1
|
Bao K, Berger J, Na E, Su Q, Halasz G, Sleeman M, Okamoto H. Hepatic PKA Mediates Liver and Pancreatic α-Cell Cross Talk. Diabetes 2025; 74:885-897. [PMID: 40095004 PMCID: PMC12097458 DOI: 10.2337/db24-0958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Glucagon stimulates hepatic glucose production, in part by promoting the uptake and catabolism of amino acids. Inhibition of the liver glucagon receptor (GCGR) results in elevated plasma amino acids, which triggers the proliferation of pancreatic α-cells, forming a liver-α-cell loop. This study aims to delineate hepatic signaling molecules downstream of GCGR that mediate the liver-α-cell loop. We knocked down liver GCGR, its G-coupled protein GNAS, and two GNAS downstream effectors, PKA and EPAC2 (RAPGEF4). Mice with GCGR, GNAS, and PKA knockdown had similar suppression of hepatic amino acid catabolism genes, hyperaminoacidemia, and α-cell hyperplasia, but those with EPAC2 knockdown did not. We then demonstrated that activating liver PKA was sufficient to reverse hyperaminoacidemia and α-cell hyperplasia caused by GCGR blockade. These results suggest that liver GCGR signals through PKA to control amino acid metabolism and that hepatic PKA plays a critical role in the liver-α-cell loop. ARTICLE HIGHLIGHTS A liver-α-cell loop exists, where inhibition of the liver glucagon receptor (GCGR) causes hyperaminoacidemia and pancreatic α-cell hyperplasia, but the GCGR downstream factors responsible for these effects are not clear. We silenced GCGR, its G-coupled protein GNAS, and two GNAS downstream effectors, PKA and EPAC2, to assess their effects on the liver-α-cell loop. Inhibition of the GCGR-GNAS-PKA pathway suppresses amino acid catabolism and causes α-cell hyperplasia, whereas PKA activation promotes amino acid catabolism and reduces alpha cell mass even when GCGR is blocked. Our study establishes hepatic PKA as the critical regulator of the liver-α-cell loop.
Collapse
Affiliation(s)
- Kehan Bao
- Regeneron Pharmaceuticals, Tarrytown, NY
| | | | - Erqian Na
- Regeneron Pharmaceuticals, Tarrytown, NY
| | - Qi Su
- Regeneron Pharmaceuticals, Tarrytown, NY
| | | | | | | |
Collapse
|
2
|
Chen X, Yang S, Shaw ND, Xia M. Kisspeptin Receptor Agonists and Antagonists: Strategies for Discovery and Implications for Human Health and Disease. Int J Mol Sci 2025; 26:4890. [PMID: 40430029 PMCID: PMC12112028 DOI: 10.3390/ijms26104890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
The kisspeptin/kisspeptin receptor (KISS1/KISS1R) system has emerged as a vital regulator of various physiological processes, including cancer progression, metabolic function, and reproduction. KISS1R, a member of the G protein-coupled receptor family, is crucial for regulating the hypothalamic/pituitary/gonadal axis. A growing number of KISS1R agonists are currently being investigated in clinical trials, whereas the number of antagonists remains limited. Most existing ligands are synthetic peptides, with only a few small-molecule compounds, such as musk ambrette, having been identified. In this article, we provide an overview of the KISS1/KISS1R system and its involvement in diseases such as reproductive disorders, cancer, diabetes, and cardiovascular disease. We also highlight the various technologies used to identify KISS1R ligands, including radioligand binding assays, calcium flux assays, IP1 formation assays, ERK phosphorylation assays, qRT-PCR, and AI-based virtual screening. Furthermore, we discuss the latest advances in identifying KISS1R agonists and antagonists, highlighting ongoing challenges and future directions in research. These insights lay the groundwork for future research aimed at leveraging this system for developing innovative therapeutic strategies across a range of medical conditions.
Collapse
Affiliation(s)
- Xing Chen
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA; (X.C.); (S.Y.)
| | - Shu Yang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA; (X.C.); (S.Y.)
| | - Natalie D. Shaw
- Pediatric Neuroendocrinology Group, Clinical Research Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, NC 27709, USA;
| | - Menghang Xia
- Pediatric Neuroendocrinology Group, Clinical Research Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, NC 27709, USA;
| |
Collapse
|
3
|
Rodriguez-Carrillo A, Vela-Soria F, Smagulova F, Fernández MF, Freire C. Association between PFAS exposure and metabolic-related biomarkers in Spanish adolescents. ENVIRONMENTAL RESEARCH 2025; 273:121171. [PMID: 39978619 DOI: 10.1016/j.envres.2025.121171] [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: 11/12/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) exert endocrine disruptive effects on the endocrine-metabolic axis. Emerging knowledge suggests that kisspeptin may play a key role in these effects. OBJECTIVE To assess the cross-sectional association of blood PFAS concentrations with kisspeptin levels, KISS1 gene DNA methylation, and metabolic-related biomarkers in adolescent males from the Spanish INMA-Granada cohort. METHODS Seven PFAS and twelve biomarkers (glucose-GLU, total cholesterol-TC, triglycerides, LDL, HDL, ALP, AST, ALT, GGT, total bilirubin-BILT, direct bilirubin-BILD, and urea) were measured in plasma and serum, respectively, from 129 adolescent males (15-17 yrs). Systolic and diastolic blood pressure (SBP, DBP), pulse, z-scored body mass index, kisspeptin protein levels (n = 104) and whole blood KISS1 DNA methylation (n = 117) were determined. Linear regression models, weighted quantile sum (WQS), and Bayesian kernel machine (BKMR) were fit. RESULTS PFHpA was associated with lower GLU levels [% change per log-unit increase in plasma concentrations (95%CI) = -4.73 (-8.98;-0.28)], and PFUnDA with higher GLU, TC, and HDL levels. In models adjusted by kisspeptin level, PFOS was associated with higher SBP [3.42 (-0.12; 7.09)]. Additionally, PFNA and total PFAS concentrations were associated with higher kisspeptin levels [3.91 (0.55; 7.37) and 6.14 (0.47; 12.13), respectively]. Mixture models showed that combined PFAS exposure was associated with higher HDL, lower hepatic biomarkers (ALT, BILD) and higher kisspeptin levels. CONCLUSION Certain PFAS (e.g. PFUnDA) and their mixture were associated with metabolic-related biomarkers, mainly GLU, HDL, and SBP. These associations may be influenced by kisspeptin levels. More studies are needed to verify these observations.
Collapse
Affiliation(s)
- Andrea Rodriguez-Carrillo
- VITO Health, Flemish Institute for Technological Research (VITO), Mol, 2400, Belgium; Toxicological Centre, University of Antwerp, Universiteitsplein, 1, Wilrijk, 2610, Belgium; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| | - Fernando Vela-Soria
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain
| | - Fatima Smagulova
- Univ Rennes, EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, Rennes, F-35000, France
| | - Mariana F Fernández
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain; Department of Radiology and Physical Medicine, School of Medicine, University of Granada, Granada, 18016, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Carmen Freire
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18016, Spain; Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain; Department of Legal Medicine, Toxicology, and Physical Anthropology, School of Medicine, University of Granada, Granada, 18016, Spain.
| |
Collapse
|
4
|
Barrea L, Caprio M, Perrini S, Camajani E, Crafa A, Gangitano E, Gorini S, Sciacca L, Verde L, Albertelli M, Ferone D, Giorgino F, Colao A, Aimaretti G, Muscogiuri G. Diabetes mellitus secondary to endocrine diseases: a position statement of the working group of the club of the Italian society of endocrinology (SIE)-Nutrition hormones and metabolism. J Endocrinol Invest 2025:10.1007/s40618-025-02589-2. [PMID: 40293649 DOI: 10.1007/s40618-025-02589-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE This position statement addressed the limited scientific literature on the management of diabetes mellitus secondary to endocrinopathies, despite its frequent occurrence in hormonal diseases such as acromegaly, Cushing's syndrome, primary hyperaldosteronism, pheochromocytoma, hyperthyroidism, and neuroendocrine tumors. The aim was to review the pathophysiological mechanisms, clinical features, and management strategies, focusing on nutritional and pharmacological approaches. METHODS A comprehensive review of existing literature was conducted regarding studies on diabetes secondary to endocrinopathies and the effects of treatments for these conditions, such as somatostatin analogues and pancreatic surgery. Particular emphasis was placed on understanding glucose metabolism derangements and the interplay between endocrine excess and therapeutic interventions. RESULTS Secondary diabetes arises not only from hormone excess but also as a consequence of treatments for endocrine disorders. For instance, somatostatin analogues, while effective in resolving hormone hypersecretion, impair glucose metabolism by inhibiting pancreatic insulin secretion. Similarly, pancreatic surgery for neuroendocrine tumors often exacerbates glycemic disturbances. The management of secondary diabetes requires a multidisciplinary approach that includes treating the underlying endocrine disorder, tailoring antidiabetic therapy, and optimizing nutritional strategies to mitigate metabolic disruptions. CONCLUSION Diabetes secondary to endocrinopathies presents unique challenges due to its complex etiology and the metabolic effects of treatments. This position statement underscores the importance of an integrated management approach, offering guidance for clinicians in addressing this multifaceted condition. Further research is needed to develop evidence-based guidelines for optimal care.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Psicologia e Scienze della Salute, Università Telematica Pegaso, Centro Direzionale Isola F2, Via Porzio, 80143, Naples, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Rome, Italy
| | - Sebastio Perrini
- Section of Endocrinology, Department of Medicine and Surgery, LUM University, Casamassima, BA, Italy
| | - Elisabetta Camajani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Rome, Italy
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, 95123, Italy
| | - Elena Gangitano
- Department of Experimental Medicine, Sapienza University of Rome, Rome, 00185, Italy
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Rome, Italy
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, University of Catania, Catania, 95123, Italy
| | - Ludovica Verde
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
| | - Manuela Albertelli
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, Genova, Italy
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Diego Ferone
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, Genova, Italy
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, 70124, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia ed Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Università degli Studi di Napoli Federico II, Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Via Sergio Pansini 5, Naples, 80131, Italy
| | - Gianluca Aimaretti
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA.
