1
|
Liu N, Liu T, Alim N, Zou J, Hu X, Zhang B, Liu F, Ding X, Sun P, Wang S, Zhang L, Zheng R, Liang R. 5-HT promotes pancreatic α-to-β cell transdifferentiation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119958. [PMID: 40222656 DOI: 10.1016/j.bbamcr.2025.119958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/31/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
The role of 5-HT in maintaining glucose homeostasis during metabolic stress and inhibiting glucagon secretion is well documented, however, its effect on α cell identity remained unclear. In this study, we demonstrated that 5-HT suppressed the expression of α cell markers, such as Arx and Gcg, while enhancing the expression of β cell markers in mouse pancreatic α cell lines. We further found that treatment with 5-HT significantly increased the percentage of Gcg+Ins+ and Gcg+Nkx6.1+ cells in isolated human and mouse islets. Using pancreatic α cell lineage-tracing Gcgcre+; tdTomato/tdTomato mice, we observed that 5-HT treatment significantly reduced random blood glucose levels and increased tdTomato+Ins+, Gcg+Ins+ and Gcg+Nkx6.1+ cells in a high fat diet and streptozotocin (HFD + STZ) induced diabetes model. Additionally, in situ detection of 5-HT production in the human pancreas revealed a reduction of 5-HT expression in β cells of human T2D patients. These findings suggest that 5-HT treatment induces the transdifferentiation of α to β cells, potentially contributing to the recovery of β cell mass in T2D.
Collapse
Affiliation(s)
- Na Liu
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China
| | - Tengli Liu
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China
| | - Nijat Alim
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jiaqi Zou
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China
| | - Xiaoyan Hu
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China
| | - Boya Zhang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China
| | - Fei Liu
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Xuejie Ding
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China
| | - Peng Sun
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China; Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, PR China; School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, PR China.
| | - Rongxiu Zheng
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin 300052, PR China.
| | - Rui Liang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, PR China; NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, PR China.
| |
Collapse
|
2
|
Zhuang M, Wang B, Shi Y, Zhou Z. Multiorgan Regulation Mechanisms and Nutritional Intervention Strategies in Gestational Diabetes Mellitus. J Nutr 2025:S0022-3166(25)00192-0. [PMID: 40222585 DOI: 10.1016/j.tjnut.2025.04.008] [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: 01/06/2025] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025] Open
Abstract
Gestational diabetes mellitus (GDM) affects millions of pregnant women worldwide and leads to both short- and long-term complications for mothers and their fetuses. Managing GDM through diet, physical activity, and medical interventions can significantly reduce these risks. Studies have identified the individual and combined roles of organs regulated by placental hormones, cytokines, and gut microbiota as key pathways contributing to impaired glucose homeostasis. In this context, placental hormones mediate the crosstalk among the placenta, pancreas, and adipose tissue, stimulating endocrine pancreas adaptation and adipose tissue expansion. However, insufficient maternal physiological adaptations, such as dysregulated adipocytokines, adipokines, and oxidative stress in the pancreas, can create an environment conducive to the onset of GDM. Furthermore, gut dysbiosis implies potential mechanisms of gut-host interaction associated with the occurrence of GDM, with short-chain fatty acids possibly serving as crucial targets. Nutritional therapy is recognized as the first-line approach for managing GDM, encompassing dietary guidance and supplementation with micro- and macronutrients as well as bioactive components. Importantly, combined interventions involving multiple nutrients, such as probiotics and prebiotics with vitamins or minerals, may exert stronger beneficial effects on the prevention and treatment of GDM and its complications. This review paper discusses the regulatory role of multiorgans in GDM and the implementation of nutritional therapy for its prevention and management, along with associated complications.
Collapse
Affiliation(s)
- Min Zhuang
- College of Food Science, Shihezi University, Shihezi, China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Bing Wang
- Gulbali Institute-Agriculture Water Environment, Charles Sturt University, Wagga Wagga, NSW, Australia.
| | - Yanchuan Shi
- Neuroendocrinology Group, Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Zhongkai Zhou
- College of Food Science, Shihezi University, Shihezi, China; College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China; Gulbali Institute-Agriculture Water Environment, Charles Sturt University, Wagga Wagga, NSW, Australia.
| |
Collapse
|
3
|
Abedini F, Amjadi O, Ahangari G. Repurposing serotonergic drugs for gastric cancer: induction of apoptosis in vitro. Mol Biol Rep 2025; 52:373. [PMID: 40202572 DOI: 10.1007/s11033-025-10474-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Gastric cancer is a highly heterogeneous and aggressive disease with limited treatment options, necessitating innovative therapeutic strategies. Drug repurposing, a cost-effective approach, offers opportunities to identify new applications for existing medications. This study systematically investigated the apoptotic effects of serotonergic drugs on MKN-45 gastric cancer cells, providing a novel perspective on serotonin signaling in cancer therapy. METHODS AND RESULTS MKN-45 cells were treated with concentrations of Tropisetron, Imipramine, Ketanserin, Citalopram, and Cyproheptadine. The IC50 values were determined using an MTT assay, while acridine orange/ethidium bromide staining and Annexin V/PI flow cytometry assessed apoptotic activity. Gene expression related to serotonin receptors (HTR2A, HTR2B, HTR3A), Serotonin transporter (SLC6A4), apoptosis (Bcl-2, Bax), and proliferation (PCNA) was evaluated via real-time PCR. Tropisetron, Imipramine, Ketanserin, and Cyproheptadine demonstrated statistically significant apoptotic induction compared to untreated cells. These treatments significantly reduced anti-apoptotic Bcl-2 and PCNA, proliferation marker, expression, while pro-apoptotic Bax expression was markedly elevated (p < 0.05). CONCLUSIONS This study highlights the potential of Tropisetron, Imipramine, Ketanserin, and Cyproheptadine as repurposed drugs for gastric cancer therapy, with Tropisetron and Imipramine showing particularly promising apoptotic effects. These findings pave the way for further preclinical and clinical investigations, offering a foundation for personalized therapeutic strategies in gastric cancer management.
Collapse
Affiliation(s)
- Fatemeh Abedini
- Neuroimmunopsychooncogenetic Group, Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, P.O. Box: 1497716316, Tehran, Iran
| | - Omolbanin Amjadi
- Neuroimmunopsychooncogenetic Group, Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, P.O. Box: 1497716316, Tehran, Iran
| | - Ghasem Ahangari
- Neuroimmunopsychooncogenetic Group, Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, P.O. Box: 1497716316, Tehran, Iran.
| |
Collapse
|
4
|
Li G, Dong S, Liu C, Yang J, Rensen PCN, Wang Y. Serotonin signaling to regulate energy metabolism: a gut microbiota perspective. LIFE METABOLISM 2025; 4:loae039. [PMID: 39926388 PMCID: PMC11803461 DOI: 10.1093/lifemeta/loae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 02/11/2025]
Abstract
Serotonin is one of the most potent gastrointestinal, peripheral, and neuronal signaling molecules and plays a key role in regulating energy metabolism. Accumulating evidence has shown the complex interplay between gut microbiota and host energy metabolism. In this review, we summarize recent findings on the role of gut microbiota in serotonin metabolism and discuss the complicated mechanisms by which serotonin, working in conjunction with the gut microbiota, affects total body energy metabolism in the host. Gut microbiota affects serotonin synthesis, storage, release, transport, and catabolism. In addition, serotonin plays an indispensable role in regulating host energy homeostasis through organ crosstalk and microbe-host communication, particularly with a wide array of serotonergic effects mediated by diverse serotonin receptors with unique tissue specificity. This fresh perspective will help broaden the understanding of serotonergic signaling in modulating energy metabolism, thus shedding light on the design of innovative serotonin-targeting strategies to treat metabolic diseases.
