1
|
Li X, Chen R, Wen J, Ji R, Chen X, Cao Y, Yu Y, Zhao C. The mechanisms in the gut microbiota regulation and type 2 diabetes therapeutic activity of resistant starches. Int J Biol Macromol 2024; 274:133279. [PMID: 38906356 DOI: 10.1016/j.ijbiomac.2024.133279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Resistant starch (RS) can potentially prevent type 2 diabetes through the modulation of intestinal microbiota and microbial metabolites. Currently, it has been wildly noted that altering the intestinal microbial composition and short-chain fatty acids levels can achieve therapeutic effects, although the specific mechanisms were rarely elucidated. This review systematically explores the structural characteristics of different RS, analyzes the cross-feeding mechanism utilized by intestinal microbiota, and outlines the pathways and targets of butyrate, a primary microbial metabolite, for treating diabetes. Different RS types may have a unique impact on microbiota composition and their cross-feeding, thus exploring regulatory mechanisms of RS on diabetes through intestinal flora interaction and their metabolites could pave the way for more effective treatment outcomes for host health. Furthermore, by understanding the mechanisms of strain-level cross-feeding and metabolites of RS, precise dietary supplementation methods targeted at intestinal composition and metabolites can be achieved to improve T2DM.
Collapse
Affiliation(s)
- Xiaoqing Li
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Ruoxin Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiahui Wen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ruya Ji
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Xu Chen
- School of Life and Health Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yigang Yu
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Chao Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
2
|
Gala K, Ghusn W, Abu Dayyeh BK. Gut motility and hormone changes after bariatric procedures. Curr Opin Endocrinol Diabetes Obes 2024; 31:131-137. [PMID: 38533785 DOI: 10.1097/med.0000000000000860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
PURPOSE OF REVIEW Metabolic and bariatric surgery (MBS) and endoscopic bariatric therapies (EBT) are being increasingly utilized for the management of obesity. They work through multiple mechanisms, including restriction, malabsorption, and changes in the gastrointestinal hormonal and motility. RECENT FINDINGS Roux-en-Y gastric bypass (RYGB) and laparoscopic sleeve gastrectomy (LSG) cause decrease in leptin, increase in GLP-1 and PYY, and variable changes in ghrelin (generally thought to decrease). RYGB and LSG lead to rapid gastric emptying, increase in small bowel motility, and possible decrease in colonic motility. Endoscopic sleeve gastroplasty (ESG) causes decrease in leptin and increase in GLP-1, ghrelin, and PYY; and delayed gastric motility. SUMMARY Understanding mechanisms of action for MBS and EBT is critical for optimal care of patients and will help in further refinement of these interventions.
Collapse
Affiliation(s)
- Khushboo Gala
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Wissam Ghusn
- Department of Internal Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| | - Barham K Abu Dayyeh
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
3
|
Miskelly MG, Lindqvist A, Piccinin E, Hamilton A, Cowan E, Nergård BJ, Del Giudice R, Ngara M, Cataldo LR, Kryvokhyzha D, Volkov P, Engelking L, Artner I, Lagerstedt JO, Eliasson L, Ahlqvist E, Moschetta A, Hedenbro J, Wierup N. RNA sequencing unravels novel L cell constituents and mechanisms of GLP-1 secretion in human gastric bypass-operated intestine. Diabetologia 2024; 67:356-370. [PMID: 38032369 PMCID: PMC10789678 DOI: 10.1007/s00125-023-06046-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/15/2023] [Indexed: 12/01/2023]
Abstract
AIMS/HYPOTHESIS Roux-en-Y gastric bypass surgery (RYGB) frequently results in remission of type 2 diabetes as well as exaggerated secretion of glucagon-like peptide-1 (GLP-1). Here, we assessed RYGB-induced transcriptomic alterations in the small intestine and investigated how they were related to the regulation of GLP-1 production and secretion in vitro and in vivo. METHODS Human jejunal samples taken perisurgically and 1 year post RYGB (n=13) were analysed by RNA-seq. Guided by bioinformatics analysis we targeted four genes involved in cholesterol biosynthesis, which we confirmed to be expressed in human L cells, for potential involvement in GLP-1 regulation using siRNAs in GLUTag and STC-1 cells. Gene expression analyses, GLP-1 secretion measurements, intracellular calcium imaging and RNA-seq were performed in vitro. OGTTs were performed in C57BL/6j and iScd1-/- mice and immunohistochemistry and gene expression analyses were performed ex vivo. RESULTS Gene Ontology (GO) analysis identified cholesterol biosynthesis as being most affected by RYGB. Silencing or chemical inhibition of stearoyl-CoA desaturase 1 (SCD1), a key enzyme in the synthesis of monounsaturated fatty acids, was found to reduce Gcg expression and secretion of GLP-1 by GLUTag and STC-1 cells. Scd1 knockdown also reduced intracellular Ca2+ signalling and membrane depolarisation. Furthermore, Scd1 mRNA expression was found to be regulated by NEFAs but not glucose. RNA-seq of SCD1 inhibitor-treated GLUTag cells identified altered expression of genes implicated in ATP generation and glycolysis. Finally, gene expression and immunohistochemical analysis of the jejunum of the intestine-specific Scd1 knockout mouse model, iScd1-/-, revealed a twofold higher L cell density and a twofold increase in Gcg mRNA expression. CONCLUSIONS/INTERPRETATION RYGB caused robust alterations in the jejunal transcriptome, with genes involved in cholesterol biosynthesis being most affected. Our data highlight SCD as an RYGB-regulated L cell constituent that regulates the production and secretion of GLP-1.