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia ed Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.
- Università degli Studi di Napoli Federico II, Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Via Sergio Pansini 5, Naples, 80131, Italy.
| |
Collapse
|
5
|
Can U, Akdu S, Aktan AH. The Investigation of Kisspeptin, Spexin and Galanin in Euthyroid Women with Hashimoto's Thyroiditis. Endocr Res 2025; 50:36-42. [PMID: 39056111 DOI: 10.1080/07435800.2024.2384576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/22/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
OBJECTIVE The hallmarks of Hashimoto's thyroiditis (HT) include the destruction of thyroid cells by leading to insulin resistance (IR), hypothyroidism, and metabolic abnormalities. Kisspeptin, spexin, and galanin control appetite and body weight (BW) to regulate metabolisms. Here, we sought to determine if galanin, kisspeptin, and spexin are linked to the pathophysiology of HT in euthyroid female individuals. METHODS Forty-five women with HT and 45 healthy control women of the same age participated in the current study. The enzyme-linked immunosorbent assay (ELISA) method was used to measure the serum levels of galanin, spexin, and kisspeptin. RESULTS In comparison to the controls, HT patients had significantly higher levels of kisspeptin (p < 0.01), galanin (p < 0.01), anti-thyroid peroxidase (anti-TPO) (p < 0.001), anti-thyroglobulin (anti-Tg) (p < 0.001), and body mass index (BMI) (p < 0.05). The two groups were comparable in terms of spexin, free triiodothyronine-3 (fT3), fT4, thyroid-stimulating hormone (TSH) levels, and homeostatic model assessment for insulin resistance (HOMA-IR). Galanin and kisspeptin were seen to have a positive correlation (p < 0.01; r = 0.786). CONCLUSIONS Euthyroid women with HT were found to have higher levels of kisspeptin and galanin. These results imply that kisspeptin and galanin may be linked to the pathogenesis of hypothyroidism, and as a result, we believe that these markers may be beneficial in the early detection and treatment of HT patients.
Collapse
Affiliation(s)
- Ummugulsum Can
- Department of Biochemistry, Konya City Hospital, Konya, Turkey
| | - Sadinaz Akdu
- Department of Biochemistry, Fethiye State Hospital, Muğla, Turkey
| | - Ahmet Hamdi Aktan
- Department of Internal Medicine, Fethiye State Hospital, Muğla, Turkey
| |
Collapse
|
6
|
Shehbaz A, Afzaal M, Akram N, Saeed F, Khan W, Ahmed F, Ahmed A, Asghar A, Faisal Z. Intermittent Fasting and Probiotics: Synergistic Modulation of Gut Health for Therapeutic Advantages. Probiotics Antimicrob Proteins 2025; 17:479-486. [PMID: 39261391 DOI: 10.1007/s12602-024-10358-5] [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] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
Intermittent fasting (IF) is an increasingly popular dietary approach involving alternating fasting and eating periods. This review aims to summarize the growing body of literature demonstrating that IF is a potential nutritional practice that involves alternating periods of fasting and eating and the numerous benefits of IF, especially in the modulation of healthy gut microbiota. The positive impact of intermittent fasting on gut microbiota not only promotes gastrointestinal health but also has far-reaching effects on critical systems throughout the body. Additionally, the evidence presented in this review highlights the significant preventive and therapeutic effects of intermittent fasting on a wide range of disorders. This includes reducing the risk of diabetes, and neurological disorders, alleviating obesity, and enhancing the functioning of the liver, ultimately contributing to the maintenance of metabolic equilibrium. Perhaps most notably, these effects play a substantial role in preventing diabetes, a global health concern of increasing significance. This comprehensive investigation delves into the scientific foundations of intermittent fasting's impact on gut microbiota and its implications for averting chronic diseases, providing valuable insights for future research and therapeutic applications.
Collapse
Affiliation(s)
- Amna Shehbaz
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Noor Akram
- Food Safety & Biotechnology Lab, Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Warda Khan
- Department of Nutritional Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Faiyaz Ahmed
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, 51452, Buraydah, Saudi Arabia
| | - Aftab Ahmed
- Department of Nutritional Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Aasma Asghar
- Department of Nutritional Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zargham Faisal
- Department of Human Nutrition and Dietetics, Iqra University Karachi, Karachi, Pakistan
| |
Collapse
|
7
|
Long BY, Liao X, Liang X. The Hypothalamus and Pituitary Gland Regulate Reproduction and Are Involved in the Development of Polycystic Ovary Syndrome. Neuroendocrinology 2025; 115:315-334. [PMID: 39894018 DOI: 10.1159/000543877] [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: 10/29/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex condition with unclear mechanisms, posing a challenge for prevention and treatment of PCOS. The role of the hypothalamus and pituitary gland in regulating female reproduction is critical. Abnormalities in the hypothalamus and pituitary can impair reproductive function. It is important to study hypothalamic and pituitary changes in patients with PCOS. SUMMARY This article reviews articles on the hypothalamus and PCOS with the goal of summarizing what abnormalities of the hypothalamic-pituitary-ovarian axis are present in patients with PCOS and to clarify the pathogenesis of PCOS. We find that the mechanisms by which the hypothalamus and pituitary regulate reproduction in girls are complex and are associated with altered sex hormone levels, obesity, and insulin resistance. Different animal models of PCOS are characterized by different alterations in the hypothalamus and pituitary and respond differently to different treatments, which correspond to the complex pathogenesis of patients with PCOS. KEY MESSAGES Arcuate nucleus (ARC) is associated with luteinizing hormone (LH) surges. Suprachiasmatic nucleus, ARC, and RP3V are associated with LH surges. Animal models of PCOS have different characteristics.
Collapse
Affiliation(s)
- Bin-Yang Long
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xipeng Liao
- Tianjin University of Technology, Tianjin, China
| | - Xin Liang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Karampelias C, Băloiu B, Rathkolb B, da Silva-Buttkus P, Bachar-Wikström E, Marschall S, Fuchs H, Gailus-Durner V, Chu L, Hrabě de Angelis M, Andersson O. Examining the liver-pancreas crosstalk reveals a role for the molybdenum cofactor in β-cell regeneration. Life Sci Alliance 2024; 7:e202402771. [PMID: 39159974 PMCID: PMC11333758 DOI: 10.26508/lsa.202402771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024] Open
Abstract
Regeneration of insulin-producing β-cells is an alternative avenue to manage diabetes, and it is crucial to unravel this process in vivo during physiological responses to the lack of β-cells. Here, we aimed to characterize how hepatocytes can contribute to β-cell regeneration, either directly or indirectly via secreted proteins or metabolites, in a zebrafish model of β-cell loss. Using lineage tracing, we show that hepatocytes do not directly convert into β-cells even under extreme β-cell ablation conditions. A transcriptomic analysis of isolated hepatocytes after β-cell ablation displayed altered lipid- and glucose-related processes. Based on the transcriptomics, we performed a genetic screen that uncovers a potential role of the molybdenum cofactor (Moco) biosynthetic pathway in β-cell regeneration and glucose metabolism in zebrafish. Consistently, molybdenum cofactor synthesis 2 (Mocs2) haploinsufficiency in mice indicated dysregulated glucose metabolism and liver function. Together, our study sheds light on the liver-pancreas crosstalk and suggests that the molybdenum cofactor biosynthesis pathway should be further studied in relation to glucose metabolism and diabetes.