Collapse
Affiliation(s)
- Guoli Li
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Sijing Dong
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Chunhao Liu
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Jing Yang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Patrick C N Rensen
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Yanan Wang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
5
|
Wu WH, Zhi H, Feng WK, Jiang L, Yang L, Qian LQ, Zhao RX, Tan YM, Yang HY, Liu XD, Liu L. Clozapine impaired glucose-stimulated insulin secretion partly by increasing plasma 5-HT levels due to the inhibition of OCT1-mediated hepatic 5-HT uptake in mice. Acta Pharmacol Sin 2025; 46:687-701. [PMID: 39472495 PMCID: PMC11845781 DOI: 10.1038/s41401-024-01401-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/22/2024] [Indexed: 02/23/2025]
Abstract
Patients taking atypical antipsychotics (AAPs), especially clozapine, are often associated with hyperglycaemia. Here, clozapine served as a representative agent for investigating how AAPs induce hyperglycaemia. In normal mice and mice fed a high fat diet (HFD), clozapine impaired glucose tolerance and glucose-stimulated insulin secretion (GSIS) following intraperitoneal glucose administration and increased plasma 5-HT levels. Intraperitoneal 5-HT administration also impaired glucose tolerance and GSIS in mice. In INS-1 cells, high 5-HT levels impaired GSIS, which was attenuated by the 5-HTR3 antagonist tropisetron or by silencing 5-HTR3a. The 5-HTR2a agonist TCB2 attenuated clozapine-induced GSIS impairment. Silencing 5-HTR2a or the 5-HTR2a antagonist ketanserin impaired GSIS. In mice, 5-HT administration impaired GSIS, which was attenuated by tropisetron but aggravated by clozapine. Clozapine increased plasma [2H]5-HT exposure following intravenous administration to mice. In HEK293-OCT1 cells, clozapine inhibited [2H]5-HT and MPP+ uptake. Clozapine or OCT1 silencing impaired 5-HT metabolism in mouse primary hepatocytes, demonstrating that clozapine increased plasma 5-HT levels via the inhibition of OCT1-mediated hepatic 5-HT uptake. Liver-specific silencing of OCT1 increased plasma [2H]5-HT exposure and 5-HT levels and impaired GSIS and glucose tolerance in mice. In conclusion, clozapine impaired GSIS and glucose tolerance by increasing plasma 5-HT levels via the inhibition of OCT1-mediated hepatic 5-HT uptake. Increased 5-HT impaired GSIS by activating islet 5-HTR3a. The antagonistic effect of clozapine on islet 5-HTR2a also contributed to GSIS impairment. The finding that clozapine-induced GSIS impairment was attributed to increased 5-HT levels via the inhibition of OCT1-mediated hepatic 5-HT uptake may partly explain hyperglycaemia caused by other AAPs.
Collapse
Affiliation(s)
- Wen-Han Wu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Hao Zhi
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Wen-Ke Feng
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Ling Jiang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Lu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Li-Qiang Qian
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Rui-Xi Zhao
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Yong-Mei Tan
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China
| | - Han-Yu Yang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China.
| | - Xiao-Dong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China.
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210029, China.
| |
Collapse
|
6
|
Lee J, Lee NK, Moon JH. Gestational Diabetes Mellitus: Mechanisms Underlying Maternal and Fetal Complications. Endocrinol Metab (Seoul) 2025; 40:10-25. [PMID: 39844628 PMCID: PMC11898322 DOI: 10.3803/enm.2024.2264] [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: 12/03/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Gestational diabetes mellitus (GDM) affects over 10% of all pregnancies, both in Korea and worldwide. GDM not only increases the risk of adverse pregnancy outcomes such as preeclampsia, preterm birth, macrosomia, neonatal hypoglycemia, and shoulder dystocia, but it also significantly increases the risk of developing postpartum type 2 diabetes mellitus and cardiovascular disease in the mother. Additionally, GDM is linked to a higher risk of childhood obesity and diabetes in offspring, as well as neurodevelopmental disorders, including autistic spectrum disorder. This review offers a comprehensive summary of clinical epidemiological studies concerning maternal and fetal complications and explores mechanistic investigations that reveal the underlying pathophysiology.
Collapse
Affiliation(s)
- Jooyeop Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Armed Forces Yangju Hospital, Yangju, Korea
| | - Na Keum Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Joon Ho Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
7
|
Sylvester-Armstrong KR, Reeder CF, Powell A, Becker MW, Hagan DW, Chen J, Mathews CE, Wasserfall CH, Atkinson MA, Egerman R, Phelps EA. Serum from pregnant donors induces human beta cell proliferation. Islets 2024; 16:2334044. [PMID: 38533763 PMCID: PMC10978022 DOI: 10.1080/19382014.2024.2334044] [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: 10/11/2023] [Revised: 02/20/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
Pancreatic beta cells are among the slowest replicating cells in the human body and have not been observed to increase in number except during the fetal and neonatal period, in cases of obesity, during puberty, as well as during pregnancy. Pregnancy is associated with increased beta cell mass to meet heightened insulin demands. This phenomenon raises the intriguing possibility that factors present in the serum of pregnant individuals may stimulate beta cell proliferation and offer insights into expansion of the beta cell mass for treatment and prevention of diabetes. The primary objective of this study was to test the hypothesis that serum from pregnant donors contains bioactive factors capable of inducing human beta cell proliferation. An immortalized human beta cell line with protracted replication (EndoC-βH1) was cultured in media supplemented with serum from pregnant and non-pregnant female and male donors and assessed for differences in proliferation. This experiment was followed by assessment of proliferation of primary human beta cells. Sera from five out of six pregnant donors induced a significant increase in the proliferation rate of EndoC-βH1 cells. Pooled serum from the cohort of pregnant donors also increased the rate of proliferation in primary human beta cells. This study demonstrates that serum from pregnant donors stimulates human beta cell proliferation. These findings suggest the existence of pregnancy-associated factors that can offer novel avenues for beta cell regeneration and diabetes prevention strategies. Further research is warranted to elucidate the specific factors responsible for this effect.
Collapse
Affiliation(s)
| | - Callie F. Reeder
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Andrece Powell
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Matthew W. Becker
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - D. Walker Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Jing Chen
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Robert Egerman
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
8
|
Toczyska K, Haq N, Lyu Z, Bewick G, Zhao M, Rosa H, Starikova J, Liu B, Persaud SJ. The selective serotonin reuptake inhibitors, sertraline and paroxetine, improve islet beta-cell mass and function in vitro. Diabetes Obes Metab 2024; 26:3606-3617. [PMID: 38888050 PMCID: PMC11639051 DOI: 10.1111/dom.15701] [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: 12/19/2023] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024]
Abstract
AIMS To investigate the effects of the selective serotonin reuptake inhibitors (SSRIs) sertraline and paroxetine at therapeutically relevant concentrations on beta-cell mass and function. METHODS Viability was quantified in mouse insulinoma (MIN6) beta cells and mouse islets after 48-h exposure to sertraline (1-10 μM) or paroxetine (0.01-1 μM) using the Trypan blue exclusion test. The effects of therapeutic concentrations of these SSRIs on insulin secretion were determined by static incubation and perifusion experiments, while islet apoptosis was investigated by Caspase-Glo 3/7 assay, TUNEL staining and quantitative PCR analysis. Finally, proliferation of MIN6 and mouse islet beta cells was assessed by bromodeoxyuridine (BrdU) enzyme-linked immunosorbent assay and immunofluorescence. RESULTS Sertraline (0.1-1 μM) and paroxetine (0.01-0.1 μM) were well tolerated by MIN6 beta cells and islets, whereas 10 μM sertraline and 1 μM paroxetine were cytotoxic. Exposure to 1 μM sertraline and 0.1 μM paroxetine significantly potentiated glucose-stimulated insulin secretion from mouse and human islets. Moreover, they showed protective effects against cytokine- and palmitate-induced apoptosis of islets, they downregulated cytokine-induced Stat1 and Traf1 mRNA expression, and they significantly increased proliferation of mouse beta cells. CONCLUSIONS Our data demonstrate that sertraline and paroxetine act directly on beta cells to enhance glucose-stimulated insulin secretion and stimulate beta-cell mass expansion by increasing proliferation and decreasing apoptosis. These drugs are therefore likely to be appropriate for treating depression in people with type 2 diabetes.
Collapse
Affiliation(s)
- Klaudia Toczyska
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Naila Haq
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Zekun Lyu
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Gavin Bewick
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Min Zhao
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Hannah Rosa
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Jessica Starikova
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Bo Liu
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| | - Shanta Jean Persaud
- Department of DiabetesSchool of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUK
| |
Collapse
|
9
|
Lee RA, Chopra DG, Nguyen V, Huang XP, Zhang Y, Shariati K, Yiv N, Schugar R, Annes J, Roth B, Ku GM. An shRNA screen in primary human beta cells identifies the serotonin 1F receptor as a negative regulator of survival during transplant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.591950. [PMID: 38746433 PMCID: PMC11092577 DOI: 10.1101/2024.05.01.591950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Islet transplantation can cure type 1 diabetes, but peri-transplant beta cell death limits this procedure to those with low insulin requirements. Improving human beta cell survival or proliferation may make islet transplantation a possibility for more type 1 patients. To identify novel regulators of beta cell survival and proliferation, we conducted a pooled small hairpin RNA (shRNA) screen in primary human beta cells transplanted into immunocompromised mice. shRNAs targeting several cyclin dependent kinase inhibitors were enriched after transplant. Here, we focused on the Gi/o-coupled GPCR, serotonin 1F receptor ( HTR1F, 5-HT 1F ) which our screen identified as a negative regulator of beta cell numbers after transplant. In vitro , 5-HT 1F knockdown induced human beta cell proliferation but only when combined with harmine and exendin-4. In vivo , knockdown of 5-HT 1F reduced beta cell death during transplant. To demonstrate the feasibility of targeting 5-HT 1F in islet transplant, we identified and validated a small molecule 5-HT 1F antagonist. This antagonist increased glucose stimulated insulin secretion from primary human islets and cAMP accumulation in primary human beta cells. Finally, the 5-HT 1F antagonist improved glycemia in marginal mass, human islet transplants into immunocompromised mice. We identify 5-HT 1F as a novel druggable target to improve human beta cell survival in the setting of islet transplantation. One Sentence Summary Serotonin 1F receptor (5-HT 1F ) negatively regulates insulin secretion and beta cell survival during transplant.
