index was minimized in SuperEGF pigs as indicated by increased villi height (P<0.05), decreased lamina propria width (P<0.05), and higher expression of anti-inflammatory cytokine, IL-13 (P<0.05). Further, goblet cell numbers and Muc2 levels were increased in SuperEGF pigs. Interestingly, the weaning-induced decrease of glucose cotransporter sodium-glucose linked transporter 1 (SGLT1) and glucagon-like peptide-2 (GLP2) levels was reversed by SuperEGF supplementation. Our findings add to our understanding of the mechanisms behind enhancing piglet performance by EGF containing fermentation product.
Collapse
Affiliation(s)
- Andrea Bedford
- Department of Animal and Poultry Science, University of Guelph, Canada
| | - Tao Chen
- Department of Animal and Poultry Science, University of Guelph, Canada; College of Veterinary Medicine, Hunan Agricultural University, PR China
| | - Evanna Huynh
- Department of Animal and Poultry Science, University of Guelph, Canada
| | - Cuilan Zhu
- Department of Animal and Poultry Science, University of Guelph, Canada
| | - Samantha Medeiros
- Department of Animal and Poultry Science, University of Guelph, Canada
| | - Doug Wey
- Department of Animal and Poultry Science, University of Guelph, Canada
| | - Cornelis de Lange
- Department of Animal and Poultry Science, University of Guelph, Canada
| | - Julang Li
- Department of Animal and Poultry Science, University of Guelph, Canada.
| |
Collapse
|
41
|
Sangild PT, Ney DM, Sigalet DL, Vegge A, Burrin D. Animal models of gastrointestinal and liver diseases. Animal models of infant short bowel syndrome: translational relevance and challenges. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1147-68. [PMID: 25342047 PMCID: PMC4269678 DOI: 10.1152/ajpgi.00088.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal failure (IF), due to short bowel syndrome (SBS), results from surgical resection of a major portion of the intestine, leading to reduced nutrient absorption and need for parenteral nutrition (PN). The incidence is highest in infants and relates to preterm birth, necrotizing enterocolitis, atresia, gastroschisis, volvulus, and aganglionosis. Patient outcomes have improved, but there is a need to develop new therapies for SBS and to understand intestinal adaptation after different diseases, resection types, and nutritional and pharmacological interventions. Animal studies are needed to carefully evaluate the cellular mechanisms, safety, and translational relevance of new procedures. Distal intestinal resection, without a functioning colon, results in the most severe complications and adaptation may depend on the age at resection (preterm, term, young, adult). Clinically relevant therapies have recently been suggested from studies in preterm and term PN-dependent SBS piglets, with or without a functional colon. Studies in rats and mice have specifically addressed the fundamental physiological processes underlying adaptation at the cellular level, such as regulation of mucosal proliferation, apoptosis, transport, and digestive enzyme expression, and easily allow exogenous or genetic manipulation of growth factors and their receptors (e.g., glucagon-like peptide 2, growth hormone, insulin-like growth factor 1, epidermal growth factor, keratinocyte growth factor). The greater size of rats, and especially young pigs, is an advantage for testing surgical procedures and nutritional interventions (e.g., PN, milk diets, long-/short-chain lipids, pre- and probiotics). Conversely, newborn pigs (preterm or term) and weanling rats provide better insights into the developmental aspects of treatment for SBS in infants owing to their immature intestines. The review shows that a balance among practical, economical, experimental, and ethical constraints will determine the choice of SBS model for each clinical or basic research question.