Collapse
Affiliation(s)
- Michael G Miskelly
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Andreas Lindqvist
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Elena Piccinin
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Bari, Italy
| | - Alexander Hamilton
- Molecular Metabolism, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Elaine Cowan
- Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | | | - Rita Del Giudice
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Biomedical Science and Biofilms - Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Mtakai Ngara
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Luis R Cataldo
- Molecular Metabolism, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dmytro Kryvokhyzha
- Bioinformatics Unit, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Petr Volkov
- Bioinformatics Unit, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Luke Engelking
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Isabella Artner
- Endocrine Cell Differentiation and Function, Stem Cell Centre, Lund University, Malmö, Sweden
| | - Jens O Lagerstedt
- Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Emma Ahlqvist
- Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Bari, Italy
- INBB National Institute for Biostructure and Biosystems, Rome, Italy
| | - Jan Hedenbro
- Department of Surgery, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Nils Wierup
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Lund University, Malmö, Sweden.
| |
Collapse
|
4
|
Reghupaty SC, Dall NR, Svensson KJ. Hallmarks of the metabolic secretome. Trends Endocrinol Metab 2024; 35:49-61. [PMID: 37845120 PMCID: PMC10841501 DOI: 10.1016/j.tem.2023.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
The identification of novel secreted factors is advancing at an unprecedented pace. However, there is a critical need to consolidate and integrate this knowledge to provide a framework of their diverse mechanisms, functional significance, and inter-relationships. Complicating this effort are challenges related to nonstandardized methods, discrepancies in sample handling, and inconsistencies in the annotation of unknown molecules. This Review aims to synthesize the rapidly expanding field of the metabolic secretome, encompassing the five major types of secreted factors: proteins, peptides, metabolites, lipids, and extracellular vesicles. By systematically defining the functions and detection of the components within the metabolic secretome, this Review provides a primer into the advances of the field, and how integration of the techniques discussed can provide a deeper understanding of the mechanisms underlying metabolic homeostasis and its disorders.
Collapse
Affiliation(s)
- Saranya C Reghupaty
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Nicholas R Dall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA.
| |
Collapse
|
5
|
Kehagias D, Lampropoulos C, Georgopoulos N, Habeos I, Kalavrizioti D, Vamvakas SS, Davoulou P, Kehagias I. Diabetes Remission After LRYGBP With and Without Fundus Resection: a Randomized Clinical Trial. Obes Surg 2023; 33:3373-3382. [PMID: 37783932 PMCID: PMC10602944 DOI: 10.1007/s11695-023-06857-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND Glycemic control, after metabolic surgery, is achieved in two stages, initially with neuroendocrine alterations and in the long-term with sustainable weight loss. The resection of the gastric fundus, as the major site of ghrelin production, is probably related with optimized glucose regulation. The aim of the present study is to investigate whether the modification of laparoscopic Roux-en-Y gastric bypass (LRYGBP) with fundus resection offers superior glycemic control, compared to typical LRYGBP. MATERIALS AND METHODS Participants were 24 patients with body mass index (BMI) ≥40kg/m2 and type II diabetes mellitus (T2DM), who were randomly assigned to undergo LRYGBP and LRYGBP with fundus resection (LRYGBP+FR). Gastrointestinal (GI) hormones [ghrelin, glucagon-like-peptide-1 (GLP-1), peptide-YY (PYY)] and glycemic parameters (glucose, insulin, HbA1c, C-peptide, insulinogenic index, HOMA-IR) were measured preoperatively, at 6 and 12 months during an oral glucose tolerance test (OGTT). RESULTS Ninety-five percent of patients showed complete remission of T2DM after 12 months. LRYGBP+FR was not related with improved glycemic control, compared to LRYGBP. Ghrelin levels were not significantly reduced at 6 and 12 months after LRYGBP+FR. GLP-1 and PYY levels were remarkably increased postprandially in both groups at 6 and 12 months postoperatively (p<0.01). Patients who underwent LRYGBP+FR achieved a significantly lower BMI at 12 months in comparison to LRYGBP (p<0.05). CONCLUSION Fundus resection is not associated with improved glycemic regulation, compared to typical LRYGBP and the significant decrease in BMI after LRYGBP+FR has to be further confirmed with longer follow-up.
