1
|
Zaman V, Matzelle D, Banik NL, Haque A. Dysregulation of Metabolic Peptides Precedes Hyperinsulinemia and Inflammation Following Exposure to Rotenone in Rats. Cells 2025; 14:124. [PMID: 39851552 PMCID: PMC11764466 DOI: 10.3390/cells14020124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Rotenone, a naturally occurring compound derived from the roots of tropical plants, is used as a broad-spectrum insecticide, piscicide, and pesticide. It is a classical, high-affinity mitochondrial complex I inhibitor that causes not only oxidative stress, α-synuclein phosphorylation, DJ-1 (Parkinson's disease protein 7) modifications, and inhibition of the ubiquitin-proteasome system but it is also widely considered an environmental contributor to Parkinson's disease (PD). While prodromal symptoms, such as loss of smell, constipation, sleep disorder, anxiety/depression, and the loss of dopaminergic neurons in the substantia nigra of rotenone-treated animals, have been reported, alterations of metabolic hormones and hyperinsulinemia remain largely unknown and need to be investigated. Whether rotenone and its effect on metabolic peptides could be utilized as a biomarker for its toxic metabolic effects, which can cause long-term detrimental effects and ultimately lead to obesity, hyperinsulinemia, inflammation, and possibly gut-brain axis dysfunction, remains unclear. Here, we show that rotenone disrupts metabolic homeostasis, altering hormonal peptides and promoting infiltration of inflammatory T cells. Specifically, our results indicate a significant decrease in glucagon-like peptide-1 (GLP-1), C-peptide, and amylin. Interestingly, levels of several hormonal peptides related to hyperinsulinemia, such as insulin, leptin, pancreatic peptide (PP), peptide YY (PYY), and gastric inhibitory polypeptide (GIP), were significantly upregulated. Administration of rotenone to rats also increased body weight and activated macrophages and inflammatory T cells. These data strongly suggest that rotenone disrupts metabolic homeostasis, leading to obesity and hyperinsulinemia. The potential implications of these findings are vast, given that monitoring these markers in the blood could not only provide a crucial tool for assessing the extent of exposure and its relevance to obesity and inflammation but could also open new avenues for future research and potential therapeutic strategies.
Collapse
Affiliation(s)
- Vandana Zaman
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA; (V.Z.); (N.L.B.)
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
| | - Naren L. Banik
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA; (V.Z.); (N.L.B.)
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
| | - Azizul Haque
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA; (V.Z.); (N.L.B.)
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA;
- Department of Pharmacology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| |
Collapse
|
2
|
Mohammad Karim A. Principles and Biomedical Applications of Self-Assembled Peptides: Potential Treatment of Type 2 Diabetes Mellitus. Pharmaceutics 2024; 16:1442. [PMID: 39598565 PMCID: PMC11597675 DOI: 10.3390/pharmaceutics16111442] [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: 09/25/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is the most prevalent metabolic disorder worldwide. There have been tremendous efforts to find a safe and prolonged effective therapy for its treatment. Peptide hormones, from certain organisms in the human body, as the pharmaceutical agents, have shown outstanding profiles of efficacy and safety in plasma glucose regulation. Their therapeutic promises have undergone intensive investigations via examining their physicochemical and pharmacokinetic properties. Their major drawback is their short half-life in vivo. To address this challenge, researchers have recently started to apply the state-of-the-art molecular self-assembly on peptide hormones to form nanofibrillar structures, as a smart nanotherapeutic drug delivery technique, to tremendously enhance their prolonged bioactivity in vivo. This revolutionary therapeutic approach would significantly improve patient compliance. First, this review provides a comprehensive summary on the pathophysiology of T2DM, various efforts to treat this chronic disorder, and the limitations and drawbacks of these treatment approaches. Next, this review lays out detailed insights on various aspects of peptide self-assembly: adverse effects, potential applications in nanobiotechnology, thermodynamics and kinetics of the process, as well as the molecular structures of the self-assembled configurations. Furthermore, this review elucidates the recent efforts on applying reversible human-derived peptide self-assembly to generate highly organized smart nanostructured drug formulations known as nanofibrils to regulate and prolong the bioactivity of the human gut hormone peptides in vivo to treat T2DM. Finally, this review highlights the future research directions to advance the knowledge on the state-of-the-art peptide self-assembly process to apply the revolutionary smart nanotherapeutics for treatment of chronic disorders such as T2DM with highly improved patient compliance.
Collapse
Affiliation(s)
- Alireza Mohammad Karim
- Nanoscience Centre, Department of Engineering, University of Cambridge, 11 J. J. Thomson Avenue, Cambridge CB3 0FF, UK
| |
Collapse
|
3
|
Hill A, Tovar Sanchez T, Delaporte E, Sokhela S, Simmons B, Kouanfack C, Mccann K, Levi J, Fairhead C, Venter F. Low CD4 counts predict excessive weight gains during first-line treatment for HIV. J Antimicrob Chemother 2024; 79:2369-2378. [PMID: 39028639 DOI: 10.1093/jac/dkae238] [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: 01/15/2024] [Accepted: 06/22/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Weight gain is common after antiretroviral initiation, especially among females, those of black race and lower baseline CD4, although this may potentially be due to lower baseline weight. Use of tenofovir disoproxil fumarate or efavirenz can suppress weight gain. METHODS Data were pooled from the ADVANCE (n = 1053), NAMSAL (n = 613) and WHRI001 (n = 536) trials investigating first-line regimen. Week 96 weight and body mass index (BMI) was stratified by baseline CD4. Multivariable models of weight change and incident obesity (BMI ≥30 kg/m2) were adjusted for baseline CD4, age, sex, tenofovir disoproxil fumarate, efavirenz, baseline BMI and trial. RESULTS Participants across all treatment arms experienced weight gain from baseline to week 96, with baseline CD4 count, baseline HIV RNA, tenofovir alafenamide and dolutegravir use, and female sex significant predictors. Mean unadjusted weight change was highest with CD4 < 100 (+8.6 kg; SD = 8.2) and lowest with CD4 ≥ 350 (+3.0 kg; SD = 6.5). This weight gain in CD4 < 100 was highest for participants on tenofovir alafenamide-inclusive treatment, such that absolute weight at week 96 was highest in the CD4 < 100 group. Although not statistically significant, obesity rate (BMI ≥ 30 kg/m2) in those taking TAF/FTC + DTG with CD4 < 100 overtook that seen in CD4 ≥ 350, despite lower baseline obesity prevalence. The unadjusted findings were corroborated in multivariable longitudinal models. CONCLUSIONS Participants with low CD4 may demonstrate significant 'overshoot' weight gain, in addition to 'return to health', with a trend towards increased risk of obesity when initiated on TAF/FTC + DTG. Use of tenofovir disoproxil fumarate and efavirenz were associated with smaller weight gains. Effective weight management strategies are needed, especially for individuals with low baseline CD4.
Collapse
Affiliation(s)
- Andrew Hill
- University of Liverpool, Department of Pharmacology and Therapeutics, 70 Pembroke Place, Liverpool L69 3GF, Liverpool, UK
| | - Tamara Tovar Sanchez
- Department of Infectious Diseases, University of Montpellier, TransVIHMI, Institute de Recherche pour le Développement, Inserm Montpellier, France
| | - Eric Delaporte
- Department of Infectious Diseases, University of Montpellier, TransVIHMI, Institute de Recherche pour le Développement, Inserm Montpellier, France
| | - Simiso Sokhela
- Department of HIV, Wits Ezintsha, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Bryony Simmons
- Department of Health Economics, London School of Economics and Political Science, LSE Health, London, UK
| | - Charles Kouanfack
- Faculty of Medicine and Pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | - Kaitlyn Mccann
- Department of Public Health, Imperial College London, Faculty of Medicine, London, UK
| | - Jacob Levi
- Department of Global Health, The Institute of Tropical Medicine and International Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Cassandra Fairhead
- Department of Global Health, The Institute of Tropical Medicine and International Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Francois Venter
- Department of HIV, Wits Ezintsha, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| |
Collapse
|
4
|
Kukucka T, Ferencova N, Visnovcova Z, Ondrejka I, Hrtanek I, Kovacova V, Macejova A, Mlyncekova Z, Tonhajzerova I. Mechanisms Involved in the Link between Depression, Antidepressant Treatment, and Associated Weight Change. Int J Mol Sci 2024; 25:4511. [PMID: 38674096 PMCID: PMC11050075 DOI: 10.3390/ijms25084511] [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: 03/29/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Major depressive disorder is a severe mood disorder associated with a marked decrease in quality of life and social functioning, accompanied by a risk of suicidal behavior. Therefore, seeking out and adhering to effective treatment is of great personal and society-wide importance. Weight changes associated with antidepressant therapy are often cited as the reason for treatment withdrawal and thus are an important topic of interest. There indeed exists a significant mechanistic overlap between depression, antidepressant treatment, and the regulation of appetite and body weight. The suggested pathomechanisms include the abnormal functioning of the homeostatic (mostly humoral) and hedonic (mostly dopaminergic) circuits of appetite regulation, as well as causing neuromorphological and neurophysiological changes underlying the development of depressive disorder. However, this issue is still extensively discussed. This review aims to summarize mechanisms linked to depression and antidepressant therapy in the context of weight change.
Collapse
Affiliation(s)
- Tomas Kukucka
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Nikola Ferencova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (N.F.); (Z.V.)
| | - Zuzana Visnovcova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia; (N.F.); (Z.V.)
| | - Igor Ondrejka
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Igor Hrtanek
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Veronika Kovacova
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Andrea Macejova
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Zuzana Mlyncekova
- Clinic of Psychiatry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, 03659 Martin, Slovakia; (T.K.); (I.O.); (I.H.); (V.K.); (A.M.); (Z.M.)
| | - Ingrid Tonhajzerova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
5
|
Steenackers N, Eksteen G, Wauters L, Augustijns P, Van der Schueren B, Vanuytsel T, Matthys C. Understanding the gastrointestinal tract in obesity: From gut motility patterns to enzyme secretion. Neurogastroenterol Motil 2024; 36:e14758. [PMID: 38342973 DOI: 10.1111/nmo.14758] [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: 03/14/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND AND PURPOSE The pathophysiology of obesity has been the product of extensive research, revealing multiple interconnected mechanisms contributing to body weight regulation. The regulation of energy balance involves an intricate network, including the gut-neuroendocrine interplay. As a consequence, research on the gut-brain-microbiota axis in obesity has grown extensively. The physiology of the gastrointestinal tract, far from being underexplored, has significant implications for the development of specific complications in people living with obesity across the fields of gastroenterology, nutrition, and pharmacology. Clinical research indicates higher fasting bile acids serum levels, and blunted postprandial increases in bilious secretions in people living with obesity. Findings are less straightforward for the impact of obesity on gastric emptying with various studies reporting accelerated, normal, or delayed gastric emptying rates. Conversely, the effect of obesity on gastrointestinal pH, gastrointestinal transit, and gastric and pancreatic enzyme secretion is largely unknown. In this review, we explore the current evidence on the gastrointestinal physiology of obesity.