Collapse
Affiliation(s)
- Christos Karampelias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Bianca Băloiu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Etty Bachar-Wikström
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Susan Marschall
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lianhe Chu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
9
|
Prasad K, Bhattacharya D, Shams SGE, Izarraras K, Hart T, Mayfield B, Blaszczyk MB, Zhou Z, Pajvani UB, Friedman SL, Bhattacharya M. Kisspeptin Alleviates Human Hepatic Fibrogenesis by Inhibiting TGFβ Signaling in Hepatic Stellate Cells. Cells 2024; 13:1651. [PMID: 39404414 PMCID: PMC11476267 DOI: 10.3390/cells13191651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
The peptide hormone kisspeptin attenuates liver steatosis, metabolic dysfunction-associated steatohepatitis (MASH), and fibrosis in mouse models by signaling via the kisspeptin 1 receptor (KISS1R). However, whether kisspeptin impacts fibrogenesis in the human liver is not known. We investigated the impact of a potent kisspeptin analog (KPA) on fibrogenesis using human precision-cut liver slices (hPCLS) from fibrotic livers from male patients, in human hepatic stellate cells (HSCs), LX-2, and in primary mouse HSCs. In hPCLS, 48 h and 72 h of KPA (3 nM, 100 nM) treatment decreased collagen secretion and lowered the expression of fibrogenic and inflammatory markers. Immunohistochemical studies revealed that KISS1R is expressed and localized to HSCs in MASH/fibrotic livers. In HSCs, KPA treatment reduced transforming growth factor b (TGFβ)-the induced expression of fibrogenic and inflammatory markers, in addition to decreasing TGFβ-induced collagen secretion, cell migration, proliferation, and colony formation. Mechanistically, KISS1R signaling downregulated TGFβ signaling by decreasing SMAD2/3 phosphorylation via the activation of protein phosphatases, PP2A, which dephosphorylates SMAD 2/3. This study revealed for the first time that kisspeptin reverses human hepatic fibrogenesis, thus identifying it as a new therapeutic target to treat hepatic fibrosis.
Collapse
Affiliation(s)
- Kavita Prasad
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.B.); (S.L.F.)
| | - Shams Gamal Eldin Shams
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Kimberly Izarraras
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Tia Hart
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| | - Brent Mayfield
- Department of Medicine, Columbia University, New York, NY 10032, USA; (B.M.); (U.B.P.)
| | - Maryjka B. Blaszczyk
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (M.B.B.); (Z.Z.)
| | - Zhongren Zhou
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (M.B.B.); (Z.Z.)
| | - Utpal B. Pajvani
- Department of Medicine, Columbia University, New York, NY 10032, USA; (B.M.); (U.B.P.)
| | - Scott L. Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.B.); (S.L.F.)
| | - Moshmi Bhattacharya
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA; (K.P.); (S.G.E.S.); (K.I.); (T.H.)
| |
Collapse
|
10
|
Hu R, Geng Y, Huang Y, Liu Z, Li F, Song K, Ma W, Dong H, Zhang M, Lei T, Song Y, Zhang Z. Jiawei Buzhong Yiqi Decoction attenuates polycystic ovary syndrome through regulating kisspeptin-GPR54-AKT-SHBG system. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155931. [PMID: 39116604 DOI: 10.1016/j.phymed.2024.155931] [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: 07/05/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common reproductive endocrine disorders. Accumulated evidence has suggested the indispensable role of kisspeptin-G protein-coupled receptor (GPR54) system and SHBG in development of PCOS. However, potential mechanisms and their relationship are unclear. Jiawei Buzhong Yiqi Decoction (JWBZYQ) has been reported to ameliorate obese PCOS. Whereas, potential mechanisms remain elusive. PURPOSE To determine whether JWBZYQ attenuates PCOS by regulating the kisspeptin-GPR54 system and SHBG production. And to explore potential mechanisms. METHODS An overweight PCOS rat model was developed with testosterone propionate (TP) and high-fat diet (HFD). The efficacy of JWBZYQ was assessed by tracking changes in weight, estrous cycle, ovarian morphology, and serum sex hormone levels. Additionally, kisspeptin-GPR54 system expression in multiple organs and PI3K-AKT pathway activity in liver of different rats were detected. Modifications in SHBG production were also measured. Kisspeptin54 was administered to establish a cellular model. The levels of AKT phosphorylation and SHBG protein within HepG2 cells were analyzed. Finally, confirmatory studies were performed using AKT phosphorylation activator and inhibitor. RESULTS JWBZYQ effectively attenuated the overweight, disrupted estrous cycle, altered sex hormone levels, and aberrant ovarian morphology in PCOS rats. Meanwhile, PCOS rats exhibited elevated levels of kisspeptin and GPR54, along with reduced SHBG levels, which could be reversed by JWBZYQ. These alterations might be connected with the activation of AKT phosphorylation. In vitro experiment identified that JWBZYQ could rectify the hyperactivated AKT phosphorylation and deficient production of SHBG caused by kisspeptin54. CONCLUSIONS Overexpressed kisspeptin-GPR54 system inhibited SHBG synthesis in PCOS. JWBZYQ curtailed the exorbitant expression of kisspeptin and GPR54, which moderated the rise in AKT phosphorylation and subsequently promoted the production of SHBG.
Collapse
Affiliation(s)
- Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Haoxu Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Lei
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yufan Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuo Zhang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
11
|
Shi Y, Ma J, Li S, Liu C, Liu Y, Chen J, Liu N, Liu S, Huang H. Sex difference in human diseases: mechanistic insights and clinical implications. Signal Transduct Target Ther 2024; 9:238. [PMID: 39256355 PMCID: PMC11387494 DOI: 10.1038/s41392-024-01929-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/26/2024] [Accepted: 07/23/2024] [Indexed: 09/12/2024] Open
Abstract
Sex characteristics exhibit significant disparities in various human diseases, including prevalent cardiovascular diseases, cancers, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Risk profiles and pathological manifestations of these diseases exhibit notable variations between sexes. The underlying reasons for these sex disparities encompass multifactorial elements, such as physiology, genetics, and environment. Recent studies have shown that human body systems demonstrate sex-specific gene expression during critical developmental stages and gene editing processes. These genes, differentially expressed based on different sex, may be regulated by androgen or estrogen-responsive elements, thereby influencing the incidence and presentation of cardiovascular, oncological, metabolic, immune, and neurological diseases across sexes. However, despite the existence of sex differences in patients with human diseases, treatment guidelines predominantly rely on male data due to the underrepresentation of women in clinical trials. At present, there exists a substantial knowledge gap concerning sex-specific mechanisms and clinical treatments for diverse diseases. Therefore, this review aims to elucidate the advances of sex differences on human diseases by examining epidemiological factors, pathogenesis, and innovative progress of clinical treatments in accordance with the distinctive risk characteristics of each disease and provide a new theoretical and practical basis for further optimizing individualized treatment and improving patient prognosis.
Collapse
Affiliation(s)
- Yuncong Shi
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jianshuai Ma
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Sijin Li
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Chao Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Yuning Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jie Chen
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ningning Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shiming Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China.
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Wu Z, Chen G, Qiu C, Yan X, Xu L, Jiang S, Xu J, Han R, Shi T, Liu Y, Gao W, Wang Q, Li J, Ye F, Pan X, Zhang Z, Ning P, Zhang B, Chen J, Du Y. Structural basis for the ligand recognition and G protein subtype selectivity of kisspeptin receptor. SCIENCE ADVANCES 2024; 10:eadn7771. [PMID: 39151001 PMCID: PMC11328905 DOI: 10.1126/sciadv.adn7771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/11/2024] [Indexed: 08/18/2024]
Abstract
Kisspeptin receptor (KISS1R), belonging to the class A peptide-GPCR family, plays a key role in the regulation of reproductive physiology after stimulation by kisspeptin and is regarded as an attractive drug target for reproductive diseases. Here, we demonstrated that KISS1R can couple to the Gi/o pathway besides the well-known Gq/11 pathway. We further resolved the cryo-electron microscopy (cryo-EM) structure of KISS1R-Gq and KISS1R-Gi complexes bound to the synthetic agonist TAK448 and structure of KISS1R-Gq complex bound to the endogenous agonist KP54. The high-resolution structures provided clear insights into mechanism of KISS1R recognition by its ligand and can facilitate the design of targeted drugs with high affinity to improve treatment effects. Moreover, the structural and functional analyses indicated that conformational differences in the extracellular loops (ECLs), intracellular loops (ICLs) of the receptor, and the "wavy hook" of the Gα subunit may account for the specificity of G protein coupling for KISS1R signaling.
Collapse
Affiliation(s)
- Zhangsong Wu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Chen Qiu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Xiaoyi Yan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Lezhi Xu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Shirui Jiang
- The Huanan Affiliated Hospital of Shenzhen University, Shenzhen University, 518000 Shenzhen, Guangdong, China
| | - Jun Xu
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Runyuan Han
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tingyi Shi
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Yiming Liu
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Qian Wang
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
- The Huanan Affiliated Hospital of Shenzhen University, Shenzhen University, 518000 Shenzhen, Guangdong, China
| | - Jiancheng Li
- Instrumental Analysis Center, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Fang Ye
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Xin Pan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Zhiyi Zhang
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Peiruo Ning
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Binghao Zhang
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, 272067 Jining, Shandong, China
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172 Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Shen S, Wang D, Liu H, He X, Cao Y, Chen J, Li S, Cheng X, Xu HE, Duan J. Structural basis for hormone recognition and distinctive Gq protein coupling by the kisspeptin receptor. Cell Rep 2024; 43:114389. [PMID: 38935498 DOI: 10.1016/j.celrep.2024.114389] [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: 12/13/2023] [Revised: 04/25/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
Kisspeptin signaling through its G protein-coupled receptor, KISS1R, plays an indispensable role in regulating reproduction via the hypothalamic-pituitary-gonadal axis. Dysregulation of this pathway underlies severe disorders like infertility and precocious puberty. Here, we present cryo-EM structures of KISS1R bound to the endogenous agonist kisspeptin-10 and a synthetic analog TAK-448. These structures reveal pivotal interactions between peptide ligands and KISS1R extracellular loops for receptor activation. Both peptides exhibit a conserved binding mode, unveiling their common activation mechanism. Intriguingly, KISS1R displays a distinct 40° angular deviation in its intracellular TM6 region compared to other Gq-coupled receptors, enabling distinct interactions with Gq. This study reveals the molecular intricacies governing ligand binding and activation of KISS1R, while highlighting its exceptional ability to couple with Gq. Our findings pave the way for structure-guided design of therapeutics targeting this physiologically indispensable receptor.