Collapse
|
10
|
Norrby K. On Connective Tissue Mast Cells as Protectors of Life, Reproduction, and Progeny. Int J Mol Sci 2024; 25:4499. [PMID: 38674083 PMCID: PMC11050338 DOI: 10.3390/ijms25084499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The connective tissue mast cell (MC), a sentinel tissue-residing secretory immune cell, has been preserved in all vertebrate classes since approximately 500 million years. No physiological role of the MC has yet been established. Considering the power of natural selection of cells during evolution, it is likely that the MCs exert essential yet unidentified life-promoting actions. All vertebrates feature a circulatory system, and the MCs interact readily with the vasculature. It is notable that embryonic MC progenitors are generated from endothelial cells. The MC hosts many surface receptors, enabling its activation via a vast variety of potentially harmful exogenous and endogenous molecules and via reproductive hormones in the female sex organs. Activated MCs release a unique composition of preformed and newly synthesized bioactive molecules, like heparin, histamine, serotonin, proteolytic enzymes, cytokines, chemokines, and growth factors. MCs play important roles in immune responses, tissue remodeling, cell proliferation, angiogenesis, inflammation, wound healing, tissue homeostasis, health, and reproduction. As recently suggested, MCs enable perpetuation of the vertebrates because of key effects-spanning generations-in ovulation and pregnancy, as in life-preserving activities in inflammation and wound healing from birth till reproductive age, thus creating a permanent life-sustaining loop. Here, we present recent advances that further indicate that the MC is a specific life-supporting and progeny-safeguarding cell.
Collapse
Affiliation(s)
- Klas Norrby
- Department of Pathology, Institute of Medical Biology, Sahlgren Academy, University of Gothenburg, 7 Ostindiefararen, SE-417 65 Gothenburg, Sweden
| |
Collapse
|
11
|
Hill TG, Smith LIF, Ruz-Maldonado I, Jones PM, Bowe JE. Kisspeptin upregulates β-cell serotonin production during pregnancy. J Endocrinol 2024; 260:e230218. [PMID: 37997938 PMCID: PMC10762540 DOI: 10.1530/joe-23-0218] [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: 07/07/2023] [Accepted: 11/23/2023] [Indexed: 11/25/2023]
Abstract
During pregnancy the maternal pancreatic islets of Langerhans undergo adaptive changes to compensate for gestational insulin resistance. The lactogenic hormones are well established to play a key role in regulating the islet adaptation to pregnancy, and one of the mechanisms through which they act is through upregulating β-cell serotonin production. During pregnancy islet serotonin levels are significantly elevated, where it is released from the β-cells to drive the adaptive response through paracrine and autocrine effects. We have previously shown that placental kisspeptin (KP) also plays a role in promoting the elevated insulin secretion and β-cell proliferation observed during pregnancy, although the precise mechanisms involved are unclear. In the present study we investigated the effects of KP on expression of pro-proliferative genes and serotonin biosynthesis within rodent islets. Whilst KP had limited effect on pro-proliferative gene expression at the time points tested, KP did significantly stimulate expression of the serotonin biosynthesis enzyme Tph-1. Furthermore, the islets of pregnant β-cell-specific GPR54 knockdown mice were found to contain significantly fewer serotonin-positive β-cells when compared to pregnant controls. Our previous studies suggested that reduced placental kisspeptin production, with consequent impaired kisspeptin-dependent β-cell compensation, may be a factor in the development of GDM in humans. These current data suggest that, similar to the lactogenic hormones, KP may also contribute to serotonin biosynthesis and subsequent islet signalling during pregnancy. Furthermore, upregulation of serotonin biosynthesis may represent a common mechanism through which multiple signals might influence the islet adaptation to pregnancy.
Collapse
Affiliation(s)
- Thomas G Hill
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, UK
| | - Lorna I F Smith
- Diabetes Research Group, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | | | - Peter M Jones
- Diabetes Research Group, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | - James E Bowe
- Diabetes Research Group, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| |
Collapse
|
12
|
Gojani EG, Wang B, Li DP, Kovalchuk O, Kovalchuk I. The Impact of Psilocybin on High Glucose/Lipid-Induced Changes in INS-1 Cell Viability and Dedifferentiation. Genes (Basel) 2024; 15:183. [PMID: 38397173 PMCID: PMC10888174 DOI: 10.3390/genes15020183] [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: 12/21/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Serotonin emerges as a pivotal factor influencing the growth and functionality of β-cells. Psilocybin, a natural compound derived from mushrooms of the Psilocybe genus, exerts agonistic effects on the serotonin 5-HT2A and 5-HT2B receptors, thereby mimicking serotonin's behavior. This study investigates the potential impacts of psilocybin on β-cell viability, dedifferentiation, and function using an in vitro system. The INS-1 832/13 Rat Insulinoma cell line underwent psilocybin pretreatment, followed by exposure to high glucose-high lipid (HG-HL) conditions for specific time periods. After being harvested from treated cells, total transcript and cellular protein were utilized for further investigation. Our findings implied that psilocybin administration effectively mitigates HG-HL-stimulated β-cell loss, potentially mediated through the modulation of apoptotic biomarkers, which is possibly related to the mitigation of TXNIP, STAT-1, and STAT-3 phosphorylation. Furthermore, psilocybin exhibits the capacity to modulate the expression of key genes associated with β-cell dedifferentiation, including Pou5f1 and Nanog, indicating its potential in attenuating β-cell dedifferentiation. This research lays the groundwork for further exploration into the therapeutic potential of psilocybin in Type II diabetes intervention.
Collapse
Affiliation(s)
| | | | | | | | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.); (B.W.); (D.-P.L.); (O.K.)
| |
Collapse
|
13
|
Schmidt MD, Ishahak M, Augsornworawat P, Millman JR. Comparative and integrative single cell analysis reveals new insights into the transcriptional immaturity of stem cell-derived β cells. BMC Genomics 2024; 25:105. [PMID: 38267908 PMCID: PMC10807170 DOI: 10.1186/s12864-024-10013-x] [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: 11/30/2023] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Abstract
Diabetes cell replacement therapy has the potential to be transformed by human pluripotent stem cell-derived β cells (SC-β cells). However, the precise identity of SC-β cells in relationship to primary fetal and adult β-cells remains unclear. Here, we used single-cell sequencing datasets to characterize the transcriptional identity of islets from in vitro differentiation, fetal islets, and adult islets. Our analysis revealed that SC-β cells share a core β-cell transcriptional identity with human adult and fetal β-cells, however SC-β cells possess a unique transcriptional profile characterized by the persistent expression and activation of progenitor and neural-biased gene networks. These networks are present in SC-β cells, irrespective of the derivation protocol used. Notably, fetal β-cells also exhibit this neural signature at the transcriptional level. Our findings offer insights into the transcriptional identity of SC-β cells and underscore the need for further investigation of the role of neural transcriptional networks in their development.
Collapse
Affiliation(s)
- Mason D Schmidt
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Punn Augsornworawat
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO, 63130, USA
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO, 63130, USA.
| |
Collapse
|
14
|
Roberts FL, Cataldo LR, Fex M. Monoamines' role in islet cell function and type 2 diabetes risk. Trends Mol Med 2023; 29:1045-1058. [PMID: 37722934 DOI: 10.1016/j.molmed.2023.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/20/2023]
Abstract
The two monoamines serotonin and melatonin have recently been highlighted as potent regulators of islet hormone secretion and overall glucose homeostasis in the body. In fact, dysregulated signaling of both amines are implicated in β-cell dysfunction and development of type 2 diabetes mellitus (T2DM). Serotonin is a key player in β-cell physiology and plays a role in expansion of β-cell mass. Melatonin regulates circadian rhythm and nutrient metabolism and reduces insulin release in human and rodent islets in vitro. Herein, we focus on the role of serotonin and melatonin in islet physiology and the pathophysiology of T2DM. This includes effects on hormone secretion, receptor expression, genetic variants influencing β-cell function, melatonin treatment, and compounds that alter serotonin availability and signaling.