Collapse
Affiliation(s)
- Nele Steenackers
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Gabriel Eksteen
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Lucas Wauters
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Bart Van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Christophe Matthys
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
6
|
NamKoong C, Kim B, Yu JH, Youn BS, Kim H, Kim E, Gil SY, Kang GM, Lee CH, Kim YB, Park KH, Kim MS, Kwon O. Stomach clusterin as a gut-derived feeding regulator. BMB Rep 2024; 57:149-154. [PMID: 37817436 PMCID: PMC10979347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/16/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023] Open
Abstract
The stomach has emerged as a crucial endocrine organ in the regulation of feeding since the discovery of ghrelin. Gut-derived hormones, such as ghrelin and cholecystokinin, can act through the vagus nerve. We previously reported the satiety effect of hypothalamic clusterin, but the impact of peripheral clusterin remains unknown. In this study, we administered clusterin intraperitoneally to mice and observed its ability to suppress fasting-driven food intake. Interestingly, we found its synergism with cholecystokinin and antagonism with ghrelin. These effects were accompanied by increased c-fos immunoreactivity in nucleus tractus solitarius, area postrema, and hypothalamic paraventricular nucleus. Notably, truncal vagotomy abolished this response. The stomach expressed clusterin at high levels among the organs, and gastric clusterin was detected in specific enteroendocrine cells and the submucosal plexus. Gastric clusterin expression decreased after fasting but recovered after 2 hours of refeeding. Furthermore, we confirmed that stomachspecific overexpression of clusterin reduced food intake after overnight fasting. These results suggest that gastric clusterin may function as a gut-derived peptide involved in the regulation of feeding through the gut-brain axis. [BMB Reports 2024; 57(3): 149-154].
Collapse
Affiliation(s)
- Cherl NamKoong
- Appetite Regulation Laboratory, Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Bohye Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Ji Hee Yu
- Division of Endocrinology and Metabolism, Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Byung Soo Youn
- Osteoneurogen, Inc., Seoul 08501, Korea, Chuncheon 24341, Korea
| | - Hanbin Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Evonne Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - So Young Gil
- Appetite Regulation Laboratory, Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Gil Myoung Kang
- Appetite Regulation Laboratory, Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Chan Hee Lee
- Appetite Regulation Laboratory, Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA, Chuncheon 24341, Korea
| | - Kyeong-Han Park
- Department of Anatomy and Cell Biology, Kangwon National University College of Medicine, Chuncheon 24341, Korea
| | - Min-Seon Kim
- Appetite Regulation Laboratory, Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul 05505, Korea
- Division of Endocrinology and Metabolism, Diabetes Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Obin Kwon
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
7
|
Busebee B, Ghusn W, Cifuentes L, Acosta A. Obesity: A Review of Pathophysiology and Classification. Mayo Clin Proc 2023; 98:1842-1857. [PMID: 37831039 PMCID: PMC10843116 DOI: 10.1016/j.mayocp.2023.05.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 10/14/2023]
Abstract
Obesity is a chronic, multifactorial, and morbid disease. In the United States, 69% of adults are overweight or have obesity, and the global prevalence of obesity is increasing. Obesity is influenced by genetic, neurologic, metabolic, enteric, and behavioral processes. It remains a key modifiable risk factor for many comorbid diseases, including cardiovascular disease, diabetes mellitus, and cancer. Whereas there are recent and significant advances in obesity therapy, including diets, lifestyle modifications, pharmacotherapies, endoscopic procedures, and bariatric surgeries, there is an immense need for a better understanding of the heterogeneity in the pathophysiologic process of obesity and outcomes. Here we review salient pathophysiologic mechanisms underlying the development and morbidity of obesity as well as pathophysiologically based classification systems that inform current obesity management and may inform improved and individualized management in the future.
Collapse
Affiliation(s)
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
8
|
Lou H, Liu X, Liu P. Mechanism and implications of pro-nature physical activity in antagonizing psychological stress: the key role of microbial-gut-brain axis. Front Psychol 2023; 14:1143827. [PMID: 37560094 PMCID: PMC10408457 DOI: 10.3389/fpsyg.2023.1143827] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023] Open
Abstract
Appropriate physical activities and a biodiversity-rich environment are conducive to the relief of psychological stress, and pro-nature physical activities are a combination of the two, which has good application potential in antagonizing psychological stress, but the intervention mechanism is still unclear. The microbiota-gut-brain axis is cyclically associated with psychological stress, and psychological stress can affect the microbiota through the gut-brain pathway, and conversely, the microbiota can also affect the psychological stress-induced symptoms. It is suggested that the microbe-gut-brain axis may provide a new perspective and target for the treatment of psychological stress-related diseases. Pro-nature physical activity can improve the number of Firmicutes, short-chain fatty acids, Akkermansia bacteria, and the gut-brain barrier and further affect the HPA axis, BDNF, and serotonin pathways of gut-brain two-way communication, thereby maintaining the body's homeostasis and reducing antagonistic psychological stress. According to the comprehensive influence of physical activities on the microbiota-gut-brain axis, a "green + exercise prescription hypothesis" in line with the holistic medical concept is revealed, which is expected to be effective in the prevention, alleviation, and treatment of irritable bowel syndrome and neurodegenerative diseases. It provides new means for treating psychological stress-related diseases such as mental disorders and mood disorders. In addition, it enlightens the construction of green infrastructure that is conducive to the diversified contact of microorganisms in outdoor physical activities venues and induces healthy interaction between the human body and the microbial population in the natural ecology. However, the current research is still in its early stages, and the intervention effect and mechanism of pro-nature physical activities need further demonstration in the future.
Collapse
|
9
|
We are what we eat: The role of lipids in metabolic diseases. ADVANCES IN FOOD AND NUTRITION RESEARCH 2023. [PMID: 37516463 DOI: 10.1016/bs.afnr.2022.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lipids play a fundamental role, both structurally and functionally, for the correct functioning of the organism. In the last two decades, they have evolved from molecules involved only in energy storage to compounds that play an important role as components of cell membranes and signaling molecules that regulate cell homeostasis. For this reason, their interest as compounds involved in human health has been gaining weight. Indeed, lipids derived from dietary sources and endogenous biosynthesis are relevant for the pathophysiology of numerous diseases. There exist pathological conditions that are characterized by alterations in lipid metabolism. This is particularly true for metabolic diseases, such as liver steatosis, type 2 diabetes, cancer and cardiovascular diseases. The main issue to be considered is lipid homeostasis. A precise control of fat homeostasis is required for a correct regulation of metabolic pathways and safe and efficient energy storage in adipocytes. When this fails, a deregulation occurs in the maintenance of systemic metabolism. This happens because an increased concentrations of lipids impair cellular homeostasis and disrupt tissue function, giving rise to lipotoxicity. Fat accumulation results in many alterations in the physiology of the affected organs, mainly in metabolic tissues. These alterations include the activation of oxidative and endoplasmic reticulum stress, mitochondrial dysfunction, increased inflammation, accumulation of bioactive molecules and modification of gene expression. In this chapter, we review the main metabolic diseases in which alterations in lipid homeostasis are involved and discuss their pathogenic mechanisms.
Collapse
|
10
|
Factors Affecting the Circulating Levels of Oxyntomodulin in Health and After Acute Pancreatitis. Pancreas 2022; 51:774-783. [PMID: 36395403 DOI: 10.1097/mpa.0000000000002114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVES To investigate the factors associated with the circulating levels of oxyntomodulin in healthy individuals and individuals after an episode of acute pancreatitis (AP). METHODS Blood samples were collected from all participants after an overnight fast and analyzed for 28 biomarkers. Participants also underwent comprehensive body composition analysis on a 3-T magnetic resonance imaging scanner. Regression analyses were done to investigate the associations between oxyntomodulin and the studied factors. RESULTS The study included 105 individuals who had a primary diagnosis of AP and 58 healthy individuals. Peptide YY (B coefficient, 0.094; 95% confidence interval [95% CI], 0.164-0.123), pancreatic polypeptide (0.048; 95% CI, 0.030-0.066), and leptin (0.394; 95% CI, 0.128-0.661) had significant associations with oxyntomodulin in healthy individuals. Peptide YY was the most prominent factor associated with oxyntomodulin, explaining 60% of its variance in health. Cholecystokinin (0.014; 95% CI, 0.010-0.018), amylin (-0.107; 95% CI, -0.192 to -0.021), and glycated hemoglobin (-0.761; 95% CI, -1.249 to -0.273) had significant associations with oxyntomodulin in individuals after AP. Cholecystokinin was the most prominent factor associated with oxyntomodulin, explaining 44% of its variance after AP. CONCLUSIONS Factors affecting the circulating levels of oxyntomodulin are different in health and after AP. These insights will enable the determination of populations that benefit from oxyntomodulin therapeutics in the future.
Collapse
|
11
|
Jahandideh F, Bourque SL, Wu J. A comprehensive review on the glucoregulatory properties of food-derived bioactive peptides. Food Chem X 2022; 13:100222. [PMID: 35498998 PMCID: PMC9039931 DOI: 10.1016/j.fochx.2022.100222] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/03/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus, a group of metabolic disorders characterized by persistent hyperglycemia, affects millions of people worldwide and is on the rise. Dietary proteins, from a wide range of food sources, are rich in bioactive peptides with antidiabetic properties. Notable examples include AGFAGDDAPR, a black tea-derived peptide, VRIRLLQRFNKRS, a β-conglycinin-derived peptide, and milk-derived peptide VPP, which have shown antidiabetic effects in diabetic rodent models through variety of pathways including improving beta-cells function, suppression of alpha-cells proliferation, inhibiting food intake, increasing portal cholecystokinin concentration, enhancing insulin signaling and glucose uptake, and ameliorating adipose tissue inflammation. Despite the immense research on glucoregulatory properties of bioactive peptides, incorporation of these bioactive peptides in functional foods or nutraceuticals is widely limited due to the existence of several challenges in the field of peptide research and commercialization. Ongoing research in this field, however, is fundamental to pave the road for this purpose.