Collapse
Affiliation(s)
- Shiyi Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongxue Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Heng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinheng He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinglong Cao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Juanhua Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shujie Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xi Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Jia Duan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
14
|
Liang C, Li X, Song G, Schmidt SF, Sun L, Chen J, Pan X, Zhao H, Yan Y. Adipose Kiss1 controls aerobic exercise-related adaptive responses in adipose tissue energy homeostasis. FASEB J 2024; 38:e23743. [PMID: 38877852 DOI: 10.1096/fj.202302598rr] [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: 12/15/2023] [Revised: 05/13/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024]
Abstract
Kisspeptin signaling regulates energy homeostasis. Adiposity is the principal source and receiver of peripheral Kisspeptin, and adipose Kiss1 metastasis suppressor (Kiss1) gene expression is stimulated by exercise. However, whether the adipose Kiss1 gene regulates energy homeostasis and plays a role in adaptive alterations during prolonged exercise remains unknown. Here, we investigated the role of Kiss1 role in mice and adipose tissues and the adaptive changes it induces after exercise, using adipose-specific Kiss1 knockout (Kiss1adipoq-/-) and adeno-associated virus-induced adipose tissue Kiss1-overexpressing (Kiss1adipoq over) mice. We found that adipose-derived kisspeptin signal regulates lipid and glucose homeostasis to maintain systemic energy homeostasis, but in a sex-dependent manner, with more pronounced metabolic changes in female mice. Kiss1 regulated adaptive alterations of genes and proteins in tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OxPhos) pathways in female gWAT following prolonged aerobic exercise. We could further show that adipose Kiss1 deficiency leads to reduced peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) protein content of soleus muscle and maximum oxygen uptake (VO2 max) of female mice after prolonged exercise. Therefore, adipose Kisspeptin may be a novel adipokine that increases organ sensitivity to glucose, lipids, and oxygen following exercise.
Collapse
Affiliation(s)
- Chunyu Liang
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
- Department of Biochemistry and Molecular Biology, Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark (SDU), Odense, Denmark
- School of Physical Education, Guangxi University (GXU), Nanning, China
| | - Xuehan Li
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Ge Song
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Søren Fisker Schmidt
- Department of Biochemistry and Molecular Biology, Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark (SDU), Odense, Denmark
| | - Lingyu Sun
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Jianhao Chen
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Xinliang Pan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Haotian Zhao
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Yi Yan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| |
Collapse
|
15
|
Wang M, Pugh SM, Daboul J, Miller D, Xu Y, Hill JW. IGF-1 Acts through Kiss1-expressing Cells to Influence Metabolism and Reproduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601722. [PMID: 39005405 PMCID: PMC11244982 DOI: 10.1101/2024.07.02.601722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Objective Kisspeptin, encoded by the Kiss1 gene, ties puberty and fertility to energy status; however, the metabolic factors that control Kiss1-expressing cells need to be clarified. Methods To evaluate the impact of IGF-1 on the metabolic and reproductive functions of kisspeptin producing cells, we created mice with IGF-1 receptor deletion driven by the Kiss1 promoter (IGF1RKiss1 mice). Previous studies have shown IGF-1 and insulin can bind to each other's receptor, permitting IGF-1 signaling in the absence of IGF1R. Therefore, we also generated mice with simultaneous deletion of the IGF1R and insulin receptor (IR) in Kiss1-expressing cells (IGF1R/IRKiss1 mice). Results Loss of IGF1R in Kiss1 cells caused stunted body length. In addition, female IGF1RKiss1 mice displayed lower body weight and food intake plus higher energy expenditure and physical activity. This phenotype was linked to higher proopiomelanocortin (POMC) expression and heightened brown adipose tissue (BAT) thermogenesis. Male IGF1RKiss1 mice had mild changes in metabolic functions. Moreover, IGF1RKiss1 mice of both sexes experienced delayed puberty. Notably, male IGF1RKiss1 mice had impaired adulthood fertility accompanied by lower gonadotropin and testosterone levels. Thus, IGF1R in Kiss1-expressing cells impacts metabolism and reproduction in a sex-specific manner. IGF1R/IRKiss1 mice had higher fat mass and glucose intolerance, suggesting IGF1R and IR in Kiss1-expressing cells together regulate body composition and glucose homeostasis. Conclusions Overall, our study shows that IGF1R and IR in Kiss1 have cooperative roles in body length, metabolism, and reproduction.
Collapse
Affiliation(s)
- Mengjie Wang
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Seamus M. Pugh
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Judy Daboul
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - David Miller
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Yong Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer W. Hill
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, USA
- Department of Obstetrics and Gynecology, University of Toledo College of Medicine, Toledo, Ohio, USA
| |
Collapse
|
16
|
Ding L, Sun Y, Liang Y, Zhang J, Fu Z, Ren C, Li P, Liu W, Xiao R, Wang H, Zhang Z, Yue X, Li C, Wu Z, Feng Y, Liang X, Ma C, Gao L. Beta-Cell Tipe1 Orchestrates Insulin Secretion and Cell Proliferation by Promoting Gαs/cAMP Signaling via USP5. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304940. [PMID: 38417114 PMCID: PMC11040358 DOI: 10.1002/advs.202304940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/09/2024] [Indexed: 03/01/2024]
Abstract
Inadequate β-cell mass and insulin secretion are essential for the development of type 2 diabetes (T2D). TNF-α-induced protein 8-like 1 (Tipe1) plays a crucial role in multiple diseases, however, a specific role in T2D pathogenesis remains largely unexplored. Herein, Tipe1 as a key regulator in T2D, contributing to the maintenance of β cell homeostasis is identified. The results show that the β-cell-specific knockout of Tipe1 (termed Ins2-Tipe1BKO) aggravated diabetic phenotypes in db/db mice or in mice with high-fat diet-induced diabetes. Notably, Tipe1 improves β cell mass and function, a process that depends on Gαs, the α subunit of the G-stimulating protein. Mechanistically, Tipe1 inhibited the K48-linked ubiquitination degradation of Gαs by recruiting the deubiquitinase USP5. Consequently, Gαs or cAMP agonists almost completely restored the dysfunction of β cells observed in Ins2-Tipe1BKO mice. The findings characterize Tipe1 as a regulator of β cell function through the Gαs/cAMP pathway, suggesting that Tipe1 may emerge as a novel target for T2D intervention.
Collapse
Affiliation(s)
- Lu Ding
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Yang Sun
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Yan Liang
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Jie Zhang
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Zhendong Fu
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Caiyue Ren
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Pengfei Li
- Department of EndocrinologyYucheng People's HospitalDezhouShandong251200P. R. China
| | - Wen Liu
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Rong Xiao
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Hao Wang
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Zhaoying Zhang
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Histology and EmbryologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Yuemin Feng
- Department of GastroenterologyShengLi Hospital of Shandong First Medical UniversityJinanShandong250012P. R. China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of EducationShandong Key Laboratory of Infection and Immunityand Department of ImmunologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| |
Collapse
|
17
|
Sliwowska JH, Woods NE, Alzahrani AR, Paspali E, Tate RJ, Ferro VA. Kisspeptin a potential therapeutic target in treatment of both metabolic and reproductive dysfunction. J Diabetes 2024; 16:e13541. [PMID: 38599822 PMCID: PMC11006622 DOI: 10.1111/1753-0407.13541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/21/2023] [Accepted: 02/03/2024] [Indexed: 04/12/2024] Open
Abstract
Kisspeptins (KPs) are proteins that were first recognized to have antimetastatic action. Later, the critical role of this peptide in the regulation of reproduction was proved. In recent years, evidence has been accumulated supporting a role for KPs in regulating metabolic processes in a sexual dimorphic manner. It has been proposed that KPs regulate metabolism both indirectly via gonadal hormones and/or directly via the kisspeptin receptor in the brain, brown adipose tissue, and pancreas. The aim of the review is to provide both experimental and clinical evidence indicating that KPs are peptides linking metabolism and reproduction. We propose that KPs could be used as a potential target to treat both metabolic and reproductive abnormalities. Thus, we focus on the consequences of disruptions in KPs and their receptors in metabolic conditions such as diabetes, undernutrition, obesity, and reproductive disorders (hypogonadotropic hypogonadism and polycystic ovary syndrome). Data from both animal models and human subjects indicate that alterations in KPs in the case of metabolic imbalance lead also to disruptions in reproductive functions. Changes both in the hypothalamic and peripheral KP systems in animal models of the aforementioned disorders are discussed. Finally, an overview of current clinical studies involving KP in fertility and metabolism show fewer studies on metabolism (15%) and only one to date on both. Presented data indicate a dynamic and emerging field of KP studies as possible therapeutic targets in treatments of both reproductive and metabolic dysfunctions.