Collapse
Affiliation(s)
- Fiona Louise Roberts
- Lund University Diabetes Centre, Department of Clinical Sciences, Unit for Molecular Metabolism, SE-21428 Malmö, Sweden
| | - Luis Rodrigo Cataldo
- Lund University Diabetes Centre, Department of Clinical Sciences, Unit for Molecular Metabolism, SE-21428 Malmö, Sweden; The Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Malin Fex
- Lund University Diabetes Centre, Department of Clinical Sciences, Unit for Molecular Metabolism, SE-21428 Malmö, Sweden.
| |
Collapse
|
15
|
Page L, Younge N, Freemark M. Hormonal Determinants of Growth and Weight Gain in the Human Fetus and Preterm Infant. Nutrients 2023; 15:4041. [PMID: 37764824 PMCID: PMC10537367 DOI: 10.3390/nu15184041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The factors controlling linear growth and weight gain in the human fetus and newborn infant are poorly understood. We review here the changes in linear growth, weight gain, lean body mass, and fat mass during mid- and late gestation and the early postnatal period in the context of changes in the secretion and action of maternal, placental, fetal, and neonatal hormones, growth factors, and adipocytokines. We assess the effects of hormonal determinants on placental nutrient delivery and the impact of preterm delivery on hormone expression and postnatal growth and metabolic function. We then discuss the effects of various maternal disorders and nutritional and pharmacologic interventions on fetal and perinatal hormone and growth factor production, growth, and fat deposition and consider important unresolved questions in the field.
Collapse
Affiliation(s)
- Laura Page
- Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Noelle Younge
- Neonatology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Michael Freemark
- Division of Pediatric Endocrinology, Duke University Medical Center, Durham, NC 27710, USA;
- The Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
16
|
Augsornworawat P, Hogrebe NJ, Ishahak M, Schmidt MD, Marquez E, Maestas MM, Veronese-Paniagua DA, Gale SE, Miller JR, Velazco-Cruz L, Millman JR. Single-nucleus multi-omics of human stem cell-derived islets identifies deficiencies in lineage specification. Nat Cell Biol 2023; 25:904-916. [PMID: 37188763 PMCID: PMC10264244 DOI: 10.1038/s41556-023-01150-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/17/2023] [Indexed: 05/17/2023]
Abstract
Insulin-producing β cells created from human pluripotent stem cells have potential as a therapy for insulin-dependent diabetes, but human pluripotent stem cell-derived islets (SC-islets) still differ from their in vivo counterparts. To better understand the state of cell types within SC-islets and identify lineage specification deficiencies, we used single-nucleus multi-omic sequencing to analyse chromatin accessibility and transcriptional profiles of SC-islets and primary human islets. Here we provide an analysis that enabled the derivation of gene lists and activity for identifying each SC-islet cell type compared with primary islets. Within SC-islets, we found that the difference between β cells and awry enterochromaffin-like cells is a gradient of cell states rather than a stark difference in identity. Furthermore, transplantation of SC-islets in vivo improved cellular identities overtime, while long-term in vitro culture did not. Collectively, our results highlight the importance of chromatin and transcriptional landscapes during islet cell specification and maturation.
Collapse
Affiliation(s)
- Punn Augsornworawat
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Mason D Schmidt
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Erica Marquez
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Marlie M Maestas
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Daniel A Veronese-Paniagua
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Sarah E Gale
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Julia R Miller
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Leonardo Velazco-Cruz
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
17
|
Zhu H, Wang G, Nguyen-Ngoc KV, Kim D, Miller M, Goss G, Kovsky J, Harrington AR, Saunders DC, Hopkirk AL, Melton R, Powers AC, Preissl S, Spagnoli FM, Gaulton KJ, Sander M. Understanding cell fate acquisition in stem-cell-derived pancreatic islets using single-cell multiome-inferred regulomes. Dev Cell 2023; 58:727-743.e11. [PMID: 37040771 PMCID: PMC10175223 DOI: 10.1016/j.devcel.2023.03.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 01/06/2023] [Accepted: 03/14/2023] [Indexed: 04/13/2023]
Abstract
Pancreatic islet cells derived from human pluripotent stem cells hold great promise for modeling and treating diabetes. Differences between stem-cell-derived and primary islets remain, but molecular insights to inform improvements are limited. Here, we acquire single-cell transcriptomes and accessible chromatin profiles during in vitro islet differentiation and pancreas from childhood and adult donors for comparison. We delineate major cell types, define their regulomes, and describe spatiotemporal gene regulatory relationships between transcription factors. CDX2 emerged as a regulator of enterochromaffin-like cells, which we show resemble a transient, previously unrecognized, serotonin-producing pre-β cell population in fetal pancreas, arguing against a proposed non-pancreatic origin. Furthermore, we observe insufficient activation of signal-dependent transcriptional programs during in vitro β cell maturation and identify sex hormones as drivers of β cell proliferation in childhood. Altogether, our analysis provides a comprehensive understanding of cell fate acquisition in stem-cell-derived islets and a framework for manipulating cell identities and maturity.
Collapse
Affiliation(s)
- Han Zhu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Gaowei Wang
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Kim-Vy Nguyen-Ngoc
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Dongsu Kim
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Michael Miller
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Georgina Goss
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Jenna Kovsky
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Austin R Harrington
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-0475, USA
| | - Alexander L Hopkirk
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-0475, USA
| | - Rebecca Melton
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Alvin C Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-0475, USA; Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-0615, USA; VA Tennessee Valley Healthcare System, Nashville, TN 37212-2637, USA
| | - Sebastian Preissl
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Francesca M Spagnoli
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Kyle J Gaulton
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Maike Sander
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0653, USA; Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
18
|
Sylvester-Armstrong KR, Reeder CF, Powell A, Becker MW, Hagan DW, Chen J, Mathews CE, Wasserfall CH, Atkinson MA, Egerman R, Phelps EA. Serum from pregnant donors induces human beta cell proliferation and insulin secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537214. [PMID: 37131658 PMCID: PMC10153135 DOI: 10.1101/2023.04.17.537214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Pancreatic beta cells are among the slowest replicating cells in the human body. Human beta cells usually do not increase in number with exceptions being during the neonatal period, in cases of obesity, and during pregnancy. This project explored maternal serum for stimulatory potential on human beta cell proliferation and insulin output. Gravid, full-term women who were scheduled to undergo cesarean delivery were recruited for this study. A human beta cell line was cultured in media supplemented with serum from pregnant and non-pregnant donors and assessed for differences in proliferation and insulin secretion. A subset of pregnant donor sera induced significant increases in beta cell proliferation and insulin secretion. Pooled serum from pregnant donors also increased proliferation in primary human beta cells but not primary human hepatocytes indicating a cell-type specific effect. This study suggests stimulatory factors in human serum during pregnancy could provide a novel approach for human beta cell expansion.
Collapse
Affiliation(s)
| | - Callie F. Reeder
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, Florida
| | - Andrece Powell
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida
| | - Matthew W. Becker
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida
| | - D. Walker Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida
| | - Jing Chen
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida
| | - Robert Egerman
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, Florida
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida
| |
Collapse
|
19
|
Type I Diabetes Pathoetiology and Pathophysiology: Roles of the Gut Microbiome, Pancreatic Cellular Interactions, and the 'Bystander' Activation of Memory CD8 + T Cells. Int J Mol Sci 2023; 24:ijms24043300. [PMID: 36834709 PMCID: PMC9964837 DOI: 10.3390/ijms24043300] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/10/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) arises from the failure of pancreatic β-cells to produce adequate insulin, usually as a consequence of extensive pancreatic β-cell destruction. T1DM is classed as an immune-mediated condition. However, the processes that drive pancreatic β-cell apoptosis remain to be determined, resulting in a failure to prevent ongoing cellular destruction. Alteration in mitochondrial function is clearly the major pathophysiological process underpinning pancreatic β-cell loss in T1DM. As with many medical conditions, there is a growing interest in T1DM as to the role of the gut microbiome, including the interactions of gut bacteria with Candida albicans fungal infection. Gut dysbiosis and gut permeability are intimately associated with raised levels of circulating lipopolysaccharide and suppressed butyrate levels, which can act to dysregulate immune responses and systemic mitochondrial function. This manuscript reviews broad bodies of data on T1DM pathophysiology, highlighting the importance of alterations in the mitochondrial melatonergic pathway of pancreatic β-cells in driving mitochondrial dysfunction. The suppression of mitochondrial melatonin makes pancreatic β-cells susceptible to oxidative stress and dysfunctional mitophagy, partly mediated by the loss of melatonin's induction of PTEN-induced kinase 1 (PINK1), thereby suppressing mitophagy and increasing autoimmune associated major histocompatibility complex (MHC)-1. The immediate precursor to melatonin, N-acetylserotonin (NAS), is a brain-derived neurotrophic factor (BDNF) mimic, via the activation of the BDNF receptor, TrkB. As both the full-length and truncated TrkB play powerful roles in pancreatic β-cell function and survival, NAS is another important aspect of the melatonergic pathway relevant to pancreatic β-cell destruction in T1DM. The incorporation of the mitochondrial melatonergic pathway in T1DM pathophysiology integrates wide bodies of previously disparate data on pancreatic intercellular processes. The suppression of Akkermansia muciniphila, Lactobacillus johnsonii, butyrate, and the shikimate pathway-including by bacteriophages-contributes to not only pancreatic β-cell apoptosis, but also to the bystander activation of CD8+ T cells, which increases their effector function and prevents their deselection in the thymus. The gut microbiome is therefore a significant determinant of the mitochondrial dysfunction driving pancreatic β-cell loss as well as 'autoimmune' effects derived from cytotoxic CD8+ T cells. This has significant future research and treatment implications.