Collapse
Key Words
- AMPK, AMP-activated protein kinase
- Akt, Protein kinase B
- Bioactive peptides
- C/EBP-α, CCAAT/ enhancer binding protein alpha
- CCK, Cholecystokinin
- CCK-1R, CCK type 1 receptor
- DPP-IV, Dipeptidyl peptidase IV
- Diabetes mellitus
- ERK1/2, Extracellular signal regulated kinase 1/2
- GIP, Glucose-dependent insulinotropic polypeptide
- GLP-1, Glucagon-like peptide 1
- GLUT, Glucose transporter
- Glucose homeostasis
- IRS-1, Insulin receptor substrate-1
- Insulin resistance
- MAPK, Mitogen activated protein kinase
- PI3K, Phosphatidylinositol 3-kinase
- PPARγ, Peroxisome proliferator associated receptor gamma
- Reproductive dysfunction
- TZD, Thiazolidinedione
- cGMP, cyclic guanosine-monophosphate
Collapse
Affiliation(s)
- Forough Jahandideh
- Department of Anesthesiology & Pain Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada.,Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Stephane L Bourque
- Department of Anesthesiology & Pain Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2G3, Canada.,Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Jianping Wu
- Cardiovascular Research Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
12
|
Andersen DB, Holst JJ. Peptides in the regulation of glucagon secretion. Peptides 2022; 148:170683. [PMID: 34748791 DOI: 10.1016/j.peptides.2021.170683] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/21/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
Glucose homeostasis is maintained by the glucoregulatory hormones, glucagon, insulin and somatostatin, secreted from the islets of Langerhans. Glucagon is the body's most important anti-hypoglycemic hormone, mobilizing glucose from glycogen stores in the liver in response to fasting, thus maintaining plasma glucose levels within healthy limits. Glucagon secretion is regulated by both circulating nutrients, hormones and neuronal inputs. Hormones that may regulate glucagon secretion include locally produced insulin and somatostatin, but also urocortin-3, amylin and pancreatic polypeptide, and from outside the pancreas glucagon-like peptide-1 and 2, peptide tyrosine tyrosine and oxyntomodulin, glucose-dependent insulinotropic polypeptide, neurotensin and ghrelin, as well as the hypothalamic hormones arginine-vasopressin and oxytocin, and calcitonin from the thyroid. Each of these hormones have distinct effects, ranging from regulating blood glucose, to regulating appetite, stomach emptying rate and intestinal motility, which makes them interesting targets for treating metabolic diseases. Awareness regarding the potential effects of the hormones on glucagon secretion is important since secretory abnormalities could manifest as hyperglycemia or even lethal hypoglycemia. Here, we review the effects of each individual hormone on glucagon secretion, their interplay, and how treatments aimed at modulating the plasma levels of these hormones may also influence glucagon secretion and glycemic control.
Collapse
Affiliation(s)
- Daniel B Andersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, 2200, Copenhagen N, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Blegdamsvej 3B, 2200, Copenhagen N, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
13
|
Spezani R, Mandarim-de-Lacerda CA. The current significance and prospects for the use of dual receptor agonism GLP-1/Glucagon. Life Sci 2022; 288:120188. [PMID: 34861287 DOI: 10.1016/j.lfs.2021.120188] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 12/20/2022]
Abstract
The therapeutic arsenal for treating type 2 diabetes mellitus (T2DM) has been enriched recently with the inclusion of type 1 glucagon-like peptide (GLP-1). GLP-1 receptor agonists (RA) secondarily reduce appetite, decrease gastric emptying, and reduce body weight. This effect has been used to treat overweight/obesity, especially with comorbidities associated with T2DM. However, the first formulations and adverse effects gradually gave way to new formulations with fewer unpleasant effects and a more extended period of action (weekly subcutaneous administration and even oral administration), which improved the acceptance and adherence to the treatment. Therefore, titration of GLP-1RA should be done gradually. Furthermore, when side effects are consistent and intolerable after weeks/months of titration, a lower dose or a combination of antidiabetic therapies should be implemented, avoiding treatment interruption. The effort to produce increasingly powerful molecules with fewer side effects is the driving force behind the pharmaceutical industry. The unimolecular dual agonism GLP-1RA plus glucagon receptor agonism (GRA) represents an updated pharmacological indication for controlling blood glucose levels in treating T2DM and its comorbidities, showing better effects with less adverse impact than mono GLP-1RA. There are currently different proposals in this way by different laboratories. Nevertheless, the experimental results are promising and show that soon, we will have the contribution of new drugs for the treatment of T2DM.
Collapse
Affiliation(s)
- Renata Spezani
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Yu Y, Fernandez ID, Meng Y, Zhao W, Groth SW. Gut hormones, adipokines, and pro- and anti-inflammatory cytokines/markers in loss of control eating: A scoping review. Appetite 2021; 166:105442. [PMID: 34111480 PMCID: PMC10683926 DOI: 10.1016/j.appet.2021.105442] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/31/2022]
Abstract
Loss of control (LOC) eating is the defining feature of binge-eating disorder, and it has particular relevance for bariatric patients. The biomarkers of LOC eating are unclear; however, gut hormones (i.e., ghrelin, cholecystokinin [CCK], peptide YY [PYY], glucagon-like peptide 1 [GLP-1], and pancreatic polypeptide [PP]), adipokines (i.e., leptin, adiponectin), and pro- and anti-inflammatory cytokines/markers (e.g., high-sensitivity C-reactive protein [hsCRP]) are candidates due to their involvement in the psychophysiological mechanisms of LOC eating. This review aimed to synthesize research that has investigated these biomarkers with LOC eating. Because LOC eating is commonly examined within the context of binge-eating disorder, is sometimes used interchangeably with subclinical binge-eating, and is the latent construct underlying disinhibition, uncontrolled eating, and food addiction, these eating behaviors were included in the search. Only studies among individuals with overweight or obesity were included. Among the identified 31 studies, 2 studies directly examined LOC eating and 4 studies were conducted among bariatric patients. Most studies were case-control in design (n = 16) and comprised female-dominant (n = 13) or female-only (n = 13) samples. Studies generally excluded fasting total ghrelin, fasting CCK, fasting PYY, and fasting PP as correlates of the examined eating behaviors. However, there was evidence that the examined eating behaviors were associated with lower levels of fasting acyl ghrelin (the active form of ghrelin) and adiponectin, higher levels of leptin and hsCRP, and altered responses of postprandial ghrelin, CCK, and PYY. The use of GLP-1 analog was able to decrease binge-eating. In conclusion, this review identified potential biomarkers of LOC eating. Future studies would benefit from a direct focus on LOC eating (especially in the bariatric population), using longitudinal designs, exploring potential mediators and moderators, and increased inclusion of the male population.
Collapse
Affiliation(s)
- Yang Yu
- School of Nursing, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| | - I Diana Fernandez
- School of Public Health, University of Rochester, 265 Crittenden Blvd, Rochester, NY, 14642, USA.
| | - Ying Meng
- School of Nursing, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| | - Wenjuan Zhao
- Department of Oncology, Shanghai Medical College, Fudan University, 138 Yixueyuan Rd, Xuhui District, Shanghai, 200032, China.
| | - Susan W Groth
- School of Nursing, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
15
|
Gjermeni E, Kirstein AS, Kolbig F, Kirchhof M, Bundalian L, Katzmann JL, Laufs U, Blüher M, Garten A, Le Duc D. Obesity-An Update on the Basic Pathophysiology and Review of Recent Therapeutic Advances. Biomolecules 2021; 11:1426. [PMID: 34680059 PMCID: PMC8533625 DOI: 10.3390/biom11101426] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity represents a major public health problem with a prevalence increasing at an alarming rate worldwide. Continuous intensive efforts to elucidate the complex pathophysiology and improve clinical management have led to a better understanding of biomolecules like gut hormones, antagonists of orexigenic signals, stimulants of fat utilization, and/or inhibitors of fat absorption. In this article, we will review the pathophysiology and pharmacotherapy of obesity including intersection points to the new generation of antidiabetic drugs. We provide insight into the effectiveness of currently approved anti-obesity drugs and other therapeutic avenues that can be explored.
Collapse
Affiliation(s)
- Erind Gjermeni
- Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, 04289 Leipzig, Germany;
- Department of Cardiology, Median Centre for Rehabilitation Schmannewitz, 04774 Dahlen, Germany;
| | - Anna S. Kirstein
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103 Leipzig, Germany; (A.S.K.); (F.K.); (A.G.)
| | - Florentien Kolbig
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103 Leipzig, Germany; (A.S.K.); (F.K.); (A.G.)
| | - Michael Kirchhof
- Department of Cardiology, Median Centre for Rehabilitation Schmannewitz, 04774 Dahlen, Germany;
| | - Linnaeus Bundalian
- Institute of Human Genetics, University Medical Center Leipzig, 04103 Leipzig, Germany;
| | - Julius L. Katzmann
- Klinik und Poliklinik für Kardiologie, University Clinic Leipzig, 04103 Leipzig, Germany; (J.L.K.); (U.L.)
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, University Clinic Leipzig, 04103 Leipzig, Germany; (J.L.K.); (U.L.)
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Antje Garten
- Pediatric Research Center, University Hospital for Children and Adolescents, Leipzig University, 04103 Leipzig, Germany; (A.S.K.); (F.K.); (A.G.)
| | - Diana Le Duc
- Institute of Human Genetics, University Medical Center Leipzig, 04103 Leipzig, Germany;
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany;
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| |
Collapse
|
16
|
Mueller A, Palilla S, Carter J. Optimal Surgical Treatment for Type 2 Diabetes: Sleeve Gastrectomy or Gastric Bypass? Adv Surg 2021; 55:1-8. [PMID: 34389085 DOI: 10.1016/j.yasu.2021.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Amanda Mueller
- Department of Surgery, University of California - San Francisco, 513 Parnassus Avenue, HSW1601, San Francisco, CA 94143, USA
| | - Sarah Palilla
- Department of Surgery, University of California - San Francisco, 513 Parnassus Avenue, HSW1601, San Francisco, CA 94143, USA
| | - Jonathan Carter
- Department of Surgery, University of California - San Francisco, 513 Parnassus Avenue, HSW1601, San Francisco, CA 94143, USA.
| |
Collapse
|
17
|
Rezaie P, Bitarafan V, Horowitz M, Feinle-Bisset C. Effects of Bitter Substances on GI Function, Energy Intake and Glycaemia-Do Preclinical Findings Translate to Outcomes in Humans? Nutrients 2021; 13:1317. [PMID: 33923589 PMCID: PMC8072924 DOI: 10.3390/nu13041317] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 12/25/2022] Open
Abstract
Bitter substances are contained in many plants, are often toxic and can be present in spoiled food. Thus, the capacity to detect bitter taste has classically been viewed to have evolved primarily to signal the presence of toxins and thereby avoid their consumption. The recognition, based on preclinical studies (i.e., studies in cell cultures or experimental animals), that bitter substances may have potent effects to stimulate the secretion of gastrointestinal (GI) hormones and modulate gut motility, via activation of bitter taste receptors located in the GI tract, reduce food intake and lower postprandial blood glucose, has sparked considerable interest in their potential use in the management or prevention of obesity and/or type 2 diabetes. However, it remains to be established whether findings from preclinical studies can be translated to health outcomes, including weight loss and improved long-term glycaemic control. This review examines information relating to the effects of bitter substances on the secretion of key gut hormones, gastric motility, food intake and blood glucose in preclinical studies, as well as the evidence from clinical studies, as to whether findings from animal studies translate to humans. Finally, the evidence that bitter substances have the capacity to reduce body weight and/or improve glycaemic control in obesity and/or type 2 diabetes, and potentially represent a novel strategy for the management, or prevention, of obesity and type 2 diabetes, is explored.