Collapse
Affiliation(s)
| | | | - Neoklis Georgopoulos
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology, University of Patras Medical School, 26504, Rio, Greece
| | - Ioannis Habeos
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital of Patras, 26504, Rio, Greece
| | - Dimitra Kalavrizioti
- Department of Nephrology and Renal Transplantation, University Hospital of Patras, 26504, Rio, Greece
| | | | - Panagiota Davoulou
- Department of Nephrology and Renal Transplantation, University Hospital of Patras, 26504, Rio, Greece
| | - Ioannis Kehagias
- Department of Surgery, Division of Bariatric and Metabolic Surgery, University Hospital of Patras, 26504, Rio, Greece
| |
Collapse
|
6
|
Chen W, Binbin G, Lidan S, Qiang Z, Jing H. Evolution of peptide YY analogs for the management of type 2 diabetes and obesity. Bioorg Chem 2023; 140:106808. [PMID: 37666110 DOI: 10.1016/j.bioorg.2023.106808] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/13/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Peptide YY (PYY) is a gastrointestinal hormone consisting of 36 amino acids, that is predominantly secreted by intestinal l-cells. Originally extracted from pig intestines, it belongs to the pancreatic polypeptide (PP) family, but has functions distinct from those of PP and neuropeptide Y (NPY). PYY is a potential treatment for type 2 diabetes mellitus (T2DM) because of its ability to delay gastric emptying, reduce appetite, decrease weight, and lower blood glucose. However, the clinical use of PYY is limited because it is rapidly cleared by the kidneys and degraded by enzymes. In recent years, researchers have conducted various structural modifications, including amino acid substitution, PEGylation, lipidation, and fusion of PYY with other proteins to prolong its half-life and enhance its biological activity. This study presents an overview of the recent progress on PYY, including its physiological functions, metabolites and structure-activity relationships.
Collapse
Affiliation(s)
- Wang Chen
- College of Medicine, Jiaxing University, The Affiliated Hospital of Jiaxing University, Jiaxing 314001, China
| | - Gong Binbin
- College of Pharmacy, Zhejiang University of Technology, Hangzhou 310000, China
| | - Sun Lidan
- College of Medicine, Jiaxing University, The Affiliated Hospital of Jiaxing University, Jiaxing 314001, China.
| | - Zhou Qiang
- College of Medicine, Jiaxing University, The Affiliated Hospital of Jiaxing University, Jiaxing 314001, China.
| | - Han Jing
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou 221116, China
| |
Collapse
|
7
|
Sridhar A, Khan D, Flatt PR, Irwin N, Moffett RC. PYY (3-36) protects against high fat feeding induced changes of pancreatic islet and intestinal hormone content and morphometry. Biochim Biophys Acta Gen Subj 2023; 1867:130359. [PMID: 37001706 DOI: 10.1016/j.bbagen.2023.130359] [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: 01/16/2023] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Prolonged high fat feeding negatively impacts pancreatic and intestinal morphology. In this regard, direct effects of PYY(3-36) on intestinal cell and pancreatic islet morphometry are yet to be fully explored in the setting of obesity. METHODS We examined the influence of 21-days twice daily treatment with PYY(3-36) on these parameters in mice fed a high fat diet (HFD). RESULTS PYY(3-36) treatment decreased food intake, body weight and circulating glucose in HFD mice. In terms of intestinal morphology, crypt depth was restored to control levels by PYY(3-36), with an additional enlargement of villi length. PYY(3-36) also reversed HFD-induced decreases of ileal PYY, and especially GLP-1, content. HFD increased numbers of PYY and GIP positive ileal cells, with PYY(3-36) fully reversing the effect on PYY cell detection. There were no obvious differences in the overall number of GLP-1 positive ileal cells in all mice, barring PYY(3-36) marginally decreasing GLP-1 villi cell immunoreactivity. Within pancreatic islets, PYY(3-36) significantly decreased alpha-cell area, whilst islet, beta-, PYY- and delta-cell areas remained unchanged. However, PYY(3-36) increased the percentage of beta-cells while also reducing percentage alpha-cell area. This was related to PYY(3-36)-induced reductions of beta-cell proliferation and apoptosis frequencies. Co-localisation of islet PYY with glucagon or somatostatin was elevated by PYY(3-36), with GLP-1/glucagon co-visualisation increased when compared to lean controls. CONCLUSION PYY(3-36) exerts protective effects on pancreatic and intestinal morphology in HFD mice linked to elevated ileal GLP-1 content. GENERAL SIGNIFICANCE These observations highlight mechanisms linked to the metabolic and weight reducing benefits of PYY(3-36).