Collapse
Affiliation(s)
- Joanna Helena Sliwowska
- Department of Veterinary Medicine and Animal Sciences, Laboratory of Neurobiology, Poznan University of Life Sciences, Poznan, Poland
| | - Nicola Elizabeth Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Abdullah Rzgallah Alzahrani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Elpiniki Paspali
- Department of Chemical Engineering, University of Strathclyde, Glasgow, UK
| | - Rothwelle Joseph Tate
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Valerie Anne Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
18
|
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
|
19
|
Patel B, Koysombat K, Mills EG, Tsoutsouki J, Comninos AN, Abbara A, Dhillo WS. The Emerging Therapeutic Potential of Kisspeptin and Neurokinin B. Endocr Rev 2024; 45:30-68. [PMID: 37467734 PMCID: PMC10765167 DOI: 10.1210/endrev/bnad023] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Kisspeptin (KP) and neurokinin B (NKB) are neuropeptides that govern the reproductive endocrine axis through regulating hypothalamic gonadotropin-releasing hormone (GnRH) neuronal activity and pulsatile GnRH secretion. Their critical role in reproductive health was first identified after inactivating variants in genes encoding for KP or NKB signaling were shown to result in congenital hypogonadotropic hypogonadism and a failure of pubertal development. Over the past 2 decades since their discovery, a wealth of evidence from both basic and translational research has laid the foundation for potential therapeutic applications. Beyond KP's function in the hypothalamus, it is also expressed in the placenta, liver, pancreas, adipose tissue, bone, and limbic regions, giving rise to several avenues of research for use in the diagnosis and treatment of pregnancy, metabolic, liver, bone, and behavioral disorders. The role played by NKB in stimulating the hypothalamic thermoregulatory center to mediate menopausal hot flashes has led to the development of medications that antagonize its action as a novel nonsteroidal therapeutic agent for this indication. Furthermore, the ability of NKB antagonism to partially suppress (but not abolish) the reproductive endocrine axis has supported its potential use for the treatment of various reproductive disorders including polycystic ovary syndrome, uterine fibroids, and endometriosis. This review will provide a comprehensive up-to-date overview of the preclinical and clinical data that have paved the way for the development of diagnostic and therapeutic applications of KP and NKB.
Collapse
Affiliation(s)
- Bijal Patel
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Kanyada Koysombat
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Edouard G Mills
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Jovanna Tsoutsouki
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Alexander N Comninos
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Ali Abbara
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| |
Collapse
|
20
|
Matuszewska J, Nowacka-Woszuk J, Radziejewska A, Grzęda E, Pruszyńska-Oszmałek E, Dylewski Ł, Chmurzyńska A, Sliwowska JH. Maternal cafeteria diet influences kisspeptin (Kiss1), kisspeptin receptor(Gpr54), and sirtuin (Sirt1) genes, hormonal and metabolic profiles, and reproductive functions in rat offspring in a sex-specific manner†. Biol Reprod 2023; 109:654-668. [PMID: 37665248 PMCID: PMC10651067 DOI: 10.1093/biolre/ioad101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/27/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023] Open
Abstract
Kisspeptin (KP, encoded by Kiss1, binding to the Gpr54 receptor) is a neuropeptide conveying information on the metabolic status to the hypothalamic-pituitary-gonadal axis. KP acts together with dynorphin A (encoded by Pdyn) and neurokinin B (encoded by Tac2) to regulate reproduction. KP is crucial for the onset of puberty and is under the control of sirtuin (encoded by Sirt1). We hypothesize that the maternal cafeteria (CAF) diet has adverse effects on the offspring's hormonal, metabolic, and reproductive functions due to sex-specific alterations in the expression of Kiss1, Gpr54, Pdyn, Tac2, and Sirt1 in the hypothalamus, and Kiss1, Gpr54, and Sirt1 in the liver. Rats were fed a CAF diet before pregnancy, during pregnancy, and during lactation. The vaginal opening was monitored. Offspring were sacrificed in three age points: PND 30, PND 35, and PND 60 (females) and PND 40, PND 45, and PND 60 (males). Their metabolic and hormonal status was assessed. mRNA for Kiss1, Gpr54, Pdyn, Tac2, and Sirt1 were measured by real-time PCR in the hypothalamus and/or livers. We found that CAF offspring had lower weight and altered body composition; increased cholesterol and triglyceride levels, sex-specific changes in glucose and insulin levels; sex-dependent changes in Sirt1/Kiss1 mRNA ratio in the hypothalamus; sex-specific alterations in Kiss1 and Sirt1 mRNA in the liver with more diversity in males; and a delayed puberty onset in females. We concluded that the mother's CAF diet leads to sex-specific alterations in metabolic and reproductive outcomes via Kiss1/Gpr54 and Sirt1 systems in offspring.
Collapse
Affiliation(s)
- Julia Matuszewska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Poznan, Poland
| | - Joanna Nowacka-Woszuk
- Department of Genetics and Animal Breeding, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Poznan, Poland
| | - Anna Radziejewska
- Department of Human Nutrition and Dietetics, Poznan University of Life Sciences, Poznan, Poland
| | - Emilia Grzęda
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Poznan, Poland
| | - Ewa Pruszyńska-Oszmałek
- Department of Animal Physiology, Biochemistry and Biostructure, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Poznan, Poland
| | - Łukasz Dylewski
- Department of Zoology, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Poznan, Poland
| | - Agata Chmurzyńska
- Department of Human Nutrition and Dietetics, Poznan University of Life Sciences, Poznan, Poland
| | - Joanna H Sliwowska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Poznan, Poland
| |
Collapse
|
21
|
Chen Q, Jiang FJ, Gao X, Li XY, Xia P. Steatotic hepatocyte-derived extracellular vesicles promote β-cell apoptosis and diabetes via microRNA-126a-3p. Liver Int 2023; 43:2560-2570. [PMID: 37337778 DOI: 10.1111/liv.15654] [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: 08/07/2022] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/21/2023]
Abstract
Extracellular vesicles (EVs) have emerged as a unique mediator of interorgan communications, playing important roles in the pathophysiologic process of various diseases, including diabetes and other metabolic diseases. Here, we reported that the EVs released by steatotic hepatocytes exerted a detrimental effect on pancreatic β cells, leading to β-cell apoptosis and dysfunction. The effect was profoundly attributable to an up-regulation of miR-126a-3p in the steatotic hepatocyte-derived EVs. Accordingly, overexpression of miR-126a-3p promoted, whereas inhibition of miR-126a-3p prevented β-cell apoptosis, through a mechanism related to its target gene, insulin receptor substrate-2. Moreover, inhibition of miR-126a-3p by its specific antagomir was able to partially reverse the loss of β-cell mass and ameliorate hyperglycaemia in diabetic mice. Thus, the findings reveal a novel pathogenic role of steatotic hepatocyte-derived EVs, which mechanistically links nonalcoholic fatty liver disease to the development of diabetes.
Collapse
Affiliation(s)
- Qi Chen
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fang-Jie Jiang
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Ying Li
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pu Xia
- Department of Endocrinology and Metabolism, Fudan Institute for Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Abstract
Precocious puberty (PP) means the appearance of secondary sexual characters before the age of eight years in girls and nine years in boys. Puberty is indicated in girls by the enlargement of the breasts (thelarche) in girls and in boys by the enlargement of the testes in either volume or length (testicular volume = 4 mL, testicular length = 25 mm, or both). Two types of PP are recognized - namely central PP (CPP) and peripheral PP (PPP). This paper aims to describe the clinical findings and laboratory workup of PP and to illustrate the new trends in the management of precocious sexual maturation. Gonadotropin-releasing hormone (GnRH)-independent type (PPP) refers to the development of early pubertal maturation not related to the central activation of the hypothalamic-pituitary-gonadal (HPG) axis. It is classified into genetic or acquired disorders. The most common forms of congenital or genetic causes involve McCune-Albright syndrome (MAS), familial male-limited PP, and congenital adrenal hyperplasia. The acquired causes include exogenous exposure to androgens, functioning tumors or cysts, and the pseudo-PP of profound primary hypothyroidism. On the other hand, CPP is the most common and it is a gonadotropin-dependent form. It is due to premature maturation of the HPG axis. CPP may occur as genetic alterations, such as MKRN3, DLK1, or KISS1;as a part of mutations in the epigenetic factors that regulate the HPG axis, such as Lin28b and let-7; or as a part of syndromes, central lesions such as hypothalamic hamartoma, and others. A full, detailed history and physical examination should be taken. Furthermore, several investigations should be conducted for both types of PP, including the estimation of serum gonadotropins such as luteinizing and follicle-stimulating hormones and sex steroids, in addition to a radiographic workup and thyroid function tests. Treatment depends on the type of PP: Long-acting GnRHa, either intramuscularly or implanted, is the norm of care for CPP management, while in PPP, especially in congenital adrenal hyperplasia, the goal of management is to suppress adrenal androgen secretion by glucocorticoids. In addition, anastrozole and letrozole - third-generation aromatase inhibitors - are more potent for MAS.