Collapse
Affiliation(s)
| | | | | | - Christine Feinle-Bisset
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5005, Australia; (P.R.); (V.B.); (M.H.)
| |
Collapse
|
18
|
Simon JJ, Stopyra MA, Mönning E, Sailer S, Lavandier N, Kihm LP, Bendszus M, Preissl H, Herzog W, Friederich HC. Neuroimaging of hypothalamic mechanisms related to glucose metabolism in anorexia nervosa and obesity. J Clin Invest 2021; 130:4094-4103. [PMID: 32315289 DOI: 10.1172/jci136782] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDGiven the heightened tolerance to self-starvation in anorexia nervosa (AN), a hypothalamic dysregulation of energy and glucose homeostasis has been hypothesized. Therefore, we investigated whether hypothalamic reactivity to glucose metabolism is impaired in AN.METHODSTwenty-four participants with AN, 28 normal-weight participants, and 24 healthy participants with obesity underwent 2 MRI sessions in a single-blind, randomized, case-controlled crossover study. We used an intragastric infusion of glucose and water to bypass the cephalic phase of food intake. The responsivity of the hypothalamus and the crosstalk of the hypothalamus with reward-related brain regions were investigated using high-resolution MRI.RESULTSNormal-weight control participants displayed the expected glucose-induced deactivation of hypothalamic activation, whereas patients with AN and participants with obesity showed blunted hypothalamic reactivity. Furthermore, patients with AN displayed blunted reactivity in the nucleus accumbens and amygdala. Compared with the normal-weight participants and control participants with obesity, the patients with AN failed to show functional connectivity between the hypothalamus and the reward-related brain regions during water infusion relative to glucose infusion. Finally, the patients with AN displayed typical baseline levels of peripheral appetite hormones during a negative energy balance.CONCLUSIONThese results indicate that blunted hypothalamic glucose reactivity might be related to the pathophysiology of AN. This study provides insights for future research, as it is an extended perspective of the traditional primary nonhomeostatic understanding of the disease.FUNDINGThis study was supported by a grant from the DFG (SI 2087/2-1).
Collapse
Affiliation(s)
- Joe J Simon
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marion A Stopyra
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Esther Mönning
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Sailer
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Nora Lavandier
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Lars P Kihm
- Endocrinology and Nephrology, Department of Internal Medicine I, and
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Hubert Preissl
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich (IDM) at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Department of Pharmacy and Biochemistry, Interfaculty Centre for Pharmacogenomics and Pharma Research, University of Tübingen, Tübingen, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Centre, Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH), Neuherberg, Germany
| | - Wolfgang Herzog
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Hans-Christoph Friederich
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
19
|
Schaper SJ, Hofmann T, Wölk E, Weibert E, Rose M, Stengel A. Pancreatic Polypeptide but Not Other Members of the Neuropeptide Y Family Shows a Moderate Association With Perceived Anxiety in Obese Men. Front Hum Neurosci 2020; 14:578578. [PMID: 33192409 PMCID: PMC7604387 DOI: 10.3389/fnhum.2020.578578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/22/2020] [Indexed: 01/01/2023] Open
Abstract
Neuropeptide Y (NPY), peptide tyrosine tyrosine (PYY), and pancreatic polypeptide (PP) are important mediators in the bidirectional communication along the gut-brain-axis. Best known for their role in the regulation of appetite and food intake they are considered to play a crucial role in the development of obesity. Additionally, mounting evidence indicates a regulatory function in anxiety, mood and stress resilience with potential sex differences. In the present study, we examined the associations of NPY, PYY, and PP plasma levels with anxiety, depressiveness and perceived stress in obese patients. We analyzed 144 inpatients (90 female, 54 male, BMI mean: 49.4 kg/m2) in a naturalistic treatment setting for obesity and its somatic and mental comorbidities. Fasting blood samples were taken, and patients completed psychometric self-assessment questionnaires (GAD-7, PHQ-9, PSQ-20) within the first week after admission and before discharge. Plasma concentrations of the peptides were measured by ELISA. Women showed significant higher anxiety (GAD-7: 8.13 ± 5.67 vs. 5.93 ± 5.42, p = 0.04) and stress scores (PSQ-20: 52.62 ± 23.5 vs. 41.23 ± 22.53, p = 0.01) than men. In the longitudinal analysis women with a clinically relevant improvement of anxiety (≥ 5 points on GAD-7, p < 0.001) also showed significant improvements in depression (PHQ-9: 38%, p = 0.002) and PSQ-20 scores (23%, p = 0.005) while anxiety-improved male patients only improved in the subscale tension of the PSQ-20 (34%, p = 0.02). In men we observed a positive correlation of PP with anxiety scores (GAD-7: r = 0.41, p = 0.007) and with age (r = 0.49, p = 0.001) on admission while NPY negatively correlated with age (r = -0.38, p = 0.01). In contrast, there were no significant associations (p > 0.05) in female subjects in the cross-sectional as well as in the longitudinal analysis. In conclusion, women suffering from morbid obesity showed greater psychological comorbidity and considerable interactions among them. Despite that we solely observed associations of PP with anxiety and age with NPY and PP in men, suggesting a possible influence of sex hormones on the NPY system. However, improvement of anxiety scores did not lead to significant changes in NPY.
Collapse
Affiliation(s)
- Selina Johanna Schaper
- Department for Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Tobias Hofmann
- Department for Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ellen Wölk
- Department for Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Elena Weibert
- Department for Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Matthias Rose
- Department for Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, United States
| | - Andreas Stengel
- Department for Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
20
|
Yin C, Zhang H, Zhang M, Xiao Y. Adropin and apelin-12 efficiently predict metabolic syndrome in obese children. Pediatr Diabetes 2020; 21:1132-1139. [PMID: 32749012 DOI: 10.1111/pedi.13101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/09/2020] [Accepted: 07/28/2020] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Metabolic syndrome (MetS) is the most common condition associated with childhood and adolescent obesity and is a challenging public health issue. Very few studies have described the specificity and sensitivity of serum levels of adropin and apelin-12 as predictors of MetS. The aim of this study was to evaluate the association between serum levels of adropin and apelin-12 and MetS, and their sensitivity as predictors of MetS in obese children. METHODS This study involved 138 children. The study group included obese subjects with MetS, and the two control groups included obese subjects without MetS and normal weight subjects. Anthropometric parameters and clinical data were collected. Plasma levels of apelin-12, adropin, leptin, adiponectin, and TNF-α were also measured. RESULTS Obese children with MetS had significantly higher levels of apelin-12 and significantly lower levels of adropin when compared with those in children without MetS. In logistic regressions, we identified that apelin-12 was a risk factor for metabolic syndrome, and adropin was a protecting factor of having MetS after adjusting for age, sex, and puberty. Furthermore, ROC analysis revealed that adropin and apelin-12 are more sensitive predictors of metabolic syndrome than leptin and adiponectin. CONCLUSION Serum adropin and apelin-12 levels can be useful biomarkers for predicting MetS in obese children. Our findings could provide a novel approach for the treatment and prevention of MetS.
Collapse
Affiliation(s)
- Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shan Xi, China
| | - Hongai Zhang
- Department of Pediatrics, The First People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Meizheng Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shan Xi, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shan Xi, China
| |
Collapse
|
21
|
Koliaki C, Liatis S, Dalamaga M, Kokkinos A. The Implication of Gut Hormones in the Regulation of Energy Homeostasis and Their Role in the Pathophysiology of Obesity. Curr Obes Rep 2020; 9:255-271. [PMID: 32647952 DOI: 10.1007/s13679-020-00396-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on the role of gut hormones and their interactions in the regulation of energy homeostasis, describes gut hormone adaptations in obesity and in response to weight loss, and summarizes the current evidence on the role of gut hormone-based therapies for obesity treatment. RECENT FINDINGS Gut hormones play a key role in regulating eating behaviour, energy and glucose homeostasis. Dysregulated gut hormone responses have been proposed to be pathogenetically involved in the development and perpetuation of obesity. Summarizing the major gut hormone changes in obesity, obese individuals are characterized by blunted postprandial ghrelin suppression, loss of premeal ghrelin peaks, impaired diurnal ghrelin variability and reduced fasting and postprandial levels of anorexigenic peptides. Adaptive alterations of gut hormone levels are implicated in weight regain, thus complicating hypocaloric dietary interventions, and can further explain the profound weight loss and metabolic improvement following bariatric surgery. A plethora of compounds mimicking gut hormone changes after bariatric surgery are currently under investigation, introducing a new era in the pharmacotherapy of obesity. The current trend is to combine different gut hormone receptor agonists and target multiple systems simultaneously, in order to replicate as closely as possible the gut hormone milieu after bariatric surgery and circumvent the counter-regulatory adaptive changes associated with dietary energy restriction. An increasing number of preclinical and early-phase clinical trials reveal the additive benefits obtained with dual or triple gut peptide receptor agonists in reducing body weight and improving glycaemia. Gut hormones act as potent regulators of energy and glucose homeostasis. Therapeutic strategies targeting their levels or receptors emerge as a promising approach to treat patients with obesity and hyperglycaemia.
Collapse
Affiliation(s)
- Chrysi Koliaki
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece.
| | - Stavros Liatis
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| |
Collapse
|
22
|
Mejido DC, Peny JA, Vieira MN, Ferreira ST, De Felice FG. Insulin and leptin as potential cognitive enhancers in metabolic disorders and Alzheimer's disease. Neuropharmacology 2020; 171:108115. [PMID: 32344008 DOI: 10.1016/j.neuropharm.2020.108115] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 03/30/2020] [Accepted: 04/20/2020] [Indexed: 02/08/2023]
|
23
|
Patterson M, Ward H, Halvai D, Holm Nilsen HA, Reeves S. Postprandial regulation of prouroguanylin in humans of a healthy weight and those who are overweight or with obesity. Peptides 2020; 123:170179. [PMID: 31697966 DOI: 10.1016/j.peptides.2019.170179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/12/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
Abstract
Uroguanylin is a peptide gut hormone proposed to have a role in signalling post meal satiety. Uroguanylin circulates as its pro-hormone, prouroguanylin. There has been limited investigation of the regulation of prouroguanylin by food; therefore we investigated prouroguanylin regulation following meals. In separate experiments we investigated the effects of high calorie (1451 kcal) and medium calorie (725 kcal), high fat meals, on plasma prouroguanylin concentrations. We then examined the effect of a 722.5 kcal high carbohydrate breakfast on prouroguanylin concentrations, comparing the response in healthy weight adults versus those who are overweight/ with obesity. The 1451 kcal meal increased prouroguanylin concentrations, versus fasting at 60 (P < 0.05), 90 (P < 0.01) and 120 (P < 0.001) minutes. After the 725 kcal meal hormone concentrations rose more slowly and were significant versus fasting concentrations at 120 min (P < 0.01). The high carbohydrate breakfast 722.5 kcal, led to an initial suppression of hormone concentrations at 30 min. post meal (P < 0.05) followed by an increase in concentrations until they were significant versus fasting at 120 min. (P < 0.01). Participants overweight/ with obesity had lower fasting prouroguanylin concentrations (P < 0.05), but post meal concentrations did not differ between the groups. Our results suggest there is a delayed increase in prouroguanylin concentrations following, large and regular sized mixed macronutrient meals rich in fat or carbohydrate. Fasting levels are suppressed in people who are overweight/ with obesity, but the post meal response remains intact. There may be potential to target post meal release of prouroguanylin in obesity.