Collapse
Affiliation(s)
- A Sridhar
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - D Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - P R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - N Irwin
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK.
| | - R C Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| |
Collapse
|
8
|
Kamakura R, Raza GS, Sodum N, Lehto V, Kovalainen M, Herzig K. Colonic Delivery of Nutrients for Sustained and Prolonged Release of Gut Peptides: A Novel Strategy for Appetite Management. Mol Nutr Food Res 2022; 66:e2200192. [PMID: 35938221 PMCID: PMC9787473 DOI: 10.1002/mnfr.202200192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/17/2022] [Indexed: 12/30/2022]
Abstract
Obesity is one of the major global threats to human health and risk factors for cardiometabolic diseases and certain cancers. Glucagon-like peptide-1 (GLP-1) plays a major role in appetite and glucose homeostasis and recently the USFDA approved GLP-1 agonists for the treatment of obesity and type 2 diabetes. GLP-1 is secreted from enteroendocrine L-cells in the distal part of the gastrointestinal (GI) tract in response to nutrient ingestion. Endogenously released GLP-1 has a very short half-life of <2 min and most of the nutrients are absorbed before reaching the distal GI tract and colon, which hinders the use of nutritional compounds for appetite regulation. The review article focuses on nutrients that endogenously stimulate GLP-1 and peptide YY (PYY) secretion via their receptors in order to decrease appetite as preventive action. In addition, various delivery technologies such as pH-sensitive, mucoadhesive, time-dependent, and enzyme-sensitive systems for colonic targeting of nutrients delivery are described. Sustained colonic delivery of nutritional compounds could be one of the most promising approaches to prevent obesity and associated metabolic diseases by, e.g., sustained GLP-1 release.
Collapse
Affiliation(s)
- Remi Kamakura
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Ghulam Shere Raza
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Nalini Sodum
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Vesa‐Pekka Lehto
- Department of Applied PhysicsFaculty of Science and ForestryUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Miia Kovalainen
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Karl‐Heinz Herzig
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
- Department of Pediatric Gastroenterology and Metabolic DiseasesPediatric InstitutePoznan University of Medical SciencesPoznań60–572Poland
| |
Collapse
|
9
|
Khan D, Moffett RC, Flatt PR, Tarasov AI. Classical and non-classical islet peptides in the control of β-cell function. Peptides 2022; 150:170715. [PMID: 34958851 DOI: 10.1016/j.peptides.2021.170715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/25/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022]
Abstract
The dual role of the pancreas as both an endocrine and exocrine gland is vital for food digestion and control of nutrient metabolism. The exocrine pancreas secretes enzymes into the small intestine aiding digestion of sugars and fats, whereas the endocrine pancreas secretes a cocktail of hormones into the blood, which is responsible for blood glucose control and regulation of carbohydrate, protein and fat metabolism. Classical islet hormones, insulin, glucagon, pancreatic polypeptide and somatostatin, interact in an autocrine and paracrine manner, to fine-tube the islet function and insulin secretion to the needs of the body. Recently pancreatic islets have been reported to express a number of non-classical peptide hormones involved in metabolic signalling, whose major production site was believed to reside outside pancreas, e.g. in the small intestine. We highlight the key non-classical islet peptides, and consider their involvement, together with established islet hormones, in regulation of stimulus-secretion coupling as well as proliferation, survival and transdifferentiation of β-cells. We furthermore focus on the paracrine interaction between classical and non-classical islet hormones in the maintenance of β-cell function. Understanding the functional relationships between these islet peptides might help to develop novel, more efficient treatments for diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| | - R Charlotte Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Andrei I Tarasov
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
10
|
Vasto S, Di Gaudio F, Raso M, Sabatino L, Caldarella R, De Pasquale C, Di Rosa L, Baldassano S. Impact on Glucose Homeostasis: Is Food Biofortified with Molybdenum a Workable Solution? A Two-Arm Study. Nutrients 2022; 14:1351. [PMID: 35405964 PMCID: PMC9002377 DOI: 10.3390/nu14071351] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
Diabetes is expected to increase up to 700 million people worldwide with type 2 diabetes being the most frequent. The use of nutritional interventions is one of the most natural approaches for managing the disease. Minerals are of paramount importance in order to preserve and obtain good health and among them molybdenum is an essential component. There are no studies about the consumption of biofortified food with molybdenum on glucose homeostasis but recent studies in humans suggest that molybdenum could exert hypoglycemic effects. The present study aims to assess if consumption of lettuce biofortified with molybdenum influences glucose homeostasis and whether the effects would be due to changes in gastrointestinal hormone levels and specifically Peptide YY (PYY), Glucagon-Like Peptide 1 (GLP-1), Glucagon-Like Peptide 2 (GLP-2), and Gastric Inhibitory Polypeptide (GIP). A cohort of 24 people was supplemented with biofortified lettuce for 12 days. Blood and urine samples were obtained at baseline (T0) and after 12 days (T2) of supplementation. Blood was analyzed for glucose, insulin, insulin resistance, β-cell function, and insulin sensitivity, PYY, GLP-1, GLP-2 and GIP. Urine samples were tested for molybdenum concentration. The results showed that consumption of lettuce biofortified with molybdenum for 12 days did not affect beta cell function but significantly reduced fasting glucose, insulin, insulin resistance and increased insulin sensitivity in healthy people. Consumption of biofortified lettuce did not show any modification in urine concentration of molybdenum among the groups. These data suggest that consumption of lettuce biofortified with molybdenum improves glucose homeostasis and PYY and GIP are involved in the action mechanism.