Collapse
Affiliation(s)
- Ahmed Alghamdi
- Pediatric Endocrinology, Faculty of Medicine, Al Baha University, Al Baha, SAU
| |
Collapse
|
23
|
Guarnotta V, Emanuele F, Salzillo R, Bonsangue M, Amato C, Mineo MI, Giordano C. Practical therapeutic approach in the management of diabetes mellitus secondary to Cushing's syndrome, acromegaly and neuroendocrine tumours. Front Endocrinol (Lausanne) 2023; 14:1248985. [PMID: 37842314 PMCID: PMC10569460 DOI: 10.3389/fendo.2023.1248985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/28/2023] [Indexed: 10/17/2023] Open
Abstract
Cushing's syndrome, acromegaly and neuroendocrine disorders are characterized by an excess of counterregulatory hormones, able to induce insulin resistance and glucose metabolism disorders at variable degrees and requiring immediate treatment, until patients are ready to undergo surgery. This review focuses on the management of diabetes mellitus in endocrine disorders related to an excess of counterregulatory hormones. Currently, the landscape of approved agents for treatment of diabetes is dynamic and is mainly patient-centred and not glycaemia-centred. In addition, personalized medicine is more and more required to provide a precise approach to the patient's disease. For this reason, we aimed to define a practical therapeutic algorithm for management of diabetes mellitus in patients with glucagonoma, pheochromocytoma, Cushing's syndrome and acromegaly, based on our practical experience and on the physiopathology of the specific endocrine disease taken into account. This document is addressed to all specialists who approach patients with diabetes mellitus secondary to endocrine disorders characterized by an excess of counterregulatory hormones, in order to take better care of these patients. Care and control of diabetes mellitus should be one of the primary goals in patients with an excess of counterregulatory hormones requiring immediate and aggressive treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Carla Giordano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, Palermo, Italy
| |
Collapse
|
24
|
Di Giorgio NP, Bizzozzero-Hiriart M, Surkin PN, Repetto E, Bonaventura MM, Tabares FN, Bourguignon NS, Converti A, Gomez JMR, Bettler B, Lux-Lantos V. Deletion of GABAB receptors from Kiss1 cells affects glucose homeostasis without altering reproduction in male mice. Am J Physiol Endocrinol Metab 2023; 324:E314-E329. [PMID: 36652400 DOI: 10.1152/ajpendo.00129.2022] [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] [Indexed: 01/19/2023]
Abstract
Kisspeptin and γ-amino butyric acid (GABA), synthesized in the central nervous system, are critical for reproduction. Both are also expressed in peripheral organs/tissues critical to metabolic control (liver/pancreas/adipose). Many kisspeptin neurons coexpress GABAB receptors (GABABR) and GABA controls kisspeptin expression and secretion. We developed a unique mouse lacking GABABR exclusively from kisspeptin cells/neurons (Kiss1-GABAB1KO) to evaluate the impact on metabolism/reproduction. We confirmed selective deletion of GABABR from Kiss1 cells in the anteroventral periventricular nucleus/periventricular nucleus continuum (AVPV/PeN; immunofluorescence and PCR) and arcuate nucleus (ARC), medial amygdala (MeA), pituitary, liver, and testes (PCR). Young Kiss1-GABAB1KO males were fertile, with normal LH and testosterone. Kiss1 expression was similar between genotypes in AVPV/PeN, ARC, MeA, bed nucleus of the stria terminalis (BNST), and peripheral organs (testis, liver, pituitary). Kiss1-GABAB1KO males presented higher fasted glycemia and insulin levels, an impaired response to a glucose overload, reduced insulin sensitivity, and marked insulin resistance. Interestingly, when Kiss1-GABAB1KO males got older (9 mo old) their body weight (BW) increased, in part due to an increase in white adipose tissue (WAT). Old Kiss1-GABAB1KO males showed higher fasted insulin, increased pancreatic insulin content, insulin resistance, and significantly decreased pancreatic kisspeptin levels. In sum, lack of GABABR specifically in Kiss1 cells severely impacts glucose homeostasis in male mice, reinforcing kisspeptin involvement in metabolic regulation. These alterations in glucose homeostasis worsened with aging. We highlight the impact of GABA through GABABR in the regulation of the pancreas kisspeptin system in contrast to liver kisspeptin that was not affected.NEW & NOTEWORTHY We developed a unique mouse lacking GABAB receptors specifically in Kiss1 cells to evaluate the impact on reproduction and metabolism. Knockout males showed a severe impact on glucose homeostasis, which worsened with aging. These results reinforce the proposed kisspeptin involvement in metabolic regulation and highlight the impact of GABA through GABABR in the regulation of the peripheral pancreas kisspeptin system.
Collapse
Affiliation(s)
- Noelia P Di Giorgio
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Marianne Bizzozzero-Hiriart
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Pablo N Surkin
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Esteban Repetto
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - María M Bonaventura
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Florencia N Tabares
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Nadia S Bourguignon
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Ayelén Converti
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Juan M Riaño Gomez
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, Basel, Switzerland
| | - Victoria Lux-Lantos
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
25
|
Dogra S, Das D, Maity SK, Paul A, Rawat P, Daniel PV, Das K, Mitra S, Chakrabarti P, Mondal P. Liver-Derived S100A6 Propels β-Cell Dysfunction in NAFLD. Diabetes 2022; 71:2284-2296. [PMID: 35899967 DOI: 10.2337/db22-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an independent predictor of systemic insulin resistance and type 2 diabetes mellitus (T2DM). However, converse correlates between excess liver fat content and β-cell function remain equivocal. Specifically, how the accumulation of liver fat consequent to the enhanced de novo lipogenesis (DNL) leads to pancreatic β-cell failure and eventually to T2DM is elusive. Here, we have identified that low-molecular-weight calcium-binding protein S100A6, or calcyclin, inhibits glucose-stimulated insulin secretion (GSIS) from β cells through activation of the receptor for the advanced glycation end products and diminution of mitochondrial respiration. Serum S100A6 level is elevated both in human patients with NAFLD and in a high-fat diet-induced mouse model of NAFLD. Although serum S100A6 levels are negatively associated with β-cell insulin secretory capacity in human patients, depletion of hepatic S100A6 improves GSIS and glycemia in mice, suggesting that S100A6 contributes to the pathophysiology of diabetes in NAFLD. Moreover, transcriptional induction of hepatic S100A6 is driven by the potent regulator of DNL, carbohydrate response element-binding protein (ChREBP), and ectopic expression of ChREBP in the liver suppresses GSIS in a S100A6-sensitive manner. Together, these data suggest elevated serum levels of S100A6 may serve as a biomarker in identifying patients with NAFLD with a heightened risk of developing β-cell dysfunction. Overall, our data implicate S100A6 as, to our knowledge, a hitherto unknown hepatokine to be activated by ChREBP and that participates in the hepato-pancreatic communication to impair insulin secretion and drive the development of T2DM in NAFLD.
Collapse
Affiliation(s)
- Surbhi Dogra
- School of Basic Sciences, Indian Institute of Technology-Mandi
| | - Debajyoti Das
- Division of Cell Biology and Physiology, Council of Scientific & Industrial Research-Indian Institute of Chemical Biology, Kolkata
| | - Sujay K Maity
- Division of Cell Biology and Physiology, Council of Scientific & Industrial Research-Indian Institute of Chemical Biology, Kolkata
| | - Avishek Paul
- Division of Cell Biology and Physiology, Council of Scientific & Industrial Research-Indian Institute of Chemical Biology, Kolkata
| | - Priya Rawat
- School of Basic Sciences, Indian Institute of Technology-Mandi
| | | | - Kausik Das
- Department of Hepatology, Institute of Post-Graduate Medical Education and Research and Seth Sukhlal Karnani Memorial Hospital, Kolkata, India
| | - Souveek Mitra
- Department of Hepatology, Institute of Post-Graduate Medical Education and Research and Seth Sukhlal Karnani Memorial Hospital, Kolkata, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, Council of Scientific & Industrial Research-Indian Institute of Chemical Biology, Kolkata
| | | |
Collapse
|
26
|
Ziarniak K, Yang T, Boycott C, Beetch M, Sassek M, Grzeda E, Ma Y, Sliwowska JH, Stefanska B. DNA hypermethylation of Kiss1r promoter and reduction of hepatic Kiss1r in female rats with type 2 diabetes. Epigenetics 2022; 17:2332-2346. [PMID: 36094166 PMCID: PMC9665141 DOI: 10.1080/15592294.2022.2119120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Kisspeptin, produced from the brain and peripheral tissues, may constitute an important link in metabolic regulation in response to external cues, such as diet. The kisspeptin system is well described in the brain. However, its function and regulation in the peripheral tissues, especially in relation to metabolic disease and sex differences, remain to be elucidated. As Kiss1 and Kiss1r, encoding for kisspeptin and kisspeptin receptors, respectively, are altered by overnutrition/fasting and regulated by DNA methylation during puberty and cancer, epigenetic mechanisms in metabolic disorders are highly probable. In the present study, we experimentally induced type 2 diabetes mellitus (DM2) in female Wistar rats using high-fat diet/streptozocin. We analysed expression and DNA methylation of Kiss1 and Kiss1r in the peripheral tissues, using quantitative-reverse-transcription PCR (qRT-PCR) and pyrosequencing. We discovered differential expression of Kiss1 and Kiss1r in peripheral organs in DM2 females, as compared with healthy controls, and the profile differed from patterns reported earlier in males. DM2 in females was linked to the increased Kiss1 mRNA in the liver and increased Kiss1r mRNA in the liver and adipose tissue. However, Kiss1r promoter was hypermethylated in the liver, suggesting gene silencing. Indeed, the increase in DNA methylation of Kiss1r promoter was accompanied by a reduction in Kiss1r protein, implying epigenetic or translational gene repression. Our results deliver novel evidence for tissue-specific differences in Kiss1 and Kiss1r expression in peripheral organs in DM2 females and suggest DNA methylation as a player in regulation of the hepatic kisspeptin system in DM2.