Collapse
Affiliation(s)
| | - Hannah Ward
- Department of Life Sciences, University of Roehampton, London, UK
| | - Delaram Halvai
- Department of Life Sciences, University of Roehampton, London, UK
| | | | - Sue Reeves
- Department of Life Sciences, University of Roehampton, London, UK
| |
Collapse
|
24
|
Nota MH, Vreeken D, Wiesmann M, Aarts EO, Hazebroek EJ, Kiliaan AJ. Obesity affects brain structure and function- rescue by bariatric surgery? Neurosci Biobehav Rev 2020; 108:646-657. [DOI: 10.1016/j.neubiorev.2019.11.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/28/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023]
|
25
|
Ilie IR. Neurotransmitter, neuropeptide and gut peptide profile in PCOS-pathways contributing to the pathophysiology, food intake and psychiatric manifestations of PCOS. Adv Clin Chem 2019; 96:85-135. [PMID: 32362321 DOI: 10.1016/bs.acc.2019.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major health problem with a heterogeneous hormone-imbalance and clinical presentation across the lifespan of women. Increased androgen production and abnormal gonadotropin-releasing hormone (GnRH) release and gonadotropin secretion, resulting in chronic anovulation are well-known features of the PCOS. The brain is both at the top of the neuroendocrine axis regulating ovarian function and a sensitive target of peripheral gonadal hormones and peptides. Current literature illustrates that neurotransmitters regulate various functions of the body, including reproduction, mood and body weight. Neurotransmitter alteration could be one of the reasons for disturbed GnRH release, consequently directing the ovarian dysfunction in PCOS, since there is plenty evidence for altered catecholamine metabolism and brain serotonin or opioid activity described in PCOS. Further, the dysregulated neurotransmitter and neuropeptide profile in PCOS could also be the reason for low self-esteem, anxiety, mood swings and depression or obesity, features closely associated with PCOS women. Can these altered central brain circuits, or the disrupted gut-brain axis be the tie that would both explain and link the pathogenesis of this disorder, the occurrence of depression, anxiety and other mood disorders as well as of obesity, insulin resistance and abnormal appetite in PCOS? This review intends to provide the reader with a comprehensive overview of what is known about the relatively understudied, but very complex role that neurotransmitters, neuropeptides and gut peptides play in PCOS. The answer to the above question may help the development of drugs to specifically target these central and peripheral circuits, thereby providing a valuable treatment for PCOS patients that present to the clinic with GnRH/LH hypersecretion, obesity or psychiatric manifestations.
Collapse
Affiliation(s)
- Ioana R Ilie
- Department of Endocrinology, University of Medicine and Pharmacy 'Iuliu-Hatieganu', Cluj-Napoca, Romania.
| |
Collapse
|
26
|
Payab M, Hasani-Ranjbar S, Shahbal N, Qorbani M, Aletaha A, Haghi-Aminjan H, Soltani A, Khatami F, Nikfar S, Hassani S, Abdollahi M, Larijani B. Effect of the herbal medicines in obesity and metabolic syndrome: A systematic review and meta-analysis of clinical trials. Phytother Res 2019; 34:526-545. [PMID: 31793087 DOI: 10.1002/ptr.6547] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 09/18/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022]
Abstract
Obesity is a medical situation in which excess body fat has gathered because of imbalance between energy intake and energy expenditure. In spite of the fact that the variety of studies are available for obesity treatment and management, its "globesity" still remains a big challenge all over the world. The current systematic review and meta-analysis aimed to evaluate the efficacy, safety, and mechanisms of effective herbal medicines in the management and treatment of obesity and metabolic syndrome in human. We systematically searched all relevant clinical trials via Web of Science, Scopus, PubMed, and the Cochrane database to assess the effects of raw or refined products derived from plants or parts of plants on obesity and metabolic syndrome in overweight and obesity adult subjects. All studies conducted by the end of May 2019 were considered in the systematic review. Data were extracted independently by two experts. The quality assessment was assessed using Consolidated Standards of Reporting Trials checklist. The main outcomes were anthropometric indices and metabolic syndrome components. Pooled effect of herbal medicines on obesity and metabolic syndrome were presented as standardized mean difference (SMD) and 95% confidence interval (CI). A total of 279 relevant clinical trials were included. Herbals containing green tea, Phaseolus vulgaris, Garcinia cambogia, Nigella sativa, puerh tea, Irvingia gabonensis, and Caralluma fimbriata and their active ingredients were found to be effective in the management of obesity and metabolic syndrome. In addition, C. fimbriata, flaxseed, spinach, and fenugreek were able to reduce appetite. Meta-analysis showed that intake of green tea resulted in a significant improvement in weight ([SMD]: -0.75 [-1.18, -0.319]), body mass index ([SMD]: -1.2 [-1.82, -0.57]), waist circumference ([SMD]: -1.71 [-2.66, -0.77]), hip circumference ([SMD]: -0.42 [-1.02, -0.19]), and total cholesterol, ([SMD]: -0.43 [-0.77, -0.09]). In addition, the intake of P. vulgaris and N. sativa resulted in a significant improvement in weight ([SMD]: -0.88, 95 % CI: [-1.13, -0.63]) and triglyceride ([SMD]: -1.67, 95 % CI: [-2.54, -0.79]), respectively. High quality trials are still needed to firmly establish the clinical efficacy of the plants in obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Hasani-Ranjbar
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazila Shahbal
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Azadeh Aletaha
- Evidence Based Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of medical Sciences, Tehran, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Science Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Akbar Soltani
- Evidence Based Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of medical Sciences, Tehran, Iran
| | - Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, and Evidence-based Evaluation of Cost-Effectiveness and Clinical Outcomes Group, Pharmaceutical Science Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Hassani
- Toxicology and Diseases Group (TDG), Pharmaceutical Science Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group (TDG), Pharmaceutical Science Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Miron I, Dumitrascu D. GASTROINTESTINAL MOTILITY DISORDERS IN OBESITY. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2019; 15:497-504. [PMID: 32377248 PMCID: PMC7200119 DOI: 10.4183/aeb.2019.497] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) motility, which is important for the digestion and absorption, may be altered in obesity. The aim of this review is to present the GI motility changes occurring in obesity, as well as their underlying mechanisms. We have conducted a systematic review of the published literature concerning GI motility and obesity and have described recent published data on the changes throughout the entire GI tract. Most recent discoveries include evidence supporting the increase of gastroesophageal reflux disease in obesity and inhibition of gastric motility. Intestinal transit of the distal small bowel generally slows down, ensuring enough time for digestion and absorption. Constipation is more frequent in obese patients than in those with a normal weight. The gut-brain axis plays an important role in the pathophysiology of GI motility disorders in obesity. This bidirectional communication is achieved by way of neurons, hormones, metabolites derived from intestinal microbiota and cytokines. The molecular mechanisms of GI motility changes in obesity are complex. Current data offer a starting point for further research needed to clarify the association of obesity with GI motility disorders.
Collapse
Affiliation(s)
- I. Miron
- “Iuliu Hatieganu” University of Medicine and Pharmacy, 3 Medical Clinic, Cluj-Napoca, Romania
| | - D.L. Dumitrascu
- “Iuliu Hatieganu” Dept of Internal Medicine, Cluj-Napoca, Romania
| |
Collapse
|
28
|
Changes in Plasma Free Fatty Acids Associated with Type-2 Diabetes. Nutrients 2019; 11:nu11092022. [PMID: 31466350 PMCID: PMC6770316 DOI: 10.3390/nu11092022] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with increased total plasma free fatty acid (FFA) concentrations and an elevated risk of cardiovascular disease. The exact mechanisms by which the plasma FFA profile of subjects with T2DM changes is unclear, but it is thought that dietary fats and changes to lipid metabolism are likely to contribute. Therefore, establishing the changes in concentrations of specific FFAs in an individual’s plasma is important. Each type of FFA has different effects on physiological processes, including the regulation of lipolysis and lipogenesis in adipose tissue, inflammation, endocrine signalling and the composition and properties of cellular membranes. Alterations in such processes due to altered plasma FFA concentrations/profiles can potentially result in the development of insulin resistance and coagulatory defects. Finally, fibrates and statins, lipid-regulating drugs prescribed to subjects with T2DM, are also thought to exert part of their beneficial effects by impacting on plasma FFA concentrations. Thus, it is also interesting to consider their effects on the concentration of FFAs in plasma. Collectively, we review how FFAs are altered in T2DM and explore the likely downstream physiological and pathological implications of such changes.
Collapse
|
29
|
Srugo SA, Bloise E, Nguyen TTTN, Connor KL. Impact of Maternal Malnutrition on Gut Barrier Defense: Implications for Pregnancy Health and Fetal Development. Nutrients 2019; 11:nu11061375. [PMID: 31248104 PMCID: PMC6628366 DOI: 10.3390/nu11061375] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022] Open
Abstract
Small intestinal Paneth cells, enteric glial cells (EGC), and goblet cells maintain gut mucosal integrity, homeostasis, and influence host physiology locally and through the gut-brain axis. Little is known about their roles during pregnancy, or how maternal malnutrition impacts these cells and their development. Pregnant mice were fed a control diet (CON), undernourished by 30% vs. control (UN), or fed a high fat diet (HF). At day 18.5 (term = 19), gut integrity and function were assessed by immunohistochemistry and qPCR. UN mothers displayed reduced mRNA expression of Paneth cell antimicrobial peptides (AMP; Lyz2, Reg3g) and an accumulation of villi goblet cells, while HF had reduced Reg3g and mucin (Muc2) mRNA and increased lysozyme protein. UN fetuses had increased mRNA expression of gut transcription factor Sox9, associated with reduced expression of maturation markers (Cdx2, Muc2), and increased expression of tight junctions (TJ; Cldn-7). HF fetuses had increased mRNA expression of EGC markers (S100b, Bfabp, Plp1), AMP (Lyz1, Defa1, Reg3g), and TJ (Cldn-3, Cldn-7), and reduced expression of an AMP-activator (Tlr4). Maternal malnutrition altered expression of genes that maintain maternal gut homeostasis, and altered fetal gut permeability, function, and development. This may have long-term implications for host-microbe interactions, immunity, and offspring gut-brain axis function.
Collapse
Affiliation(s)
- Sebastian A Srugo
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Enrrico Bloise
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil.
| | | | - Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada.
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
30
|
|
31
|
Weltens N, Iven J, Van Oudenhove L, Kano M. The gut-brain axis in health neuroscience: implications for functional gastrointestinal disorders and appetite regulation. Ann N Y Acad Sci 2019; 1428:129-150. [PMID: 30255954 DOI: 10.1111/nyas.13969] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/07/2018] [Accepted: 08/23/2018] [Indexed: 12/20/2022]
Abstract
Over the past few years, scientific interest in the gut-brain axis (i.e., the bidirectional communication system between the gastrointestinal tract and the brain) has exploded, mostly due to the identification of the gut microbiota as a novel key player in this communication. However, important progress has also been made in other aspects of gut-brain axis research, which has been relatively underemphasized in the review literature. Therefore, in this review, we provide a comprehensive, although not exhaustive, overview of recent research on the functional neuroanatomy of the gut-brain axis and its relevance toward the multidisciplinary field of health neuroscience, excluding studies on the role of the gut microbiota. More specifically, we first focus on irritable bowel syndrome, after which we outline recent findings on the role of the gut-brain axis in appetite and feeding regulation, primarily focusing on the impact of subliminal nutrient-related gut-brain signals. We conclude by providing future perspectives to facilitate translation of the findings from gut-brain axis neuroscientific research to clinical applications in these domains.