Collapse
Affiliation(s)
- Per T. Sangild
- 1Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark; ,2Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark;
| | - Denise M. Ney
- 3Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin;
| | | | - Andreas Vegge
- 1Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark; ,5Diabetes Pharmacology, Novo Nordisk, Måløv, Denmark; and
| | - Douglas Burrin
- 6USDA-ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
42
|
Lim DW, Wales PW, Josephson JK, Nation PN, Wizzard PR, Sergi CM, Field CJ, Sigalet DL, Turner JM. Glucagon-Like Peptide 2 Improves Cholestasis in Parenteral Nutrition--Associated Liver Disease. JPEN J Parenter Enteral Nutr 2014; 40:14-21. [PMID: 25280755 DOI: 10.1177/0148607114551968] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 08/27/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Parenteral nutrition-associated liver disease (PNALD) remains a significant cause of morbidity and mortality in neonates with intestinal failure. Although glucagon-like peptide-2 (GLP-2) is being advanced as therapy, the effect of GLP-2 treatment on PNALD is unknown. We aim to investigate the effect of exogenous GLP-2 administration on hepatic function in a neonatal piglet model of PNALD. METHODS Neonatal piglets (aged 2-6 days) underwent jugular venous catheterization to receive isonitrogenous, isocaloric parenteral nutrition (PN). Piglets were allocated to 2 groups: group 1 (n = 8) received saline while group 2 (n = 7) received GLP-2 (at 11 nmol/kg/d). After 17 days, piglets underwent terminal laparotomy, and bile flow was measured. Liver specimens were analyzed histologically and with immunoperoxidase staining. Age-matched sow-reared control piglets (group 3, n = 8) were used for comparison. RESULTS Both groups 1 and 2 receiving PN developed cholestasis relative to sow-reared controls, as evidenced by a decrease in bile flow and increase in serum total bilirubin. However, group 2 had improved bile flow (1.35 vs 0.73 µL/g; P = .02) and diminished bilirubin (38.0 vs 78.5 µmol/L; P = .008) compared with group 1. Group 2 also had lower serum alanine aminotransferase levels, a marker of liver injury. Histologically, the liver specimens in group 1 had marked hepatocyte pigmentation, which was decreased in group 2 specimens. CONCLUSIONS The exogenous administration of GLP-2 is associated with the improvement of cholestasis and liver injury. This study introduces a novel role for GLP-2 in improving PNALD in the setting of prolonged PN duration.
Collapse
Affiliation(s)
- David W Lim
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Paul W Wales
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada Department of Surgery, Hospital for Sick Children & University of Toronto, Toronto, Ontario, Canada
| | | | - Patrick N Nation
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Pamela R Wizzard
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Consolato M Sergi
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - David L Sigalet
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Justine M Turner
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada Department of Agricultural Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
43
|
Moran A, Al-Rammahi M, Zhang C, Bravo D, Calsamiglia S, Shirazi-Beechey S. Sweet taste receptor expression in ruminant intestine and its activation by artificial sweeteners to regulate glucose absorption. J Dairy Sci 2014; 97:4955-72. [DOI: 10.3168/jds.2014-8004] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/23/2014] [Indexed: 12/30/2022]
|
44
|
Acute effects of the glucagon-like peptide 2 analogue, teduglutide, on intestinal adaptation in short bowel syndrome. J Pediatr Gastroenterol Nutr 2014; 58:694-702. [PMID: 24399211 DOI: 10.1097/mpg.0000000000000295] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neonatal short bowel syndrome following massive gut resection is associated with malabsorption of nutrients. The intestinotrophic factor glucagon-like peptide 2 (GLP-2) improves gut function in adult patients with short bowel syndrome, but its effect in pediatric patients remains unknown. Our objective was to test the efficacy of the long-acting synthetic human GLP-2 analogue, teduglutide (ALX-0600), in a neonatal piglet jejunostomy model. Two-day-old pigs were subjected to resection of 50% of the small intestine (distal part), and the remnant intestine was exteriorized on the abdominal wall as a jejunostomy. All pigs were given total parenteral nutrition for 7 days and a single daily injection of the following doses of teduglutide: 0.01 (n = 6), 0.02 (n = 6), 0.1 (n = 5), or 0.2 mg · kg · day (n = 6), and compared with placebo (n = 9). Body weight increment was similar for all 4 teduglutide groups but higher than placebo (P < 0.05). There was a dose-dependent increase in weight per length of the remnant intestine (P < 0.01) and fractional protein synthesis rate in the intestine was increased in the 0.2 mg · kg · day group versus placebo (P < 0.001); however, functional and structural endpoints including activity of digestive enzymes, absorption of enteral nutrients, and immunohistochemistry (Ki67, villin, FABP2, ChgA, and GLP-2R) were not affected by the treatment. Teduglutide induces trophicity on the remnant intestine but has limited acute effects on functional endpoints. Significant effects of teduglutide on gut function may require a longer adaptation period and/or a more frequent administration of the peptide. In perspective, GLP-2 or its analogues may be relevant to improve intestinal adaptation in pediatric patients with short bowel syndrome.