Collapse
Affiliation(s)
- Sonya Vasto
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy;
- Euro-Mediterranean Institutes of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Francesca Di Gaudio
- Department of Promoting Health, Maternal-Infant, Excellence and Internal and Specialized Medicine (ProMISE) G. D’Alessandro, University of Palermo, 90127 Palermo, Italy;
| | - Maria Raso
- Chromatography and Mass Spectrometry Section, Quality Control and Chemical Risk (CQRC), Department PROMISE, University Palermo, 90133 Palermo, Italy;
| | - Leo Sabatino
- Dipartimento Scienze Agrarie, Alimentari e Forestali (SAAF), University of Palermo, Viale delle Scienze, Ed. 5, 90128 Palermo, Italy; (L.S.); (C.D.P.)
| | - Rosalia Caldarella
- Department of Laboratory Medicine, “P. Giaccone” University Hospital, 90128 Palermo, Italy;
| | - Claudio De Pasquale
- Dipartimento Scienze Agrarie, Alimentari e Forestali (SAAF), University of Palermo, Viale delle Scienze, Ed. 5, 90128 Palermo, Italy; (L.S.); (C.D.P.)
| | - Luigi Di Rosa
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy;
| | - Sara Baldassano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy;
| |
Collapse
|
11
|
Jurkowska K, Szymańska B, Knysz B, Piwowar A. Effect of Combined Antiretroviral Therapy on the Levels of Selected Parameters Reflecting Metabolic and Inflammatory Disturbances in HIV-Infected Patients. J Clin Med 2022; 11:jcm11061713. [PMID: 35330038 PMCID: PMC8954290 DOI: 10.3390/jcm11061713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 01/25/2023] Open
Abstract
Subjects infected with human immunodeficiency virus (HIV) treated with combined antiretroviral therapy (cART) show a greater predisposition to metabolic disturbances compared to the general population. The aim of the study was to assess the effect of cART on the level of selected parameters related to carbohydrate and lipid metabolism, cardiovascular diseases and inflammation in the plasma of HIV-infected patients against the uninfected. The levels of irisin (IRS), myostatin (MSTN), peptide YY (PYY), glucagon-like peptide-1 (GLP-1), dipeptidyl peptidase IV (DPP-4), fetuin A (FETU-A), pentraxin 3 (PTX 3), chemokine stromal cell-derived factor 1 (SDF-1), and regulated on activation normal T cell expressed and secreted (RANTES) in the plasma of HIV-infected patients and the control group were measured by immunoassay methods. HIV-infected patients were analyzed in terms of CD4+ T cells and CD8+ T cell count, HIV RNA viral load, and the type of therapeutic regimen containing either protease inhibitors (PIs) or integrase transfer inhibitors (INSTIs). The analysis of HIV-infected patients before and after cART against the control group showed statistically significant differences for the following parameters: IRS (p = 0.02), MSTN (p = 0.03), PYY (p = 0.03), GLP-1 (p = 0.03), PTX3 (p = 0.03), and RANTES (p = 0.02), but no significant differences were found for DPP-4, FETU-A, and SDF-1. Comparing the two applied therapeutic regimens, higher levels of all tested parameters were shown in HIV-infected patients treated with INSTIs compared to HIV-infected patients treated with PIs, but the differences were not statistically significant. The obtained results indicated significant changes in the expression of selected parameters in the course of HIV infection and cART. There is need for further research on the clinical usefulness of the selected parameters and for new information on the pathogenesis of HIV-related comorbidities to be provided. The obtained data may allow for better monitoring of the course of HIV infection and optimization of therapy in order to prevent the development of comorbidities as a result of long-term use of cART.
Collapse
Affiliation(s)
- Karolina Jurkowska
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.J.); (A.P.)
| | - Beata Szymańska
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.J.); (A.P.)
- Correspondence: ; Tel.: +48-71-784-0457
| | - Brygida Knysz
- Department of Infectious Diseases, Liver Diseases and Acquired Immune Deficiencies, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.J.); (A.P.)