Collapse
Affiliation(s)
- Nathalie Weltens
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), University of Leuven, Leuven, Belgium.,Leuven Brain Institute, University of Leuven, Leuven, Belgium
| | - Julie Iven
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), University of Leuven, Leuven, Belgium.,Leuven Brain Institute, University of Leuven, Leuven, Belgium
| | - Lukas Van Oudenhove
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), University of Leuven, Leuven, Belgium.,Leuven Brain Institute, University of Leuven, Leuven, Belgium.,Consultation-Liaison Psychiatry, University Psychiatric Centre KU Leuven, Campus Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Michiko Kano
- Frontiers Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Sendai, Japan.,Department of Behavioral Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
32
|
Scopelliti A, Bauer C, Yu Y, Zhang T, Kruspig B, Murphy DJ, Vidal M, Maddocks ODK, Cordero JB. A Neuronal Relay Mediates a Nutrient Responsive Gut/Fat Body Axis Regulating Energy Homeostasis in Adult Drosophila. Cell Metab 2019; 29:269-284.e10. [PMID: 30344016 PMCID: PMC6370946 DOI: 10.1016/j.cmet.2018.09.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 08/10/2018] [Accepted: 09/25/2018] [Indexed: 02/05/2023]
Abstract
The control of systemic metabolic homeostasis involves complex inter-tissue programs that coordinate energy production, storage, and consumption, to maintain organismal fitness upon environmental challenges. The mechanisms driving such programs are largely unknown. Here, we show that enteroendocrine cells in the adult Drosophila intestine respond to nutrients by secreting the hormone Bursicon α, which signals via its neuronal receptor DLgr2. Bursicon α/DLgr2 regulate energy metabolism through a neuronal relay leading to the restriction of glucagon-like, adipokinetic hormone (AKH) production by the corpora cardiaca and subsequent modulation of AKH receptor signaling within the adipose tissue. Impaired Bursicon α/DLgr2 signaling leads to exacerbated glucose oxidation and depletion of energy stores with consequent reduced organismal resistance to nutrient restrictive conditions. Altogether, our work reveals an intestinal/neuronal/adipose tissue inter-organ communication network that is essential to restrict the use of energy and that may provide insights into the physiopathology of endocrine-regulated metabolic homeostasis.
Collapse
Affiliation(s)
| | - Christin Bauer
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Yachuan Yu
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Tong Zhang
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Björn Kruspig
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Daniel J Murphy
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Marcos Vidal
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Oliver D K Maddocks
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
| |
Collapse
|
33
|
Choi HN, Yim JE. Plasma Adropin as a Potential Marker Predicting Obesity and Obesity-associated Cancer in Korean Patients With Type 2 Diabetes Mellitus. J Cancer Prev 2018; 23:191-196. [PMID: 30671402 PMCID: PMC6330988 DOI: 10.15430/jcp.2018.23.4.191] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 01/20/2023] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) and cancer are serious health problems worldwide, and their prevalences have been on the rise in recent years. It has been reported that adropin plays an important role in the development of T2DM, oxidative stress, inflammation, and obesity. However, there is limited information available on T2DM from human studies, especially for the Korean population. In this study, we aimed to investigate the correlation between adropin levels and obesity of Korean T2DM patients. Methods Thirty-six T2DM patients were recruited for this study. The participants were further classified into female (n = 12) and male (n = 24). Their body composition, metabolic parameters, inflammatory factors, and oxidative stress were measured. Results The severity of obesity is more manifested in male than in female. Plasma triglyceride (TG) and high-sensitivity C-reactive protein (hs-CRP) levels of male were significantly higher than female. The plasma adropin and adiponectin level of female was significantly higher than male. The body weight, body mass index (BMI), body fat mass were negatively correlated with the plasma adropin level in female, whereas adropin has positive correlation with adiponectin in female. The hs-CRP was negatively correlated with the plasma adropin level in female and male. malondialdehyde, reactive oxidative species, and TNF-α was not significantly correlated with adropin in patients with T2DM. Conclusions These findings suggest that adropin may be more used as a biomarker for predicting the risk of obesity and inflammation in Korean patients with T2DM, especially women.
Collapse
Affiliation(s)
- Ha-Neul Choi
- Department of Food and Nutrition, College of Natural Science, Changwon National University, Changwon, Korea
| | - Jung-Eun Yim
- Department of Food and Nutrition, College of Natural Science, Changwon National University, Changwon, Korea
| |
Collapse
|
34
|
Diabesity and mood disorders: Multiple links through the microbiota-gut-brain axis. Mol Aspects Med 2018; 66:80-93. [PMID: 30513310 DOI: 10.1016/j.mam.2018.11.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/30/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023]
Abstract
The global prevalence of diabesity is on the rise, and the clinical, social and economic health burden arising from this epidemic is aggravated by a significant co-morbidity of diabesity with neuropsychiatric disease, particularly depression. Importantly, not only is the prevalence of mood disorders elevated in patients with type 2 diabetes, depressed patients are also more prone to develop diabetes. This reciprocal relationship calls for a molecular and systemic analysis of diabesity-brain interactions to guide preventive and therapeutic strategies. The analysis we are presenting in this review is modelled on the microbiota-gut-brain axis, which provides the brain with information from the gut not only via the nervous system, but also via a continuous stream of microbial, endocrine, metabolic and immune messages. This communication network offers important clues as to how obesity and diabetes could target the brain to provoke neuropsychiatric disease. There is emerging evidence that the gut microbiota is orchestrating a multiplicity of bodily functions that are intimately related to the immune, metabolic and nervous systems and that gut dysbiosis spoils the homeostasis between these systems. In our article we highlight two groups of molecular links that seem to have a significant bearing on the impact of diabesity on the brain. On the one hand, we focus on microbiota-related metabolites such as short-chain fatty acids, tryptophan metabolites, immune stimulants and endocannabinoids that are likely to play a mediator role. On the other hand, we discuss signalling molecules that operate primarily in the brain, specifically neuropeptide Y, brain-derived neurotrophic factor and γ-amino butyric acid, that are disturbed by microbial factors, obesity and diabetes and are relevant to mental illness. Finally, we address the usefulness of diet-related interventions to suspend the deleterious relationship between diabesity and mood disorders.
Collapse
|
35
|
Maciel MG, Beserra BTS, Oliveira FCB, Ribeiro CM, Coelho MS, Neves FDAR, Amato AA. The effect of glucagon-like peptide 1 and glucagon-like peptide 1 receptor agonists on energy expenditure: A systematic review and meta-analysis. Diabetes Res Clin Pract 2018; 142:222-235. [PMID: 29857094 DOI: 10.1016/j.diabres.2018.05.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 12/27/2022]
Abstract
AIM We reviewed clinical trials addressing the effect of glucacon-like peptide 1 (GLP-1) or GLP-1 receptor agonists (GLP-1RA) on energy expenditure (EE) in adults. MATERIALS AND METHODS PubMed, Science Direct and Web of Science were searched for clinical trials investigating the effect of GLP-1 or GLP-1RA on EE in adults. RESULTS Ten trials (93 participants) assessed the effect of GLP-1 administration over 1 to 48 h and found no change in resting EE (REE). Two out of three trials (62 participants) reported a significant decrease in diet-induced thermogenesis (DIT) following GLP-1 administration. Ten trials with exenatide (10 μg bid, for 10-52 weeks) or liraglutide (0.6, 1.2, 1.8 or 3 mg, for 3 days-52 weeks), with a total of 282 participants, indicated a neutral effect of these GLP-1RA on REE, DIT or physical activity-induced EE. Importantly, the longest trial with GLP-1RA reported a significant increase in REE in response to treatment with both exenatide or liraglutide and most trials reported that GLP-1RA-induced weight loss was not accompanied by decreased REE. CONCLUSIONS This review indicates that GLP-1 has no short-term effect on REE but may decrease DIT. The GLP-1RA exenatide and liraglutide have a neutral effect on REE, although it is not possible to rule out an increase in REE following prolonged treatment.
Collapse
Affiliation(s)
- Michel Garcia Maciel
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil; Campus Universitario Darcy Ribeiro, Asa Norte, Brasilia, DF 70910-900, Brazil
| | - Bruna Teles Soares Beserra
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil; Campus Universitario Darcy Ribeiro, Asa Norte, Brasilia, DF 70910-900, Brazil
| | - Fernanda Cerqueira Barroso Oliveira
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil; Campus Universitario Darcy Ribeiro, Asa Norte, Brasilia, DF 70910-900, Brazil
| | - Carolina Martins Ribeiro
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil; Campus Universitario Darcy Ribeiro, Asa Norte, Brasilia, DF 70910-900, Brazil
| | - Michella Soares Coelho
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil; Campus Universitario Darcy Ribeiro, Asa Norte, Brasilia, DF 70910-900, Brazil
| | - Francisco de Assis Rocha Neves
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil; Campus Universitario Darcy Ribeiro, Asa Norte, Brasilia, DF 70910-900, Brazil
| | - Angélica Amorim Amato
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil; Campus Universitario Darcy Ribeiro, Asa Norte, Brasilia, DF 70910-900, Brazil.
| |
Collapse
|
36
|
Bower RL, Yule L, Rees TA, Deganutti G, Hendrikse ER, Harris PWR, Kowalczyk R, Ridgway Z, Wong AG, Swierkula K, Raleigh DP, Pioszak AA, Brimble MA, Reynolds CA, Walker CS, Hay DL. Molecular Signature for Receptor Engagement in the Metabolic Peptide Hormone Amylin. ACS Pharmacol Transl Sci 2018; 1:32-49. [PMID: 32219203 DOI: 10.1021/acsptsci.8b00002] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Indexed: 11/30/2022]
Abstract
The pancreatic peptide hormone, amylin, plays a critical role in the control of appetite, and synergizes with other key metabolic hormones such as glucagon-like peptide 1 (GLP-1). There is opportunity to develop potent and long-acting analogues of amylin or hybrids between these and GLP-1 mimetics for treating obesity. To achieve this, interrogation of how the 37 amino acid amylin peptide engages with its complex receptor system is required. We synthesized an extensive library of peptides to profile the human amylin sequence, determining the role of its disulfide loop, amidated C-terminus and receptor "capture" and "activation" regions in receptor signaling. We profiled four signaling pathways with different ligands at multiple receptor subtypes, in addition to exploring selectivity determinants between related receptors. Distinct roles for peptide subregions in receptor binding and activation were identified, resulting in peptides with greater activity than the native sequence. Enhanced peptide activity was preserved in the brainstem, the major biological target for amylin. Interpretation of our data using full-length active receptor models supported by molecular dynamics, metadynamics, and supervised molecular dynamics simulations guided the synthesis of a potent dual agonist of GLP-1 and amylin receptors. The data offer new insights into the function of peptide amidation, how allostery drives peptide-receptor interactions, and provide a valuable resource for the development of novel amylin agonists for treating diabetes and obesity.