Collapse
|
45
|
Thymann T, Le Huërou-Luron I, Petersen YM, Hedemann MS, Elinf J, Jensen BB, Holst JJ, Hartmann B, Sangild PT. Glucagon-like peptide 2 treatment may improve intestinal adaptation during weaning. J Anim Sci 2014; 92:2070-9. [PMID: 24663206 DOI: 10.2527/jas.2013-7015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transition from sow's milk to solid feed is associated with intestinal atrophy and diarrhea. We hypothesized that the intestinotrophic hormone glucagon-like peptide 2 (GLP-2) would induce a dose- and health status-dependent effect on gut adaptation. In Exp. 1, weaned pigs (average BW at weaning 4.98 ± 0.18 kg) were kept in a high-sanitary environment and injected with saline or short-acting GLP-2 (80 μg/(kg BW·12 h); n = 8). Under these conditions, there was no diarrhea and GLP-2 did not improve gastrointestinal structure or function. In Exp. 2, weaned pigs (average BW at weaning 6.68 ± 0.27 kg) were kept in a low-sanitary environment, leading to weaning diarrhea, and injected with saline or short-acting GLP-2 (200 µg/(kg BW·12 h); n = 11). Treatment with GLP-2 increased goblet cell density (P < 0.05) and reduced short chain fatty acid concentration in the colon (P < 0.01) but had limited effects on diarrhea. In Exp. 3, weaned pigs (average BW at weaning 6.90 ± 0.32 kg) were kept in a low-sanitary environment and injected with saline or a long-acting acylated GLP-2 analogue (25 µg/(kg BW·12 h); n = 8). In this experiment, GLP-2 increased intestinal weight (+22%; P < 0.01) and activity of brush border enzymes (+50-100%; P < 0.05). Circulating GLP-2 levels were in the pharmacological range in Exp. 3 (constant levels >20,000 pmol/L) and Exp. 2 (increases to 20,000 pmol/L for a few hours each day) while they were in the supraphysiological range in Exp. 1 (50-200 pmol/L). In conclusion, GLP-2 may improve gut structure and function in weanling pigs. However, the effects may be significant only under conditions of diarrhea and if GLP-2 exposure time is extended using long-acting analogues.
Collapse
Affiliation(s)
- T Thymann
- University of Copenhagen, Dep. of Human Nutrition, Exercise and Sports, DK-1958 Frederiksberg, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
l-Methionine supplementation maintains the integrity and barrier function of the small-intestinal mucosa in post-weaning piglets. Amino Acids 2014; 46:1131-42. [DOI: 10.1007/s00726-014-1675-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 01/15/2014] [Indexed: 11/27/2022]
|
47
|
Drucker DJ, Yusta B. Physiology and pharmacology of the enteroendocrine hormone glucagon-like peptide-2. Annu Rev Physiol 2013; 76:561-83. [PMID: 24161075 DOI: 10.1146/annurev-physiol-021113-170317] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glucagon-like peptide-2 (GLP-2) is a 33-amino-acid proglucagon-derived peptide secreted from enteroendocrine L cells. GLP-2 circulates at low basal levels in the fasting period, and plasma levels rise rapidly after food ingestion. Renal clearance and enzymatic inactivation control the elimination of bioactive GLP-2. GLP-2 increases mesenteric blood flow and activates proabsorptive pathways in the gut, facilitating nutrient absorption. GLP-2 also enhances gut barrier function and induces proliferative and cytoprotective pathways in the small bowel. The actions of GLP-2 are transduced via a single G protein-coupled receptor (GLP-2R), expressed predominantly within the gastrointestinal tract. Disruption of GLP-2R signaling increases susceptibility to gut injury and impairs the adaptive mucosal response to refeeding. Sustained augmentation of GLP-2R signaling reduces the requirement for parenteral nutrition in human subjects with short-bowel syndrome. Hence GLP-2 integrates nutrient-derived signals to optimize mucosal integrity and energy absorption.