Collapse
|
20
|
Application of Caenorhabditis elegans in Lipid Metabolism Research. Int J Mol Sci 2023; 24:ijms24021173. [PMID: 36674689 PMCID: PMC9860639 DOI: 10.3390/ijms24021173] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Over the last decade, the development and prevalence of obesity have posed a serious public health risk, which has prompted studies on the regulation of adiposity. With the ease of genetic manipulation, the diversity of the methods for characterizing body fat levels, and the observability of feeding behavior, Caenorhabditis elegans (C. elegans) is considered an excellent model for exploring energy homeostasis and the regulation of the cellular fat storage. In addition, the homology with mammals in the genes related to the lipid metabolism allows many aspects of lipid modulation by the regulators of the central nervous system to be conserved in this ideal model organism. In recent years, as the complex network of genes that maintain an energy balance has been gradually expanded and refined, the regulatory mechanisms of lipid storage have become clearer. Furthermore, the development of methods and devices to assess the lipid levels has become a powerful tool for studies in lipid droplet biology and the regulation of the nematode lipid metabolism. Herein, based on the rapid progress of C. elegans lipid metabolism-related studies, this review outlined the lipid metabolic processes, the major signaling pathways of fat storage regulation, and the primary experimental methods to assess the lipid content in nematodes. Therefore, this model system holds great promise for facilitating the understanding, management, and therapies of human obesity and other metabolism-related diseases.
Collapse
|
21
|
Guo Y, Li L, Yao Y, Li H. Regeneration of Pancreatic β-Cells for Diabetes Therapeutics by Natural DYRK1A Inhibitors. Metabolites 2022; 13:metabo13010051. [PMID: 36676976 PMCID: PMC9865674 DOI: 10.3390/metabo13010051] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/07/2022] [Accepted: 11/23/2022] [Indexed: 12/31/2022] Open
Abstract
The pathogenesis of diabetes mellitus is characterized by insulin resistance and islet β-cell dysfunction. Up to now, the focus of diabetes treatment has been to control blood glucose to prevent diabetic complications. There is an urgent need to develop a therapeutic approach to restore the mass and function of β-cells. Although exogenous islet cell transplantation has been used to help patients control blood glucose, it is costly and has very narrow application scenario. So far, small molecules have been reported to stimulate β-cell proliferation and expand β-cell mass, increasing insulin secretion. Dual-specificity tyrosine-regulated kinase 1A (DYRK1A) inhibitors can induce human β-cell proliferation in vitro and in vivo, and show great potential in the field of diabetes therapeutics. From this perspective, we elaborated on the mechanism by which DYRK1A inhibitors regulate the proliferation of pancreatic β-cells, and summarized several effective natural DYRK1A inhibitors, hoping to provide clues for subsequent structural optimization and drug development in the future.
Collapse
Affiliation(s)
- Yichuan Guo
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lingqiao Li
- Zhejiang Starry Pharmaceutical Co., Ltd., Taizhou 317306, China
| | - Yuanfa Yao
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (Y.Y.); (H.L.)
| | - Hanbing Li
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (Y.Y.); (H.L.)
| |
Collapse
|
22
|
Castell AL, Goubault C, Ethier M, Fergusson G, Tremblay C, Baltz M, Dal Soglio D, Ghislain J, Poitout V. β Cell mass expansion during puberty involves serotonin signaling and determines glucose homeostasis in adulthood. JCI Insight 2022; 7:160854. [PMID: 36107617 PMCID: PMC9675460 DOI: 10.1172/jci.insight.160854] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/14/2022] [Indexed: 01/07/2023] Open
Abstract
Puberty is associated with transient insulin resistance that normally recedes at the end of puberty; however, in overweight children, insulin resistance persists, leading to an increased risk of type 2 diabetes. The mechanisms whereby pancreatic β cells adapt to pubertal insulin resistance, and how they are affected by the metabolic status, have not been investigated. Here, we show that puberty is associated with a transient increase in β cell proliferation in rats and humans of both sexes. In rats, β cell proliferation correlated with a rise in growth hormone (GH) levels. Serum from pubertal rats and humans promoted β cell proliferation, suggesting the implication of a circulating factor. In pubertal rat islets, expression of genes of the GH/serotonin (5-hydroxytryptamine [5-HT]) pathway underwent changes consistent with a proliferative effect. Inhibition of the pro-proliferative 5-HT receptor isoform HTR2B blocked the increase in β cell proliferation in pubertal islets ex vivo and in vivo. Peripubertal metabolic stress blunted β cell proliferation during puberty and led to altered glucose homeostasis later in life. This study identifies a role of GH/GH receptor/5-HT/HTR2B signaling in the control of β cell mass expansion during puberty and identifies a mechanistic link between pubertal obesity and the risk of developing type 2 diabetes.
Collapse
Affiliation(s)
- Anne-Laure Castell
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine and
| | - Clara Goubault
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, University of Montreal, Quebec, Canada
| | - Mélanie Ethier
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Grace Fergusson
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Caroline Tremblay
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marie Baltz
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Dorothée Dal Soglio
- CHU Sainte-Justine, Montreal, Quebec, Canada.,Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de recherche du centre hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine and
| |
Collapse
|
23
|
Barra NG, Kwon YH, Morrison KM, Steinberg GR, Wade MG, Khan WI, Vijayan MM, Schertzer JD, Holloway AC. Increased gut serotonin production in response to bisphenol A structural analogs may contribute to their obesogenic effects. Am J Physiol Endocrinol Metab 2022; 323:E80-E091. [PMID: 35575233 DOI: 10.1152/ajpendo.00049.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesogens are synthetic, environmental chemicals that can disrupt endocrine control of metabolism and contribute to the risk of obesity and metabolic disease. Bisphenol A (BPA) is one of the most studied obesogens. There is considerable evidence that BPA exposure is associated with weight gain, increased adiposity, poor blood glucose control, and nonalcoholic fatty liver disease in animal models and human populations. Increased usage of structural analogs of BPA has occurred in response to legislation banning their use in some commercial products. However, BPA analogs may also cause some of the same metabolic impairments because of common mechanisms of action. One key effector that is altered by BPA and its analogs is serotonin, however, it is unknown if BPA-induced changes in peripheral serotonin pathways underlie metabolic perturbations seen with BPA exposure. Upon ingestion, BPA and its analogs act as endocrine-disrupting chemicals in the gastrointestinal tract to influence serotonin production by the gut, where over 95% of serotonin is produced. The purpose of this review is to evaluate how BPA and its analogs alter gut serotonin regulation and then discuss how disruption of serotonergic networks influences host metabolism. We also provide evidence that BPA and its analogs enhance serotonin production in gut enterochromaffin cells. Taken together, we propose that BPA and many BPA analogs represent endocrine-disrupting chemicals that can influence host metabolism through the endogenous production of gut-derived factors, such as serotonin.