Collapse
Affiliation(s)
- Rebekah L Bower
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Lauren Yule
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand.,School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand.,School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Tayla A Rees
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Giuseppe Deganutti
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, U.K
| | - Erica R Hendrikse
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Paul W R Harris
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand.,School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand.,School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Renata Kowalczyk
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand.,School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Zachary Ridgway
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Amy G Wong
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States
| | - Katarzyna Swierkula
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, U.K
| | - Daniel P Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794-3400, United States.,Department of Structural and Molecular Biology, University College London, London WC1E 6BT, U.K
| | - Augen A Pioszak
- Department of Biochemistry and Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Margaret A Brimble
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand.,School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Christopher A Reynolds
- School of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, U.K
| | - Christopher S Walker
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| | - Debbie L Hay
- School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand.,School of Biological Sciences, School of Chemical Sciences, and Maurice Wilkins Centre, The University of Auckland, Auckland, 1010, New Zealand
| |
Collapse
|
37
|
Abstract
Islets of Langerhans are islands of endocrine cells scattered throughout the pancreas. A number of new studies have pointed to the potential for conversion of non-β islet cells in to insulin-producing β-cells to replenish β-cell mass as a means to treat diabetes. Understanding normal islet cell mass and function is important to help advance such treatment modalities: what should be the target islet/β-cell mass, does islet architecture matter to energy homeostasis, and what may happen if we lose a particular population of islet cells in favour of β-cells? These are all questions to which we will need answers for islet replacement therapy by transdifferentiation of non-β islet cells to be a reality in humans. We know a fair amount about the biology of β-cells but not quite as much about the other islet cell types. Until recently, we have not had a good grasp of islet mass and distribution in the human pancreas. In this review, we will look at current data on islet cells, focussing more on non-β cells, and on human pancreatic islet mass and distribution.
Collapse
Affiliation(s)
- Gabriela Da Silva Xavier
- Section of Functional Genomics and Cell Biology, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston B15 2TT, UK.
| |
Collapse
|
38
|
Müller M, Canfora EE, Blaak EE. Gastrointestinal Transit Time, Glucose Homeostasis and Metabolic Health: Modulation by Dietary Fibers. Nutrients 2018; 10:nu10030275. [PMID: 29495569 PMCID: PMC5872693 DOI: 10.3390/nu10030275] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/16/2018] [Accepted: 02/26/2018] [Indexed: 12/19/2022] Open
Abstract
Gastrointestinal transit time may be an important determinant of glucose homeostasis and metabolic health through effects on nutrient absorption and microbial composition, among other mechanisms. Modulation of gastrointestinal transit may be one of the mechanisms underlying the beneficial health effects of dietary fibers. These effects include improved glucose homeostasis and a reduced risk of developing metabolic diseases such as obesity and type 2 diabetes mellitus. In this review, we first discuss the regulation of gastric emptying rate, small intestinal transit and colonic transit as well as their relation to glucose homeostasis and metabolic health. Subsequently, we briefly address the reported health effects of different dietary fibers and discuss to what extent the fiber-induced health benefits may be mediated through modulation of gastrointestinal transit.
Collapse
Affiliation(s)
- Mattea Müller
- Department of Human Biology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Emanuel E Canfora
- Department of Human Biology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
39
|
Pekmez CT, Dragsted LO, Brahe LK. Gut microbiota alterations and dietary modulation in childhood malnutrition - The role of short chain fatty acids. Clin Nutr 2018; 38:615-630. [PMID: 29496274 DOI: 10.1016/j.clnu.2018.02.014] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 02/02/2018] [Accepted: 02/08/2018] [Indexed: 12/12/2022]
Abstract
The gut microbiome affects the health status of the host through different mechanisms and is associated with a wide variety of diseases. Both childhood undernutrition and obesity are linked to alterations in composition and functionality of the gut microbiome. One of the possible mechanisms underlying the interplay between microbiota and host metabolism is through appetite-regulating hormones (including leptin, ghrelin, glucagon-like peptide-1). Short chain fatty acids, the end product of bacterial fermentation of non-digestible carbohydrates, might be able to alter energy harvest and metabolism through enteroendocrine cell signaling, adipogenesis and insulin-like growth factor-1 production. Elucidating these mechanisms may lead to development of new modulation practices of the gut microbiota as a potential prevention and treatment strategy for childhood malnutrition. The present overview will briefly outline the gut microbiota development in the early life, gut microbiota alterations in childhood undernutrition and obesity, and whether this relationship is causal. Further we will discuss possible underlying mechanisms in relation to the gut-brain axis and short chain fatty acids, and the potential of probiotics, prebiotics and synbiotics for modulating the gut microbiota during childhood as a prevention and treatment strategy against undernutrition and obesity.
Collapse
Affiliation(s)
- Ceyda Tugba Pekmez
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark; Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey.
| | - Lars Ove Dragsted
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Lena Kirchner Brahe
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
40
|
Abstract
Importance of the neuroendocrine brain for health and happiness has become clear since the 1960s. Foundations laid 100 years ago culminated in Geoffrey W Harris's model of control by the brain of secretion of anterior and posterior pituitary gland hormones through, respectively, releasing factors secreted into the hypothalamic-hypophysial portal system, and directly from axon terminals into the systemic circulation. Confirmation, expansion and deepening of knowledge and understanding have followed increasingly sophisticated technology. This allowed chemical characterisation of the posterior pituitary hormones, oxytocin and vasopressin, the releasing factors, their receptors and genes, location of the neurosecretory neurons in the hypothalamus, and how their activity is controlled, including by neural and hormonal feedback, and how hormone rhythms are generated. Wider roles of these neurons and their peptides in the brain are now recognised: in reproductive and social behaviours, emotions and appetite. Plasticity and epigenetic programming of neuroendocrine systems have emerged as important features.
Collapse
Affiliation(s)
- John A. Russell
- Professor Emeritus, Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, UK
| |
Collapse
|
41
|
Lach G, Schellekens H, Dinan TG, Cryan JF. Anxiety, Depression, and the Microbiome: A Role for Gut Peptides. Neurotherapeutics 2018; 15:36-59. [PMID: 29134359 PMCID: PMC5794698 DOI: 10.1007/s13311-017-0585-0] [Citation(s) in RCA: 358] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complex bidirectional communication between the gut and the brain is finely orchestrated by different systems, including the endocrine, immune, autonomic, and enteric nervous systems. Moreover, increasing evidence supports the role of the microbiome and microbiota-derived molecules in regulating such interactions; however, the mechanisms underpinning such effects are only beginning to be resolved. Microbiota-gut peptide interactions are poised to be of great significance in the regulation of gut-brain signaling. Given the emerging role of the gut-brain axis in a variety of brain disorders, such as anxiety and depression, it is important to understand the contribution of bidirectional interactions between peptide hormones released from the gut and intestinal bacteria in the context of this axis. Indeed, the gastrointestinal tract is the largest endocrine organ in mammals, secreting dozens of different signaling molecules, including peptides. Gut peptides in the systemic circulation can bind cognate receptors on immune cells and vagus nerve terminals thereby enabling indirect gut-brain communication. Gut peptide concentrations are not only modulated by enteric microbiota signals, but also vary according to the composition of the intestinal microbiota. In this review, we will discuss the gut microbiota as a regulator of anxiety and depression, and explore the role of gut-derived peptides as signaling molecules in microbiome-gut-brain communication. Here, we summarize the potential interactions of the microbiota with gut hormones and endocrine peptides, including neuropeptide Y, peptide YY, pancreatic polypeptide, cholecystokinin, glucagon-like peptide, corticotropin-releasing factor, oxytocin, and ghrelin in microbiome-to-brain signaling. Together, gut peptides are important regulators of microbiota-gut-brain signaling in health and stress-related psychiatric illnesses.
Collapse
Affiliation(s)
- Gilliard Lach
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Harriet Schellekens
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Food for Health Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland.
- Food for Health Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
42
|
Holzer P, Farzi A, Hassan AM, Zenz G, Jačan A, Reichmann F. Visceral Inflammation and Immune Activation Stress the Brain. Front Immunol 2017; 8:1613. [PMID: 29213271 PMCID: PMC5702648 DOI: 10.3389/fimmu.2017.01613] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022] Open
Abstract
Stress refers to a dynamic process in which the homeostasis of an organism is challenged, the outcome depending on the type, severity, and duration of stressors involved, the stress responses triggered, and the stress resilience of the organism. Importantly, the relationship between stress and the immune system is bidirectional, as not only stressors have an impact on immune function, but alterations in immune function themselves can elicit stress responses. Such bidirectional interactions have been prominently identified to occur in the gastrointestinal tract in which there is a close cross-talk between the gut microbiota and the local immune system, governed by the permeability of the intestinal mucosa. External stressors disturb the homeostasis between microbiota and gut, these disturbances being signaled to the brain via multiple communication pathways constituting the gut-brain axis, ultimately eliciting stress responses and perturbations of brain function. In view of these relationships, the present article sets out to highlight some of the interactions between peripheral immune activation, especially in the visceral system, and brain function, behavior, and stress coping. These issues are exemplified by the way through which the intestinal microbiota as well as microbe-associated molecular patterns including lipopolysaccharide communicate with the immune system and brain, and the mechanisms whereby overt inflammation in the GI tract impacts on emotional-affective behavior, pain sensitivity, and stress coping. The interactions between the peripheral immune system and the brain take place along the gut-brain axis, the major communication pathways of which comprise microbial metabolites, gut hormones, immune mediators, and sensory neurons. Through these signaling systems, several transmitter and neuropeptide systems within the brain are altered under conditions of peripheral immune stress, enabling adaptive processes related to stress coping and resilience to take place. These aspects of the impact of immune stress on molecular and behavioral processes in the brain have a bearing on several disturbances of mental health and highlight novel opportunities of therapeutic intervention.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Geraldine Zenz
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Angela Jačan
- CBmed GmbH-Center for Biomarker Research in Medicine, Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| |
Collapse
|
43
|
Chen S, Zeng K, Liu QC, Guo Z, Zhang S, Chen XR, Lin JH, Wen JP, Zhao CF, Lin XH, Gao F. Adropin deficiency worsens HFD-induced metabolic defects. Cell Death Dis 2017; 8:e3008. [PMID: 28837146 PMCID: PMC5596552 DOI: 10.1038/cddis.2017.362] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/13/2022]
Abstract
The limited efficacy of current treatment methods and increased type 2 diabetes mellitus (T2DM) incidence constitute an incentive for investigating how metabolic homeostasis is maintained, to improve treatment efficacy and identify novel treatment methods. We analyzed a three-generation family of Chinese origin with the common feature of T2DM attacks and fatty pancreas (FP), alongside 19 unrelated patients with FP and 58 cases with T2DM for genetic variations in Enho, serum adropin, and relative Treg amounts. Functional studies with adropin knockout (AdrKO) in C57BL/6J mice were also performed. It showed serum adropin levels were significantly lower in FP and T2DM patients than in healthy subjects; relative Treg amounts were also significantly decreased in FP and T2DM patients, and positively associated with adropin (r=0.7220, P=0.0001). Sequencing revealed that the patients shared a Cys56Trp mutation in Enho. In vivo, adropin-deficiency was associated with increased severity of glucose homeostasis impairment and fat metabolism disorder. AdrKO mice exhibited reduced endothelial nitric oxide synthase (eNOS) phosphorylation (Ser1177), impaired glycosphingolipid biosynthesis, adipocytes infiltrating, and loss of Treg, and developed FP and T2DM. Adropin-deficiency contributed to loss of Treg and the development of FP disease and T2DM.