Collapse
Affiliation(s)
- Kamil Ziarniak
- Laboratory of Neurobiology, Department of Zoology, Faculty of Veterinary Medicine and Animal Sciences, Poznan University of Life Sciences, Poznan, Poland.,Molecular and Cell Biology Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Tony Yang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, Canada
| | - Cayla Boycott
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, Canada
| | - Megan Beetch
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, Canada
| | - Maciej Sassek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Poznan, Poland
| | - Emilia Grzeda
- Laboratory of Neurobiology, Department of Zoology, Faculty of Veterinary Medicine and Animal Sciences, Poznan University of Life Sciences, Poznan, Poland
| | - Yuexi Ma
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, Canada
| | - Joanna H Sliwowska
- Laboratory of Neurobiology, Department of Zoology, Faculty of Veterinary Medicine and Animal Sciences, Poznan University of Life Sciences, Poznan, Poland
| | - Barbara Stefanska
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
27
|
Masumi S, Lee EB, Dilower I, Upadhyaya S, Chakravarthi VP, Fields PE, Rumi MAK. The role of Kisspeptin signaling in Oocyte maturation. Front Endocrinol (Lausanne) 2022; 13:917464. [PMID: 36072937 PMCID: PMC9441556 DOI: 10.3389/fendo.2022.917464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Kisspeptins (KPs) secreted from the hypothalamic KP neurons act on KP receptors (KPRs) in gonadotropin (GPN) releasing hormone (GnRH) neurons to produce GnRH. GnRH acts on pituitary gonadotrophs to induce secretion of GPNs, namely follicle stimulating hormone (FSH) and luteinizing hormone (LH), which are essential for ovarian follicle development, oocyte maturation and ovulation. Thus, hypothalamic KPs regulate oocyte maturation indirectly through GPNs. KPs and KPRs are also expressed in the ovarian follicles across species. Recent studies demonstrated that intraovarian KPs also act directly on the KPRs expressed in oocytes to promote oocyte maturation and ovulation. In this review article, we have summarized published reports on the role of hypothalamic and ovarian KP-signaling in oocyte maturation. Gonadal steroid hormones regulate KP secretion from hypothalamic KP neurons, which in turn induces GPN secretion from the hypothalamic-pituitary (HP) axis. On the other hand, GPNs secreted from the HP axis act on the granulosa cells (GCs) and upregulate the expression of ovarian KPs. While KPs are expressed predominantly in the GCs, the KPRs are in the oocytes. Expression of KPs in the ovaries increases with the progression of the estrous cycle and peaks during the preovulatory GPN surge. Intrafollicular KP levels in the ovaries rise with the advancement of developmental stages. Moreover, loss of KPRs in oocytes in mice leads to failure of oocyte maturation and ovulation similar to that of premature ovarian insufficiency (POI). These findings suggest that GC-derived KPs may act on the KPRs in oocytes during their preovulatory maturation. In addition to the intraovarian role of KP-signaling in oocyte maturation, in vivo, a direct role of KP has been identified during in vitro maturation of sheep, porcine, and rat oocytes. KP-stimulation of rat oocytes, in vitro, resulted in Ca2+ release and activation of the mitogen-activated protein kinase, extracellular signal-regulated kinase 1 and 2. In vitro treatment of rat or porcine oocytes with KPs upregulated messenger RNA levels of the factors that favor oocyte maturation. In clinical trials, human KP-54 has also been administered successfully to patients undergoing assisted reproductive technologies (ARTs) for increasing oocyte maturation. Exogenous KPs can induce GPN secretion from hypothalamus; however, the possibility of direct KP action on the oocytes cannot be excluded. Understanding the direct in vivo and in vitro roles of KP-signaling in oocyte maturation will help in developing novel KP-based ARTs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
28
|
Socs3 ablation in kisspeptin cells partially prevents lipopolysaccharide-induced body weight loss. Cytokine 2022; 158:155999. [PMID: 35985175 DOI: 10.1016/j.cyto.2022.155999] [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: 03/15/2022] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/22/2022]
Abstract
Many cytokines have been proposed to regulate reproduction due to their actions on hypothalamic kisspeptin cells, the main modulators of gonadotropin-releasing hormone (GnRH) neurons. Hormones such as leptin, prolactin and growth hormone are good examples of cytokines that lead to Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway activation, consequently exerting effects in kisspeptin neurons. Different studies have investigated how specific components of the JAK/STAT signaling pathway affect the functions of kisspeptin cells, but the role of the suppressor of cytokine signaling 3 (SOCS3) in mediating cytokine actions in kisspeptin cells remains unknown. Cre-Loxp technology was used in the present study to ablate Socs3 expression in kisspeptin cells (Kiss1/Socs3-KO). Then, male and female control and Kiss1/Socs3-KO mice were evaluated for sexual maturation, energy homeostasis features, and fertility. It was found that hypothalamic Kiss1 mRNA expression is significantly downregulated in Kiss1/Socs3-KO mice. Despite reduced hypothalamic Kiss1 mRNA content, these mice did not present any sexual maturation or fertility impairments. Additionally, body weight gain, leptin sensitivity and glucose homeostasis were similar to control mice. Interestingly, Kiss1/Socs3-KO mice were partially protected against lipopolysaccharide (LPS)-induced body weight loss. Our results suggest that Socs3 ablation in kisspeptin cells partially prevents the sickness behavior induced by LPS, suggesting that kisspeptin cells can modulate energy metabolism in mice in certain situations.
Collapse
|
29
|
Guzman S, Dragan M, Kwon H, de Oliveira V, Rao S, Bhatt V, Kalemba KM, Shah A, Rustgi VK, Wang H, Bech PR, Abbara A, Izzi-Engbeaya C, Manousou P, Guo JY, Guo GL, Radovick S, Dhillo WS, Wondisford FE, Babwah AV, Bhattacharya M. Targeting hepatic kisspeptin receptor ameliorates nonalcoholic fatty liver disease in a mouse model. J Clin Invest 2022; 132:145889. [PMID: 35349482 PMCID: PMC9106350 DOI: 10.1172/jci145889] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/23/2022] [Indexed: 01/27/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most common liver disease, has become a silent worldwide pandemic. The incidence of NAFLD correlates with the rise in obesity, type 2 diabetes, and metabolic syndrome. A hallmark featureof NAFLD is excessive hepatic fat accumulation or steatosis, due to dysregulated hepatic fat metabolism, which can progress to nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. Currently, there are no approved pharmacotherapies to treat this disease. Here, we have found that activation of the kisspeptin 1 receptor (KISS1R) signaling pathway has therapeutic effects in NAFLD. Using high-fat diet-fed mice, we demonstrated that a deletion of hepatic Kiss1r exacerbated hepatic steatosis. In contrast, enhanced stimulation of KISS1R protected against steatosis in wild-type C57BL/6J mice and decreased fibrosis using a diet-induced mouse model of NASH. Mechanistically, we found that hepatic KISS1R signaling activates the master energy regulator, AMPK, to thereby decrease lipogenesis and progression to NASH. In patients with NAFLD and in high-fat diet-fed mice, hepatic KISS1/KISS1R expression and plasma kisspeptin levels were elevated, suggesting a compensatory mechanism to reduce triglyceride synthesis. These findings establish KISS1R as a therapeutic target to treat NASH.