Collapse
Affiliation(s)
- Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Yun Han Kwon
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Wade
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Alison C Holloway
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
24
|
Moon JH, Oh CM, Kim H. Serotonin in the regulation of systemic energy metabolism. J Diabetes Investig 2022; 13:1639-1645. [PMID: 35762288 PMCID: PMC9533050 DOI: 10.1111/jdi.13879] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 11/29/2022] Open
Abstract
Serotonin is a well‐known neurotransmitter that is synthesized from the amino acid, tryptophan. To date, more than 14 different serotonin receptors have been discovered; they exist universally in our body and enable diverse biological functions in different organs. Central serotonin regulates mood and behavior, and impacts the systemic energy balance by decreasing appetite. A number of drugs that modulate central serotonin function (e.g., fenfluramine, sibutramine and lorcaserin) were approved and used as anti‐obesity drugs, but then later withdrawn due to adverse cardiovascular and carcinogenic effects. Over the past decade, the role of peripheral serotonin in regulating systemic energy metabolism has been extensively explored using tissue‐specific knockout animal models. By inhibiting the action of serotonin in liver and adipose tissues, hepatic steatosis was improved and lipid accumulation was mitigated, respectively. Recent findings show that modulation of the serotonergic system is a promising therapeutic target for metabolic diseases. This review summarizes the role of serotonin in regulating energy metabolism in different organs, and discusses the potential of serotonin modulation for treating metabolic diseases.
Collapse
Affiliation(s)
- Joon Ho Moon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea
| |
Collapse
|
25
|
Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, Pastori RL, Russ HA, Wagner BK, Dominguez-Bendala J. Emerging diabetes therapies: Bringing back the β-cells. Mol Metab 2022; 60:101477. [PMID: 35331962 PMCID: PMC8987999 DOI: 10.1016/j.molmet.2022.101477] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Stem cell therapies are finally coming of age as a viable alternative to pancreatic islet transplantation for the treatment of insulin-dependent diabetes. Several clinical trials using human embryonic stem cell (hESC)-derived β-like cells are currently underway, with encouraging preliminary results. Remaining challenges notwithstanding, these strategies are widely expected to reduce our reliance on human isolated islets for transplantation procedures, making cell therapies available to millions of diabetic patients. At the same time, advances in our understanding of pancreatic cell plasticity and the molecular mechanisms behind β-cell replication and regeneration have spawned a multitude of translational efforts aimed at inducing β-cell replenishment in situ through pharmacological means, thus circumventing the need for transplantation. SCOPE OF REVIEW We discuss here the current state of the art in hESC transplantation, as well as the parallel quest to discover agents capable of either preserving the residual mass of β-cells or inducing their proliferation, transdifferentiation or differentiation from progenitor cells. MAJOR CONCLUSIONS Stem cell-based replacement therapies in the mold of islet transplantation are already around the corner, but a permanent cure for type 1 diabetes will likely require the endogenous regeneration of β-cells aided by interventions to restore the immune balance. The promise of current research avenues and a strong pipeline of clinical trials designed to tackle these challenges bode well for the realization of this goal.
Collapse
Affiliation(s)
- G Basile
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - M M F Qadir
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - F Mauvais-Jarvis
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - A Vetere
- Broad Institute, Cambridge, MA, USA
| | - V Shoba
- Broad Institute, Cambridge, MA, USA
| | | | - R L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H A Russ
- Barbara Davis Center for Diabetes, Colorado University Anschutz Medical Campus, Aurora, CO, USA.
| | | | - J Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
26
|
Resendes C, Horan DJ, Robling AG, Gao B, Milne GL, Warman ML. Transiently increased serotonin has modest or no effects on bone mass accrual in growing female C57BL6/J or growing male and female Lrp5 A214V mice. Bone 2022; 158:116307. [PMID: 34973493 DOI: 10.1016/j.bone.2021.116307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022]
Abstract
Serotonin (5HT) is a chemical messenger with biologic activities affecting multiple organs. Within the skeletal system, studies in mice and humans suggest blood 5HT levels affect bone, with elevations impairing and reductions enhancing bone accrual. Other studies, however, have not supported this hypothesis. Recently, administering 5HT to a Piezo1 mutant mouse strain with hyposerotonemia, intestinal dysmotility, and a doubling of femoral trabecular bone mass at 2 months of age, was reported to return the animals' intestinal motility and bone mass to normal. However, whether the 5HT directly affected bone metabolism or indirectly affected metabolism by improving intestinal function was not determined. Therefore, we administered 5HT to mice with normal intestinal function. We randomized female C57BL6/J mice and male and female mice that have increased bone mass due to a missense mutation in the WNT co-receptor LRP5 (Lrp5A214V) to receive 5HT or vehicle via daily IP injection from 4 weeks to 8 weeks of age. We did not observe consistent significant changes for distal femur trabecular, midshaft femur cortical, or vertebral body trabecular bone mass between 5HT treated and vehicle treated mice of either genotype. These data are compatible with other studies that have not observed a correlation between blood 5HT level and bone mass.
Collapse
Affiliation(s)
- Caitlin Resendes
- Orthopedic Research Laboratories, Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA, United States; Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Daniel J Horan
- Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Benlian Gao
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| | - Ginger L Milne
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| | - Matthew L Warman
- Orthopedic Research Laboratories, Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA, United States; Department of Genetics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
27
|
Grajales D, Vázquez P, Ruíz-Rosario M, Tudurí E, Mirasierra M, Ferreira V, Hitos AB, Koller D, Zubiaur P, Cigudosa JC, Abad-Santos F, Vallejo M, Quesada I, Tirosh B, Leibowitz G, Valverde ÁM. The second-generation antipsychotic drug aripiprazole modulates the serotonergic system in pancreatic islets and induces beta cell dysfunction in female mice. Diabetologia 2022; 65:490-505. [PMID: 34932133 PMCID: PMC8803721 DOI: 10.1007/s00125-021-05630-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Second-generation antipsychotic (SGA) drugs have been associated with the development of type 2 diabetes and the metabolic syndrome in patients with schizophrenia. In this study, we aimed to investigate the effects of two different SGA drugs, olanzapine and aripiprazole, on metabolic state and islet function and plasticity. METHODS We analysed the functional adaptation of beta cells in 12-week-old B6;129 female mice fed an olanzapine- or aripiprazole-supplemented diet (5.5-6.0 mg kg-1 day-1) for 6 months. Glucose and insulin tolerance tests, in vivo glucose-stimulated insulin secretion and indirect calorimetry were performed at the end of the study. The effects of SGAs on beta cell plasticity and islet serotonin levels were assessed by transcriptomic analysis and immunofluorescence. Insulin secretion was assessed by static incubations and Ca2+ fluxes by imaging techniques. RESULTS Treatment of female mice with olanzapine or aripiprazole for 6 months induced weight gain (p<0.01 and p<0.05, respectively), glucose intolerance (p<0.01) and impaired insulin secretion (p<0.05) vs mice fed a control chow diet. Aripiprazole, but not olanzapine, induced serotonin production in beta cells vs controls, likely by increasing tryptophan hydroxylase 1 (TPH1) expression, and inhibited Ca2+ flux. Of note, aripiprazole increased beta cell size (p<0.05) and mass (p<0.01) vs mice fed a control chow diet, along with activation of mechanistic target of rapamycin complex 1 (mTORC1)/S6 signalling, without preventing beta cell dysfunction. CONCLUSIONS/INTERPRETATION Both SGAs induced weight gain and beta cell dysfunction, leading to glucose intolerance; however, aripiprazole had a more potent effect in terms of metabolic alterations, which was likely a result of its ability to modulate the serotonergic system. The deleterious metabolic effects of SGAs on islet function should be considered while treating patients as these drugs may increase the risk for development of the metabolic syndrome and diabetes.