Collapse
Affiliation(s)
- Daniel J Drucker
- Department of Medicine, Mount Sinai Hospital, Lunenfeld Tanenbaum Research Institute, University of Toronto, Toronto, Ontario, Canada M5G 1X5; ,
| | | |
Collapse
|
48
|
Vegge A, Thymann T, Lund P, Stoll B, Bering SB, Hartmann B, Jelsing J, Qvist N, Burrin DG, Jeppesen PB, Holst JJ, Sangild PT. Glucagon-like peptide-2 induces rapid digestive adaptation following intestinal resection in preterm neonates. Am J Physiol Gastrointest Liver Physiol 2013; 305:G277-85. [PMID: 23764891 PMCID: PMC4073902 DOI: 10.1152/ajpgi.00064.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Short bowel syndrome (SBS) is a frequent complication after intestinal resection in infants suffering from intestinal disease. We tested whether treatment with the intestinotrophic hormone glucagon-like peptide-2 (GLP-2) increases intestinal volume and function in the period immediately following intestinal resection in preterm pigs. Preterm pigs were fed enterally for 48 h before undergoing resection of 50% of the small intestine and establishment of a jejunostomy. Following resection, pigs were maintained on total parenteral nutrition (TPN) without (SBS, n = 8) or with GLP-2 treatment (3.5 μg/kg body wt per h, SBS+GLP-2, n = 7) and compared with a group of unresected preterm pigs (control, n = 5). After 5 days of TPN, all piglets were fed enterally for 24 h, and a nutrient balance study was performed. Intestinal resection was associated with markedly reduced endogenous GLP-2 levels. GLP-2 increased the relative absorption of wet weight (46 vs. 22%), energy (79 vs. 64%), and all macronutrients (all parameters P < 0.05). These findings were supported by a 200% increase in sucrase and maltase activities, a 50% increase in small intestinal epithelial volume (P < 0.05), as well as increased DNA and protein contents and increased total protein synthesis rate in SBS+GLP-2 vs. SBS pigs (+100%, P < 0.05). Following intestinal resection in preterm pigs, GLP-2 induced structural and functional adaptation, resulting in a higher relative absorption of fluid and macronutrients. GLP-2 treatment may be a promising therapy to enhance intestinal adaptation and improve digestive function in preterm infants with jejunostomy following intestinal resection.
Collapse
Affiliation(s)
- Andreas Vegge
- Dept. of Human Nutrition, Faculty of Life Sciences, Univ. of Copenhagen, 30 Rolighedsvej, DK-1870 Frederiksberg C, Denmark.