| |
Collapse
|
12
|
Reverter-Branchat G, Eugster PJ, Kuenzli C, Rindlisbacher B, Stauffer T, Nakas CT, Herzig D, Grouzmann E, Bally L. Multiplexed Assay to Quantify the PP-Fold Family of Peptides in Human Plasma Using Microflow Liquid Chromatography-Tandem Mass Spectrometry. Clin Chem 2022; 68:584-594. [PMID: 35015868 DOI: 10.1093/clinchem/hvab229] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/29/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Peptide Tyr-Tyr (PYY1-36), pancreatic polypeptide (PP1-36) and neuropeptide Y (NPY1-36) constitute the PP-fold family of peptides that is involved in metabolic regulation. Very low plasma concentrations and cleavage into active 3-36 fragments challenge bioanalytical assays used for the quantification of these peptides. METHODS We developed a multiplexed isotopic dilution assay to quantify PYY1-36, PP1-36, and NPY1-36 and their dipeptidyl peptidase-4 (DPP4)-derived metabolites PYY3-36, PP3-36 and NPY3-36. All peptides were immunocaptured from plasma using a monoclonal antibody and quantified by micro-ultra-HPLC-MS/MS. Blood samples from healthy volunteers were collected fasting and 30 min after nutrient stimulation. Method comparison was performed with commercial immunoassays. RESULTS Linearity was shown in the measured intervals (r2 > 0.99). The lower limit of quantification (LLOQ) with a CV at 20% was 1.5 pM for PYY1-36 and PYY3-36, 3.0 pM for PP1-36 and PP3-36, 0.8 pM for NPY1-36 and 0.5 pM for NPY3-36. In all cases, intra- and inter-assay bias and imprecision were <21%. Pre-analytical stability required addition of a protease inhibitor cocktail. Physiological concentrations of PYY3-36, NPY3-36, PP1-36 and PP3-36 were above the LLOQ in 43% to 100% of the samples. PYY1-36 and NPY1-36 were above the LLOQ in 9% and 0% of the samples, respectively. Immunoassays showed higher concentrations of measurands and poor agreement when compared with micro-UHPLC-MS/MS. CONCLUSIONS The assay allowed for specific multiplexed analysis of the PP-fold family of peptides and their DPP4-cleaved fragments in a single sample, thereby offering new perspectives to study the role and therapeutic potential of these essential peptide hormones in health and metabolic disease.
Collapse
Affiliation(s)
- Gemma Reverter-Branchat
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Philippe J Eugster
- Laboratoire des Catécholamines et Peptides, Service de Pharmacologie Clinique, University Hospital of Lausanne, Lausanne, Switzerland
| | | | | | | | - Christos T Nakas
- Laboratory of Biometry, School of Agriculture, University of Thessaly, Nea Ionia Magnesia, Greece.,University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David Herzig
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Eric Grouzmann
- Laboratoire des Catécholamines et Peptides, Service de Pharmacologie Clinique, University Hospital of Lausanne, Lausanne, Switzerland
| | - Lia Bally
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Choong YS, Lim YY, Soong JX, Savoo N, Guida C, Rhyman L, Ramracheya R, Ramasami P. Theoretical study of the interactions between peptide tyrosine tyrosine [PYY (1-36)], a newly identified modulator in type 2 diabetes pathophysiology, with receptors NPY1R and NPY4R. Hormones (Athens) 2021; 20:557-569. [PMID: 33782920 DOI: 10.1007/s42000-021-00278-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/10/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE Diabetes mellitus is a common condition in the clinically obese. Bariatric surgery is one of the ways to put type 2 diabetes in remission. Recent findings propose the appetite-regulator peptide tyrosine tyrosine (PYY) as a therapeutic option for patients with type 2 diabetes. This novel gut hormone restores impaired insulin and glucagon secretion in pancreatic islets and is implicated in type 2 diabetes reversal after bariatric surgery. The current study elucidates the interactions between PYY and the NPY1R and NPY4R receptors using computational methods. METHODS Protein structure prediction, molecular docking simulation, and molecular dynamics (MD) simulation were performed to elucidate the interactions of PYY with NPY1R and NPY4R. RESULTS The predicted binding models of PYY-NPY receptors are in agreement with those described in the literature, although different interaction partners are presented for the C-terminal tail of PYY. Non-polar interactions are predicted to drive the formation of the protein complex. The calculated binding energies show that PYY has higher affinity for NPY4R (ΔGGBSA = -65.08 and ΔGPBSA = -87.62 kcal/mol) than for NPY1R (ΔGGBSA = -23.11 and ΔGPBSA = -50.56 kcal/mol). CONCLUSIONS Based on the constructed models, the binding conformations obtained from docking and MD simulation for both the PYY-NPY1R and PYY-NPY4R complexes provide a detailed map of possible interactions. The calculated binding energies show a higher affinity of PYY for NPY4R. These findings may help to understand the mechanisms behind the improvement of diabetes following bariatric surgery.
Collapse
Affiliation(s)
- Yee Siew Choong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia.
| | - Yee Ying Lim
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Jia Xin Soong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Nandini Savoo
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius
| | - Claudia Guida
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom
| | - Lydia Rhyman
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius
- Department of Chemical Sciences, University of Johannesburg, Doornfontein, Johannesburg 2028, South Africa
| | - Reshma Ramracheya
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom.
- Pharmaceutical Operations, Centre International de Dévelopment Pharmaceutique, BioPark Mauritius, Socota Phoenicia, Phoenix 73408, Mauritius.
| | - Ponnadurai Ramasami
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius, Réduit 80837, Mauritius.