Collapse
Affiliation(s)
- Shi Chen
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China
| | - Kai Zeng
- Department of Anesthesiology, 1st Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qi-cai Liu
- Department of Laboratory Medicine, 1st Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zheng Guo
- Department of Bioinformatics, Fujian Medical University, Fuzhou, China
| | - Sheng Zhang
- Department of Pathology, 1st Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiao-rong Chen
- Department of Radiology, 1st Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jian-hua Lin
- Department of Central Laboratory, 1st Affiliated Hospital, Fuzhou, China
| | - Jun-ping Wen
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
| | - Cheng-fei Zhao
- Department of Pharmaceutical Analysis, Putian University, Putian, China
| | - Xin-hua Lin
- Department of Pharmaceutical Analysis, Fujian Medical University, Fuzhou, China
| | - Feng Gao
- Department of Pathology, 1st Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
44
|
Rahat-Rozenbloom S, Fernandes J, Cheng J, Wolever TMS. Acute increases in serum colonic short-chain fatty acids elicited by inulin do not increase GLP-1 or PYY responses but may reduce ghrelin in lean and overweight humans. Eur J Clin Nutr 2017; 71:953-958. [PMID: 27966574 PMCID: PMC5423780 DOI: 10.1038/ejcn.2016.249] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/19/2016] [Accepted: 10/26/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Colonic fermentation of dietary fibre to short-chain fatty acids (SCFA) influences appetite hormone secretion in animals, but SCFA production is excessive in obese animals. This suggests there may be resistance to the effect of SCFA on appetite hormones in obesity. OBJECTIVES To determine the effects of inulin (IN) and resistant starch (RS) on postprandial SCFA, and gut hormone (glucagon-like peptide (GLP-1), peptide-tyrosine-tyrosine (PYY) and ghrelin) responses in healthy overweight/obese (OWO) vs lean (LN) humans. SUBJECTS/METHODS Overnight-fasted participants (13 OWO and 12 LN) consumed 300 ml water containing 75 g glucose (GLU) as control or 75 g GLU plus 24 g IN, or 28.2 g RS using a randomised, single-blind, cross-over design. Blood for appetite hormones and SCFA was collected at intervals over 6 h. A standard lunch was served 4 h after the test drink. RESULTS Relative to GLU, IN, but not RS, significantly increased SCFA areas under the curve (AUC) from 4-6 h (AUC4-6). Neither IN nor RS affected GLP-1 or PYY-AUC4-6. Although neither IN nor RS reduced ghrelin-AUC4-6 compared with GLU, ghrelin at 6 h after IN was significantly lower than that after GLU (P<0.05). After IN, relative to GLU, the changes in SCFA-AUC4-6 were negatively related to the changes in ghrelin-AUC4-6 (P=0.017). SCFA and hormone responses did not differ significantly between LN and OWO. CONCLUSIONS Acute increases in colonic SCFA do not affect GLP-1 or PYY responses in LN or OWO subjects, but may reduce ghrelin. The results do not support the hypothesis that SCFA acutely stimulate PYY and GLP-1 secretion; however, a longer adaptation to increased colonic fermentation or a larger sample size may yield different results.
Collapse
Affiliation(s)
- Sari Rahat-Rozenbloom
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Judlyn Fernandes
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ji Cheng
- Biostatistics Unit, St. Joseph’s Healthcare Hamilton, Charlton East, Hamilton, ON, Canada L8N 4A6
| | - Thomas MS Wolever
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Clinical Nutrition and Risk Factor Modification Centre, and Division of Endocrinology and Metabolism and Li Ka Shing Knowledge Institute, St. Michael’s Hospital Toronto, Ontario, Canada
| |
Collapse
|
45
|
Distribution of Pancreatic Polypeptide–secreting Endocrine Cells in Nondiabetic and Diabetic Cases. Appl Immunohistochem Mol Morphol 2017; 25:422-431. [DOI: 10.1097/pai.0000000000000310] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
46
|
Martel J, Ojcius DM, Chang CJ, Lin CS, Lu CC, Ko YF, Tseng SF, Lai HC, Young JD. Anti-obesogenic and antidiabetic effects of plants and mushrooms. Nat Rev Endocrinol 2017; 13:149-160. [PMID: 27636731 DOI: 10.1038/nrendo.2016.142] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Obesity is reaching global epidemic proportions as a result of factors such as high-calorie diets and lack of physical exercise. Obesity is now considered to be a medical condition, which not only contributes to the risk of developing type 2 diabetes mellitus, cardiovascular disease and cancer, but also negatively affects longevity and quality of life. To combat this epidemic, anti-obesogenic approaches are required that are safe, widely available and inexpensive. Several plants and mushrooms that are consumed in traditional Chinese medicine or as nutraceuticals contain antioxidants, fibre and other phytochemicals, and have anti-obesogenic and antidiabetic effects through the modulation of diverse cellular and physiological pathways. These effects include appetite reduction, modulation of lipid absorption and metabolism, enhancement of insulin sensitivity, thermogenesis and changes in the gut microbiota. In this Review, we describe the molecular mechanisms that underlie the anti-obesogenic and antidiabetic effects of these plants and mushrooms, and propose that combining these food items with existing anti-obesogenic approaches might help to reduce obesity and its complications.
Collapse
Affiliation(s)
- Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, 155 Fifth Street, San Francisco, California 94103, USA
| | - Chih-Jung Chang
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Department of Microbiology and Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Research Center of Bacterial Pathogenesis, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
| | - Chuan-Sheng Lin
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Department of Microbiology and Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Research Center of Bacterial Pathogenesis, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, 510 Zhong-Zheng Street, New Taipei City 24205, Taiwan, Republic of China
| | - Yun-Fei Ko
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Chang Gung Biotechnology Corporation, 201 Tung-Hua North Road, Taipei 10508, Taiwan, Republic of China
- Biochemical Engineering Research Center, Ming Chi University of Technology, 84 Gungjuan Road, New Taipei City 24301, Taiwan, Republic of China
| | - Shun-Fu Tseng
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Research Center of Bacterial Pathogenesis, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
| | - Hsin-Chih Lai
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Department of Microbiology and Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Research Center of Bacterial Pathogenesis, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, 261 Wen-Hua First Road, Taoyuan 33303, Taiwan, Republic of China
- Graduate Institute of Health Industry and Technology, College of Human Ecology, Chang Gung University of Science and Technology, 261 Wen-Hua First Road, Taoyuan 33303, Taiwan, Republic of China
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan, Republic of China
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan, Republic of China
- Chang Gung Biotechnology Corporation, 201 Tung-Hua North Road, Taipei 10508, Taiwan, Republic of China
- Biochemical Engineering Research Center, Ming Chi University of Technology, 84 Gungjuan Road, New Taipei City 24301, Taiwan, Republic of China
- Laboratory of Cellular Physiology and Immunology, Rockefeller University, 1230 York Avenue, New York, New York 10021, USA
| |
Collapse
|
47
|
Ly HG, Dupont P, Van Laere K, Depoortere I, Tack J, Van Oudenhove L. Differential brain responses to gradual intragastric nutrient infusion and gastric balloon distension: A role for gut peptides? Neuroimage 2017; 144:101-112. [DOI: 10.1016/j.neuroimage.2016.09.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 08/18/2016] [Accepted: 09/13/2016] [Indexed: 12/15/2022] Open
|
48
|
Amato A, Baldassano S, Caldara GF, Mulè F. Pancreatic polypeptide stimulates mouse gastric motor activity through peripheral neural mechanisms. Neurogastroenterol Motil 2017; 29. [PMID: 27381051 DOI: 10.1111/nmo.12901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/10/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Pancreatic polypeptide (PP) is supposed to be one of the major endogenous agonists of the neuropeptide Y4 receptor. Pancreatic polypeptide can influence gastrointestinal motility, acting mainly through vagal mechanisms, but whether PP acts directly on the stomach has not been explored yet. The aims of this study were to investigate the effects of PP on mouse gastric emptying, on spontaneous tone of whole stomach in vitro and to examine the mechanism of action. METHODS Gastric emptying was measured by red phenol method after i.p. PP administration (1-3 nmol per mouse). Responses induced by PP (1-300 mmol L-1 ) on gastric endoluminal pressure were analyzed in vitro in the presence of different drugs. Gastric genic expression of Y4 receptor was verified by RT-PCR. KEY RESULTS Pancreatic polypeptide dose-dependently increased non-nutrient liquid gastric emptying rate. In vitro, PP produced a concentration-dependent contraction that was abolished by tetrodotoxin, a neural blocker of Na+ voltage-dependent channels. The contractile response was significantly reduced by atropine, a muscarinic receptor antagonist, and by SR48968, an NK2 receptor antagonist, while it was potentiated by neostigmine, an inhibitor of acetylcholinesterase. The joint application of atropine and SR48968 fully abolished PP contractile effect. Reverse transcriptase-polymerase chain reaction analysis revealed the presence of Y4 receptor mRNA in mouse stomach with a greater expression in antrum than in fundus. CONCLUSIONS & INFERENCES The present findings demonstrate that exogenous PP stimulates mouse gastric motor activity, by acting directly on the stomach. This effect appears due to the activation of enteric excitatory neurons releasing acetylcholine and tachykinins.
Collapse
Affiliation(s)
- A Amato
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - S Baldassano
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - G F Caldara
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - F Mulè
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| |
Collapse
|
49
|
Thongrod S, Changklungmoa N, Chansela P, Siangcham T, Kruangkum T, Suwansa-Ard S, Saetan J, Sroyraya M, Tinikul Y, Wanichanon C, Sobhon P. Characterization and tissue distribution of neuropeptide F in the eyestalk and brain of the male giant freshwater prawn, Macrobrachium rosenbergii. Cell Tissue Res 2016; 367:181-195. [DOI: 10.1007/s00441-016-2538-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/16/2016] [Indexed: 10/20/2022]
|
50
|
Kim KH, Jang HJ. Development of GLP-1 secretagogue using microarray in enteroendocrine L cells. BIOCHIP JOURNAL 2016. [DOI: 10.1007/s13206-016-0403-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|