Collapse
Affiliation(s)
- Diana Grajales
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Vázquez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Eva Tudurí
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
| | - Mercedes Mirasierra
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Vítor Ferreira
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana B Hitos
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Dora Koller
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | | | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Mario Vallejo
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Iván Quesada
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
| | - Boaz Tirosh
- The Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gil Leibowitz
- Endocrinology and Metabolism Service, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
28
|
Wang QJ, Guo Y, Zhang KH, Zhang L, Geng SX, Shan CH, Liu P, Zhu MQ, Jin QY, Liu ZY, Wang MZ, Li MY, Liu M, An L, Tian JH, Wu ZH. Night-Restricted Feeding Improves Gut Health by Synchronizing Microbe-Driven Serotonin Rhythm and Eating Activity-Driven Body Temperature Oscillations in Growing Rabbits. Front Cell Infect Microbiol 2022; 11:771088. [PMID: 34976857 PMCID: PMC8718905 DOI: 10.3389/fcimb.2021.771088] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/02/2021] [Indexed: 01/04/2023] Open
Abstract
The circadian misalignment of the gut microbiota caused by unusual eating times in adult animals is related to disease development. However, whether the composition and diurnal rhythm of gut microbiota can be optimized by synchronizing the window period of eating with natural eating habits to reduce the risk of diarrhea remains unclear, especially in growing animals. In this study, 108 5-week-old weaned rabbits (nocturnal animals) were randomly subjected to daytime feeding (DF) and night-restricted feeding (NRF). At age 12 weeks, six rabbits were selected from each group, and caecum and cecal contents, as well as serum samples were collected at 4-h intervals during 24 h. Overall, NRF was found to reduce the risk of diarrhea in growing rabbits, improved the diurnal rhythm and abundance of beneficial microorganisms, along with the production of beneficial metabolites, whereas reduced the abundance of potential pathogens (Synergistes, Desulfovibrio, and Alistipes). Moreover, NRF improved diurnal rhythm of tryptophan hydroxylase isoform 1 and serotonin. Furthermore, NRF strengthened the diurnal amplitude of body core temperature, and promoted the diurnal expression of intestinal clock genes (BMAL1, CLOCK, REV-ERBα, and PER1), and genes related to the regulation of the intestinal barrier (CLAUDIN-1), and intestinal epithelial cell self-proliferation and renewal (BMI1). In vitro simulation experiments further revealed that synchronization of microbial-driven serotonin rhythm and eating activity-driven body temperature oscillations, which are important zeitgebers, could promote the diurnal expression of clock genes and CLAUDIN-1 in rabbit intestinal epithelial cells (RIEC), and enhance RIEC proliferation. This is the first study to reveal that NRF reprograms the diurnal rhythm of the gut microbiome, promotes the diurnal expression of clock genes and tight junction genes via synchronization of microbial-driven serotonin rhythm and eating activity-driven body temperature oscillations, thereby improving intestinal health and reducing the risk of diarrhea in growing rabbits. Collectively, these results provide a new perspective for the healthy feeding and management of growing animals.
Collapse
Affiliation(s)
- Qiang-Jun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yao Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ke-Hao Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lei Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shi-Xia Geng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chun-Hua Shan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Peng Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Meng-Qi Zhu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiong-Yu Jin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhong-Ying Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mei-Zhi Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ming-Yong Li
- National Rabbit Industry Technology System Qingdao Comprehensive Experimental Station, Qingdao, China
| | - Man Liu
- National Rabbit Industry Technology System Qingdao Comprehensive Experimental Station, Qingdao, China
| | - Lei An
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jian-Hui Tian
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhong-Hong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
29
|
Kurian J, Mohanthy S, Nanjumdaiah RM. Mechanism of action of yoga on prevention and management of type 2 diabetes mellitus: Narrative review. J Bodyw Mov Ther 2022; 29:134-139. [DOI: 10.1016/j.jbmt.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/23/2021] [Accepted: 10/17/2021] [Indexed: 10/20/2022]
|
30
|
Park J, Jeong W, Yun C, Kim H, Oh CM. Serotonergic Regulation of Hepatic Energy Metabolism. Endocrinol Metab (Seoul) 2021; 36:1151-1160. [PMID: 34911172 PMCID: PMC8743581 DOI: 10.3803/enm.2021.1331] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/25/2021] [Indexed: 01/01/2023] Open
Abstract
The liver is a vital organ that regulates systemic energy metabolism and many physiological functions. Nonalcoholic fatty liver disease (NAFLD) is the commonest cause of chronic liver disease and end-stage liver failure. NAFLD is primarily caused by metabolic disruption of lipid and glucose homeostasis. Serotonin (5-hydroxytryptamine [5-HT]) is a biogenic amine with several functions in both the central and peripheral systems. 5-HT functions as a neurotransmitter in the brain and a hormone in peripheral tissues to regulate systemic energy homeostasis. Several recent studies have proposed various roles of 5-HT in hepatic metabolism and inflammation using tissue-specific knockout mice and 5-HT-receptor agonists/antagonists. This review compiles the most recent research on the relationship between 5-HT and hepatic metabolism, and the role of 5-HT signaling as a potential therapeutic target in NAFLD.
Collapse
Affiliation(s)
- Jiwon Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| | - Wooju Jeong
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| | - Chahyeon Yun
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon,
Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju,
Korea
| |
Collapse
|
31
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
32
|
Liang Y, Li H, Gan Y, Tu H. Shedding Light on the Role of Neurotransmitters in the Microenvironment of Pancreatic Cancer. Front Cell Dev Biol 2021; 9:688953. [PMID: 34395421 PMCID: PMC8363299 DOI: 10.3389/fcell.2021.688953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/13/2021] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy with a 5-year survival rate of less than 8%. The fate of PC is determined not only by the malignant behavior of the cancer cells, but also by the surrounding tumor microenvironment (TME), consisting of various cellular (cancer cells, immune cells, stromal cells, endothelial cells, and neurons) and non-cellular (cytokines, neurotransmitters, and extracellular matrix) components. The pancreatic TME has the unique characteristic of exhibiting increased neural density and altered microenvironmental concentration of neurotransmitters. The neurotransmitters, produced by both neuron and non-neuronal cells, can directly regulate the biological behavior of PC cells via binding to their corresponding receptors on tumor cells and activating the intracellular downstream signals. On the other hand, the neurotransmitters can also communicate with other cellular components such as the immune cells in the TME to promote cancer growth. In this review, we will summarize the pleiotropic effects of neurotransmitters on the initiation and progression of PC, and particularly discuss the emerging mechanisms of how neurotransmitters influence the innate and adaptive immune responses in the TME in an autocrine or paracrine manner. A better understanding of the interplay between neurotransmitters and the immune cells in the TME might facilitate the development of new effective therapies for PC.
Collapse
Affiliation(s)
| | | | - Yu Gan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Gu P, Lin Y, Wan Q, Su D, Shu Q. Oxytocin signal contributes to the adaptative growth of islets during gestation. Endocr Connect 2021; 10:694-706. [PMID: 34077390 PMCID: PMC8240721 DOI: 10.1530/ec-21-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/02/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Increased insulin production and secretion by pancreatic β-cells are important for ensuring the high insulin demand during gestation. However, the underlying mechanism of β-cell adaptation during gestation or gestational diabetes mellitus (GDM) remains unclear. Oxytocin is an important physiological hormone in gestation and delivery, and it also contributes to the maintenance of β-cell function. The aim of this study was to investigate the role of oxytocin in β-cell adaptation during pregnancy. METHODS The relationship between the blood oxytocin level and pancreatic β-cell function in patients with GDM and healthy pregnant women was investigated. Gestating and non-gestating mice were used to evaluate the in vivo effect of oxytocin signal on β-cells during pregnancy. In vitro experiments were performed on INS-1 insulinoma cells. RESULTS The blood oxytocin levels were lower in patients with GDM than in healthy pregnant women and were associated with impaired pancreatic β-cell function. Acute administration of oxytocin increased insulin secretion in both gestating and non-gestating mice. A 3-week oxytocin treatment promoted the proliferation of pancreatic β-cells and increased the β-cell mass in gestating but not non-gestating mice. Antagonism of oxytocin receptors by atosiban impaired insulin secretion and induced GDM in gestating but not non-gestating mice. Oxytocin enhanced glucose-stimulated insulin secretion, activated the mitogen-activated protein kinase pathway, and promoted cell proliferation in INS-1 cells. CONCLUSIONS These findings provide strong evidence that oxytocin is needed for β-cell adaptation during pregnancy to maintain β-cell function, and the lack of oxytocin could be associated with the risk of GDM.
Collapse
Affiliation(s)
- Ping Gu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuege Lin
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Qi Wan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Correspondence should be addressed to Q Shu or D Su or Q Wan: or or
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, China
- Correspondence should be addressed to Q Shu or D Su or Q Wan: or or
| | - Qun Shu
- Department of Obstetrics, Shanghai First Maternity and Infant Health Hospital, School of Medicine, Tongji University, Shanghai, China
- Correspondence should be addressed to Q Shu or D Su or Q Wan: or or
| |
Collapse
|
34
|
Obesity-induced changes in human islet G protein-coupled receptor expression: Implications for metabolic regulation. Pharmacol Ther 2021; 228:107928. [PMID: 34174278 DOI: 10.1016/j.pharmthera.2021.107928] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022]
Abstract
G protein-coupled receptors (GPCRs) are a large family of cell surface receptors that are the targets for many different classes of pharmacotherapy. The islets of Langerhans are central to appropriate glucose homeostasis through their secretion of insulin, and islet function can be modified by ligands acting at the large number of GPCRs that islets express. The human islet GPCRome is not a static entity, but one that is altered under pathophysiological conditions and, in this review, we have compared expression of GPCR mRNAs in human islets obtained from normal weight range donors, and those with a weight range classified as obese. We have also considered the likely outcomes on islet function that the altered GPCR expression status confers and the possible impact that adipokines, secreted from expanded fat depots, could have at those GPCRs showing altered expression in obesity.