- Department of Chemical Sciences, University of Johannesburg, Doornfontein, Johannesburg 2028, South Africa.
| |
Collapse
|
14
|
Lampropoulos C, Alexandrides T, Tsochatzis S, Kehagias D, Kehagias I. Are the Changes in Gastrointestinal Hormone Secretion Necessary for the Success of Bariatric Surgery? A Critical Review of the Literature. Obes Surg 2021; 31:4575-4584. [PMID: 34304379 DOI: 10.1007/s11695-021-05568-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/21/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Ghrelin, glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) are involved in energy balance regulation and glucose homeostasis. Obesity is characterized by lower fasting levels and blunted postprandial responses of ghrelin, GLP-1, and possibly PYY. Both Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) have been shown to increase postprandial GLP-1 and PYY levels. Human studies have shown that enhanced postprandial GLP-1 and PYY release are associated with favorable weight loss outcomes after RYGB. However, studies in knockout mice have shown that GI hormones are not required for the primary metabolic effects of bariatric surgery. Here, we summarize the complex interaction between obesity, bariatric surgery, and GI hormones in order to determine the exact role of GI hormones in the success of bariatric surgery.
Collapse
Affiliation(s)
| | - Theodoros Alexandrides
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 26504, Rio, Greece
| | - Stylianos Tsochatzis
- Department of General Surgery, Saint Andrew's General Hospital, 26335, Patras, Greece
| | - Dimitrios Kehagias
- Department of General Surgery, General University Hospital of Patras, 26504, Rio, Greece
| | - Ioannis Kehagias
- Division of Bariatric and Metabolic Surgery, Department of Surgery, General University Hospital of Patras, 26504, Rio, Greece
| |
Collapse
|
15
|
Lafferty RA, Flatt PR, Irwin N. Established and emerging roles peptide YY (PYY) and exploitation in obesity-diabetes. Curr Opin Endocrinol Diabetes Obes 2021; 28:253-261. [PMID: 33395088 DOI: 10.1097/med.0000000000000612] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The antiobesity effects of activation of hypothalamic neuropeptide Y2 receptors (NPYR2) by the gut-derived hormone, peptide YY (PYY), are established. However, more recent insight into the biology of PYY has demonstrated remarkable benefits of sustained activation of pancreatic beta-cell NPYR1, that promises to open a new therapeutic avenue in diabetes. RECENT FINDINGS The therapeutic applicability of NPYR2 agonists for obesity has been considered for many years. An alternative pathway for the clinical realisation of PYY-based drugs could be related to the development of NPYR1 agonists for treatment of diabetes. Thus, although stimulation of NPYR1 on pancreatic beta-cells has immediate insulinostatic effects, prolonged activation of these receptors leads to well defined beta-cell protective effects, with obvious positive implications for the treatment of diabetes. In this regard, NPYR1-specific, long-acting enzyme resistant PYY analogues, have been recently developed with encouraging preclinical effects observed on pancreatic islet architecture in diabetes. In agreement, the benefits of certain types of bariatric surgeries on beta-cell function and responsiveness have also been linked to elevated PYY secretion and NPY1 receptor activation. SUMMARY Enzymatically stable forms of PYY, that selectively activate NPYR1, may have significant potential for preservation of beta-cell mass and the treatment of diabetes.
Collapse
Affiliation(s)
- Ryan A Lafferty
- Diabetes Research Group, Ulster University, Coleraine, Northern Ireland, UK
| | | | | |
Collapse
|
16
|
Xie C, Huang W, Young RL, Jones KL, Horowitz M, Rayner CK, Wu T. Role of Bile Acids in the Regulation of Food Intake, and Their Dysregulation in Metabolic Disease. Nutrients 2021; 13:nu13041104. [PMID: 33800566 PMCID: PMC8066182 DOI: 10.3390/nu13041104] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Bile acids are cholesterol-derived metabolites with a well-established role in the digestion and absorption of dietary fat. More recently, the discovery of bile acids as natural ligands for the nuclear farnesoid X receptor (FXR) and membrane Takeda G-protein-coupled receptor 5 (TGR5), and the recognition of the effects of FXR and TGR5 signaling have led to a paradigm shift in knowledge regarding bile acid physiology and metabolic health. Bile acids are now recognized as signaling molecules that orchestrate blood glucose, lipid and energy metabolism. Changes in FXR and/or TGR5 signaling modulates the secretion of gastrointestinal hormones including glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), hepatic gluconeogenesis, glycogen synthesis, energy expenditure, and the composition of the gut microbiome. These effects may contribute to the metabolic benefits of bile acid sequestrants, metformin, and bariatric surgery. This review focuses on the role of bile acids in energy intake and body weight, particularly their effects on gastrointestinal hormone secretion, the changes in obesity and T2D, and their potential relevance to the management of metabolic disorders.