| | - Thomas Thymann
- Departments of 1Nutrition, Exercise and Sports, Faculty of Science, and
| | - Pernille Lund
- Departments of 1Nutrition, Exercise and Sports, Faculty of Science, and
| | - Barbara Stoll
- 2USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas;
| | - Stine B. Bering
- Departments of 1Nutrition, Exercise and Sports, Faculty of Science, and
| | - Bolette Hartmann
- 3Biomedical Science, Faculty of Health and Medical Sciences University of Copenhagen, Frederiksberg, Denmark;
| | | | - Niels Qvist
- 5Surgical Department A, Odense University Hospital, Odense, Denmark; and
| | - Douglas G. Burrin
- 2USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas;
| | - Palle B. Jeppesen
- 6Department of Gastroenterology, Rigshospitalet, Copenhagen, Denmark
| | - Jens J. Holst
- 3Biomedical Science, Faculty of Health and Medical Sciences University of Copenhagen, Frederiksberg, Denmark;
| | - Per T. Sangild
- Departments of 1Nutrition, Exercise and Sports, Faculty of Science, and
| |
Collapse
|
49
|
Sangild PT, Thymann T, Schmidt M, Stoll B, Burrin DG, Buddington RK. Invited review: the preterm pig as a model in pediatric gastroenterology. J Anim Sci 2013; 91:4713-29. [PMID: 23942716 DOI: 10.2527/jas.2013-6359] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
At birth, the newborn mammal undergoes a transition from a sterile uterine environment with a constant nutrient supply, to a microbe-rich environment with intermittent oral intake of complex milk nutrients via the gastrointestinal tract (GIT). These functional challenges partly explain the relatively high morbidity and mortality of neonates. Preterm birth interrupts prenatal organ maturation, including that of the GIT, and increases disease risk. Exemplary is necrotizing enterocolitis (NEC), which is associated closely with GIT immaturity, enteral feeding, and bacterial colonization. Infants with NEC may require resection of the necrotic parts of the intestine, leading to short bowel syndrome (SBS), characterized by reduced digestive capacity, fluid loss, and dependency on parenteral nutrition. This review presents the preterm pig as a translational model in pediatric gastroenterology that has provided new insights into important pediatric diseases such as NEC and SBS. We describe protocols for delivery, care, and handling of preterm pigs, and show how the immature GIT responds to delivery method and different nutritional and therapeutic interventions. The preterm pig may also provide a sensitive model for postnatal adaptation of weak term piglets showing high mortality. Attributes of the preterm pig model include close similarities with preterm infants in body size, organ development, and many clinical features, thereby providing a translational advantage relative to rodent models of GIT immaturity. On the other hand, the need for a sow surgical facility, a piglet intensive care unit, and clinically trained personnel may limit widespread use of preterm pigs. Studies on organ adaptation in preterm pigs help to identify the physiological basis of neonatal survival for hypersensitive newborns and aid in defining the optimal diet and rearing conditions during the critical neonatal period.
Collapse
Affiliation(s)
- P T Sangild
- Department of Nutrition, Exercise, and Sports
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
OBJECTIVES Necrotizing enterocolitis (NEC) is complex disease thought to occur as a result of an immaturity of the gastrointestinal tract of preterm infants. Intestinal dysfunction induced by total parental nutrition (TPN) may increase the risk for NEC upon introduction of enteral feeding. We hypothesized that the intestinal trophic and anti-inflammatory actions previously ascribed to the gut hormone, glucagon-like peptide-2 (GLP-2), would reduce the incidence of NEC when given in combination with TPN in preterm piglets. METHODS Preterm, newborn piglets were nourished by TPN and infused continuously with either human GLP-2 (100 μg · kg⁻¹ · day⁻¹) or control saline for 2 days (n = 12/group). On day 3, TPN was discontinued and pigs were given orogastric formula feeding every 3 hours, and continued GLP-2 or control treatment until the onset of clinical signs of NEC for an additional 96 hours and tissue was collected for molecular and histological endpoints. RESULTS GLP-2 treatment delayed the onset of NEC but was unable to prevent a high NEC incidence (~70%) and severity that occurred in both groups. GLP-2-treated pigs had less histological injury and increased proximal intestinal weight and mucosal villus height, but not crypt depth or Ki-67-positive cells. Inflammatory markers of intestinal myeloperoxidase were unchanged and serum amyloid A levels were higher in GLP-2-treated pigs. CONCLUSIONS GLP-2 did not prevent NEC and a proinflammatory response despite some reduction in mucosal injury and increased trophic effect.
Collapse
|