Collapse
|
35
|
Moon JH, Kim H, Kim H, Park J, Choi W, Choi W, Hong HJ, Ro HJ, Jun S, Choi SH, Banerjee RR, Shong M, Cho NH, Kim SK, German MS, Jang HC, Kim H. Lactation improves pancreatic β cell mass and function through serotonin production. Sci Transl Med 2021; 12:12/541/eaay0455. [PMID: 32350130 DOI: 10.1126/scitranslmed.aay0455] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 12/23/2019] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
Pregnancy imposes a substantial metabolic burden on women through weight gain and insulin resistance. Lactation reduces the risk of maternal postpartum diabetes, but the mechanisms underlying this benefit are unknown. Here, we identified long-term beneficial effects of lactation on β cell function, which last for years after the cessation of lactation. We analyzed metabolic phenotypes including β cell characteristics in lactating and non-lactating humans and mice. Lactating and non-lactating women showed comparable glucose tolerance at 2 months after delivery, but after a mean of 3.6 years, glucose tolerance in lactated women had improved compared to non-lactated women. In humans, the disposition index, a measure of insulin secretory function of β cells considering the degree of insulin sensitivity, was higher in lactated women at 3.6 years after delivery. In mice, lactation improved glucose tolerance and increased β cell mass at 3 weeks after delivery. Amelioration of glucose tolerance and insulin secretion were maintained up to 4 months after delivery in lactated mice. During lactation, prolactin induced serotonin production in β cells. Secreted serotonin stimulated β cell proliferation through serotonin receptor 2B in an autocrine and paracrine manner. In addition, intracellular serotonin acted as an antioxidant to mitigate oxidative stress and improved β cell survival. Together, our results suggest that serotonin mediates the long-term beneficial effects of lactation on female metabolic health by increasing β cell proliferation and reducing oxidative stress in β cells.
Collapse
Affiliation(s)
- Joon Ho Moon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyeongseok Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.,Department of Biochemistry, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Hyunki Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Jungsun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Wonsuk Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Wongun Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Hyun Jung Hong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Hyun-Joo Ro
- Center for Research Equipment, Korea Basic Science Institute, Cheongju 28119, Korea.,Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
| | - Sangmi Jun
- Center for Research Equipment, Korea Basic Science Institute, Cheongju 28119, Korea.,Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ronadip R Banerjee
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama School of Medicine, Birmingham, AL 35294, USA
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Nam Han Cho
- Department of Preventive Medicine, Ajou University School of Medicine, Suwon 16499, Korea
| | - Seung K Kim
- Department of Developmental Biology and Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael S German
- Diabetes Center, Hormone Research Institute and Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Hak Chul Jang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| |
Collapse
|
36
|
De Felice E, Giaquinto D, Damiano S, Salzano A, Fabroni S, Ciarcia R, Scocco P, de Girolamo P, D’Angelo L. Distinct Pattern of NPY in Gastro-Entero-Pancreatic System of Goat Kids Fed with a New Standardized Red Orange and Lemon Extract (RLE). Animals (Basel) 2021; 11:ani11020449. [PMID: 33572145 PMCID: PMC7914828 DOI: 10.3390/ani11020449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary In the last decades the European ban towards antibiotics resulted in an increase of the number of studies on the effects of natural feed additives, that can enhance the health of farm animals intended for human consumption. Polyphenols such as flavanones and anthocyanins (responsible of the red, purple or blue colors) are bioactive compounds found in fruits and vegetables. Polyphenols possess multiple pharmacological characteristics, like antioxidant, anti-inflammatory and immunostimulant properties. Although many of the biological effects of polyphenols are known, only a limited number of studies has been focused on the effects of their supplementation in ruminant diet. Therefore, we evaluated the effect of a diet supplemented with a standardized powder extract, red (blood) orange and lemon extract (RLE), rich in flavanones, anthocyanins and other polyphenols on the neuropeptide Y (NPY) distribution in the gastro–entero–pancreatic system of goat kids. In mammals, NPY occurs in both the central and peripheral nervous systems and it is involved in the control of different physiological processes, including food intake regulation. For the first time, we document that NPY is widely distributed in the abomasum, duodenum and pancreas of goat kids and that significantly increases in the abomasum and pancreas of RLE supplemented feed animals. Abstract The use of natural compounds as feed additive is also increasing in farm animals, thanks to the beneficial effect on both animals and consumers health. Here, we questioned whether natural extracts, such as red orange and lemon extract (RLE) rich in flavanones, anthocyanins, and other polyphenols, used as feed additives could display an effect on the neuropeptide Y (NPY) in the gastro–entero–pancreatic tract of goat kids. NPY is one of the most abundant neuropeptides in mammals, known for its orexigenic role although it is involved in many central and peripheral functions. We carried out immunohistochemical analyses on samples of abomasum, duodenum and pancreas collected from two experimental groups: one fed with standard diet and one with standard diet + RLE. For the first time we document NPY distribution in the abomasum, duodenum and pancreas of goats and observe the highest number of NPY positive cells in neuroendocrine cells of duodenum. Remarkably, upon RLE feed supplementation, NPY immunoreactive cells increased significantly in abomasal epithelium and pancreatic islets but not in duodenum, likely due to pH variation of abomasum and duodenum. Our observations represent a baseline for future studies on the interaction between neuropeptides and polyphenols, used as feed additive.
Collapse
Affiliation(s)
- Elena De Felice
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Pontoni 5, 62032 Camerino, Italy; (E.D.F.); (D.G.); (P.S.)
| | - Daniela Giaquinto
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Pontoni 5, 62032 Camerino, Italy; (E.D.F.); (D.G.); (P.S.)
| | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (A.S.); (R.C.); (L.D.)
| | - Angela Salzano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (A.S.); (R.C.); (L.D.)
| | - Simona Fabroni
- Research Centre for Olive, Fruit and Citrus Crops, Council for Agricultural Research and Economics (CREA), 95024 Acireale, Italy;
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (A.S.); (R.C.); (L.D.)
| | - Paola Scocco
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Pontoni 5, 62032 Camerino, Italy; (E.D.F.); (D.G.); (P.S.)
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (A.S.); (R.C.); (L.D.)
- Correspondence:
| | - Livia D’Angelo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (A.S.); (R.C.); (L.D.)
| |
Collapse
|
37
|
Abstract
Pancreatic beta cells are the only cell type in our body capable of producing and secreting insulin to instruct the insulin-sensitive cells and tissues of our bodies to absorb nutrients after a meal. Accurate control of insulin release is of critical importance; too little insulin leads to diabetes, while an excess of insulin can cause potentially fatal hypoglycaemia. Yet, the pancreas of most people will control insulin secretion safely and effectively over decades and in response to glucose excursions driven by tens of thousands of meals. Because we only become aware of the important contributions of the pancreas when it fails to maintain glucose homeostasis, it is easy to forget just how well insulin release from a healthy pancreas is matched to insulin need to ensure stable blood glucose levels. Beta cells achieve this feat by extensive crosstalk with the rest of the endocrine cell types in the islet, notably the glucagon-producing alpha cells and somatostatin-producing delta cells. Here I will review the important paracrine contributions that each of these cells makes to the stimulation and subsequent inhibition of insulin release in response to a transient nutrient stimulation, and make the case that a breakdown of this local crosstalk contributes to the pathophysiology of diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, 196 Briggs Hall, 1 Shields Avenue, Davis, CA, 95616, USA.
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
38
|
Yang B, Covington BA, Chen W. In vivo generation and regeneration of β cells in zebrafish. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:9. [PMID: 32613468 PMCID: PMC7329966 DOI: 10.1186/s13619-020-00052-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
Abstract
The pathological feature of diabetes, hyperglycemia, is a result of an inadequate number and/or function of insulin producing β cells. Replenishing functional β cells is a strategy to cure the disease. Although β-cell regeneration occurs in animal models under certain conditions, human β cells are refractory to proliferation. A better understanding of both the positive and the negative regulatory mechanisms of β-cell regeneration in animal models is essential to develop novel strategies capable of inducing functional β cells in patients. Zebrafish are an attractive model system for studying β-cell regeneration due to the ease to which genetic and chemical-genetic approaches can be used as well as their high regenerative capacity. Here, we highlight the current state of β-cell regeneration studies in zebrafish with an emphasis on cell signaling mechanisms.
Collapse
Affiliation(s)
- Bingyuan Yang
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN, 37232, USA
| | - Brittney A Covington
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN, 37232, USA
| | - Wenbiao Chen
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN, 37232, USA.
| |
Collapse
|