Collapse
Affiliation(s)
- Cong Xie
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
| | - Weikun Huang
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- The ARC Center of Excellence for Nanoscale BioPhotonics, Institute for Photonics and Advanced Sensing, School of Physical Sciences, The University of Adelaide, Adelaide 5005, Australia
| | - Richard L. Young
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute, Adelaide 5005, Australia
| | - Karen L. Jones
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide 5005, Australia
| | - Michael Horowitz
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide 5005, Australia
| | - Christopher K. Rayner
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide 5005, Australia
| | - Tongzhi Wu
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide 5005, Australia
- Institute of Diabetes, School of Medicine, Southeast University, Nanjing 210009, China
- Correspondence:
| |
Collapse
|
17
|
The Intestinal Fatty Acid-Enteroendocrine Interplay, Emerging Roles for Olfactory Signaling and Serotonin Conjugates. Molecules 2021; 26:molecules26051416. [PMID: 33807994 PMCID: PMC7961910 DOI: 10.3390/molecules26051416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Intestinal enteroendocrine cells (EECs) respond to fatty acids from dietary and microbial origin by releasing neurotransmitters and hormones with various paracrine and endocrine functions. Much has become known about the underlying signaling mechanisms, including the involvement of G-protein coupled receptors (GPCRs), like free fatty acids receptors (FFARs). This review focusses on two more recently emerging research lines: the roles of odorant receptors (ORs), and those of fatty acid conjugates in gut. Odorant receptors belong to a large family of GPCRs with functional roles that only lately have shown to reach beyond the nasal-oral cavity. In the intestinal tract, ORs are expressed on serotonin (5-HT) and glucagon-like-peptide-1 (GLP-1) producing enterochromaffin and enteroendocrine L cells, respectively. There, they appear to function as chemosensors of microbiologically produced short-, and branched-chain fatty acids. Another mechanism of fatty acid signaling in the intestine occurs via their conjugates. Among them, conjugates of unsaturated long chain fatty acids and acetate with 5-HT, N-acyl serotonins have recently emerged as mediators with immune-modulatory effects. In this review, novel findings in mechanisms and molecular players involved in intestinal fatty acid biology are highlighted and their potential relevance for EEC-mediated signaling to the pancreas, immune system, and brain is discussed.
Collapse
|
18
|
Lafferty RA, Tanday N, Moffett RC, Reimann F, Gribble FM, Flatt PR, Irwin N. Positive Effects of NPY1 Receptor Activation on Islet Structure Are Driven by Pancreatic Alpha- and Beta-Cell Transdifferentiation in Diabetic Mice. Front Endocrinol (Lausanne) 2021; 12:633625. [PMID: 33716983 PMCID: PMC7949013 DOI: 10.3389/fendo.2021.633625] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/20/2021] [Indexed: 02/03/2023] Open
Abstract
Enzymatically stable and specific neuropeptide Y1 receptor (NPYR1) agonists, such as sea lamprey PYY(1-36) (SL-PYY(1-36)), are believed to improve glucose regulation in diabetes by targeting pancreatic islets. In this study, streptozotocin (STZ) diabetic transgenic GluCreERT2 ;ROSA26-eYFP and Ins1Cre/+;Rosa26-eYFP mouse models have been used to study effects of sustained NPYR1 activation on islet cell composition and alpha- and beta-cell lineage transitioning. STZ induced a particularly severe form of diabetes in Ins1Cre/+;Rosa26-eYFP mice, but twice-daily administration (25 nmol/kg) of SL-PYY(1-36) for 11 days consistently improved metabolic status. Blood glucose was decreased (p < 0.05 - p < 0.001) and both fasted plasma and pancreatic insulin significantly increased by SL-PYY(1-36). In both GluCreERT2 ;ROSA26-eYFP and Ins1Cre/+; Rosa26-eYFP mice, STZ provoked characteristic losses (p < 0.05 - p < 0.001) of islet numbers, beta-cell and pancreatic islet areas together with increases in area and central islet location of alpha-cells. With exception of alpha-cell area, these morphological changes were fully, or partially, returned to non-diabetic control levels by SL-PYY(1-36). Interestingly, STZ apparently triggered decreased (p < 0.001) alpha- to beta-cell transition in GluCreERT2 ;ROSA26-eYFP mice, together with increased loss of beta-cell identity in Ins1Cre/+;Rosa26-eYFP mice, but both effects were significantly (p < 0.001) reversed by SL-PYY(1-36). SL-PYY(1-36) also apparently reduced (p < 0.05) beta- to alpha-cell conversion in Ins1Cre/+;Rosa26-eYFP mice and glucagon expressing alpha-cells in GluCreERT2 ;ROSA26-eYFP mice. These data indicate that islet benefits of prolonged NPY1R activation, and especially restoration of beta-cell mass, are observed irrespective of diabetes status, being linked to cell lineage alterations including transdifferentiation of alpha- to beta-cells.
Collapse
Affiliation(s)
- Ryan A. Lafferty
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Neil Tanday
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - R. Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Fiona M. Gribble
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Peter R. Flatt
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, Ulster University, Coleraine, United Kingdom
| |
Collapse
|
19
|
Lewis PL, Wells JM. Engineering-inspired approaches to study β-cell function and diabetes. Stem Cells 2021; 39:522-535. [PMID: 33497522 DOI: 10.1002/stem.3340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand β-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and β-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human β-cells, to cell lines, to pluripotent stem cell-derived β-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are β-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying β-cell biology, improving β-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic.
Collapse
Affiliation(s)
- Phillip L Lewis
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|