Collapse
Affiliation(s)
- Stephania Guzman
- Department of Medicine, Robert Wood Johnson Medical School, and,Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, USA
| | | | - Hyokjoon Kwon
- Department of Medicine, Robert Wood Johnson Medical School, and
| | | | - Shivani Rao
- Department of Medicine, Robert Wood Johnson Medical School, and
| | - Vrushank Bhatt
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | | | - Ankit Shah
- Department of Medicine, Robert Wood Johnson Medical School, and
| | - Vinod K. Rustgi
- Department of Medicine, Robert Wood Johnson Medical School, and
| | - He Wang
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Paul R. Bech
- Section of Endocrinology and Investigative Medicine and
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine and
| | | | - Pinelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Jessie Y. Guo
- Department of Medicine, Robert Wood Johnson Medical School, and,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Grace L. Guo
- Department of Pharmacology and Toxicology, School of Pharmacy, and
| | - Sally Radovick
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | | | | | - Andy V. Babwah
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA.,Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Moshmi Bhattacharya
- Department of Medicine, Robert Wood Johnson Medical School, and,Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.,Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
30
|
Ma Y, Awe O, Radovick S, Yang X, Divall S, Wolfe A, Wu S. Lower FSH With Normal Fertility in Male Mice Lacking Gonadotroph Kisspeptin Receptor. Front Physiol 2022; 13:868593. [PMID: 35557961 PMCID: PMC9089166 DOI: 10.3389/fphys.2022.868593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/31/2022] [Indexed: 12/03/2022] Open
Abstract
The kisspeptin receptor, crucial for hypothalamic control of puberty and reproduction, is also present in the pituitary gland. Its role in the pituitary gland is not defined. Kisspeptin signaling via the Kiss1r could potentially regulate reproductive function at the level of pituitary gonadotrope. Using Cre/Lox technology, we deleted the Kiss1r gene in pituitary gonadotropes (PKiRKO). PKiRKO males have normal genital development (anogenital distance WT: 19.1 ± 0.4 vs. PKiRKO: 18.5 ± 0.4 mm), puberty onset, testes cell structure on gross histology, normal testes size, and fertility. PKiRKO males showed significantly decreased serum FSH levels compared to WT males (5.6 ± 1.9 vs. 10.2 ± 1.8 ng/ml) with comparable LH (1.1 ± 0.2 vs. 1.8 ± 0.4 ng/ml) and testosterone levels (351.8 ± 213.0 vs. 342.2 ± 183.0 ng/dl). PKiRKO females have normal puberty onset, cyclicity, LH and FSH levels and fertility. Overall, these findings indicate that absence of pituitary Kiss1r reduces FSH levels in male mice without affecting testis function. PKiRKO mice have normal reproductive function in both males and females.
Collapse
Affiliation(s)
- Yaping Ma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Olubusayo Awe
- Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sally Radovick
- Department of Pediatrics, Rutgers University Medical School, New Brunswick, NJ, United States
| | - Xiaofeng Yang
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Sara Divall
- Department of Pediatrics, University of Washington, Seattle's Children's Hospital, Seattle, United States
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sheng Wu
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cellular and Molecular Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
31
|
Lee EB, Dilower I, Marsh CA, Wolfe MW, Masumi S, Upadhyaya S, Rumi MAK. Sexual Dimorphism in Kisspeptin Signaling. Cells 2022; 11:1146. [PMID: 35406710 PMCID: PMC8997554 DOI: 10.3390/cells11071146] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Kisspeptin (KP) and kisspeptin receptor (KPR) are essential for the onset of puberty, development of gonads, and maintenance of gonadal function in both males and females. Hypothalamic KPs and KPR display a high degree of sexual dimorphism in expression and function. KPs act on KPR in gonadotropin releasing hormone (GnRH) neurons and induce distinct patterns of GnRH secretion in males and females. GnRH acts on the anterior pituitary to secrete gonadotropins, which are required for steroidogenesis and gametogenesis in testes and ovaries. Gonadal steroid hormones in turn regulate the KP neurons. Gonadal hormones inhibit the KP neurons within the arcuate nucleus and generate pulsatile GnRH mediated gonadotropin (GPN) secretion in both sexes. However, the numbers of KP neurons in the anteroventral periventricular nucleus and preoptic area are greater in females, which release a large amount of KPs in response to a high estrogen level and induce the preovulatory GPN surge. In addition to the hypothalamus, KPs and KPR are also expressed in various extrahypothalamic tissues including the liver, pancreas, fat, and gonads. There is a remarkable difference in circulating KP levels between males and females. An increased level of KPs in females can be linked to increased numbers of KP neurons in female hypothalamus and more KP production in the ovaries and adipose tissues. Although the sexually dimorphic features are well characterized for hypothalamic KPs, very little is known about the extrahypothalamic KPs. This review article summarizes current knowledge regarding the sexual dimorphism in hypothalamic as well as extrahypothalamic KP and KPR system in primates and rodents.
Collapse
Affiliation(s)
- Eun Bee Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Iman Dilower
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Courtney A. Marsh
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Saeed Masumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Sameer Upadhyaya
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Mohammad A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| |
Collapse
|
32
|
Hudson AD, Kauffman AS. Metabolic actions of kisspeptin signaling: Effects on body weight, energy expenditure, and feeding. Pharmacol Ther 2022; 231:107974. [PMID: 34530008 PMCID: PMC8884343 DOI: 10.1016/j.pharmthera.2021.107974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022]
Abstract
Kisspeptin (encoded by the Kiss1 gene) and its receptor, KISS1R (encoded by the Kiss1r gene), have well-established roles in stimulating reproduction via central actions on reproductive neural circuits, but recent evidence suggests that kisspeptin signaling also influences metabolism and energy balance. Indeed, both Kiss1 and Kiss1r are expressed in many metabolically-relevant peripheral tissues, including both white and brown adipose tissue, the liver, and the pancreas, suggesting possible actions on these tissues or involvement in their physiology. In addition, there may be central actions of kisspeptin signaling, or factors co-released from kisspeptin neurons, that modulate metabolic, feeding, or thermoregulatory processes. Accumulating data from animal models suggests that kisspeptin signaling regulates a wide variety of metabolic parameters, including body weight and energy expenditure, adiposity and adipose tissue function, food intake, glucose metabolism, respiratory rates, locomotor activity, and thermoregulation. Herein, the current evidence for the involvement of kisspeptin signaling in each of these physiological parameters is reviewed, gaps in knowledge identified, and future avenues of important research highlighted. Collectively, the discussed findings highlight emerging non-reproductive actions of kisspeptin signaling in metabolism and energy balance, in addition to previously documented roles in reproductive control, but also emphasize the need for more research to resolve current controversies and uncover underlying molecular and physiological mechanisms.
Collapse
Affiliation(s)
- Alexandra D Hudson
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Alexander S Kauffman
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| |
Collapse
|
33
|
Kavanagh GS, Tadi J, Balkenhol SM, Kauffman AS, Maloney SK, Smith JT. Kisspeptin impacts on circadian and ultradian rhythms of core body temperature: Evidence in kisspeptin receptor knockout and kisspeptin knockdown mice. Mol Cell Endocrinol 2022; 542:111530. [PMID: 34896241 PMCID: PMC9907773 DOI: 10.1016/j.mce.2021.111530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 01/26/2023]
Abstract
Kisspeptin is vital for the regulation of both fertility and metabolism. Kisspeptin receptor (Kiss1r) knockout (KO) mice exhibit increased adiposity and reduced energy expenditure in adulthood. Kiss1r mRNA is expressed in brown adipose tissue (BAT) and Kiss1r KO mice exhibit reduced Ucp1 mRNA in BAT and impaired thermogenesis. We hypothesised that mice with diminished kisspeptin signalling would exhibit reduced core body temperature (Tc) and altered dynamics of circadian and ultradian rhythms of Tc. Tc was recorded every 15-min over 14-days in gonadectomised wild-type (WT), Kiss1r KO, and also Kiss1-Cre (95% reduction in Kiss1 transcription) mice. Female Kiss1r KOs had higher adiposity and lower Ucp1 mRNA in BAT than WTs. No change was detected in Kiss1-Cre mice. Mean Tc during the dark phase was lower in female Kiss1r KOs versus WTs, but not Kiss1-Cre mice. Female Kiss1r KOs had a lower mesor and amplitude of the circadian rhythm of Tc than did WTs. In WT mice, there were more episodic ultradian events (EUEs) of Tc during the dark phase than the light phase, but this measure was similar between dark and light phases in Kiss1r KO and Kiss1-Cre mice. The amplitude of EUEs was higher in the dark phase in female Kiss1r KO and male Kiss1-Cre mice. Given the lack of clear metabolic phenotype in Kiss1-Cre mice, 5% of Kiss1 transcription may be sufficient for proper metabolic control, as was shown for fertility. Moreover, the observed alterations in Tc suggest that kisspeptin has a role in circadian and ultradian rhythm-driven pathways.
Collapse
Affiliation(s)
- Georgia S Kavanagh
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Jason Tadi
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Sydney M Balkenhol
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Shane K Maloney
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Jeremy T Smith
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia.
| |
Collapse
|
34
|
Sivalingam M, Ogawa S, Trudeau VL, Parhar IS. Conserved functions of hypothalamic kisspeptin in vertebrates. Gen Comp Endocrinol 2022; 317:113973. [PMID: 34971635 DOI: 10.1016/j.ygcen.2021.113973] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022]
Abstract
Hypothalamic kisspeptin encoded by KISS1/Kiss1 gene emerged as a regulator of the reproductive axis in mammals following the discovery of the kisspeptin receptor (Kissr) and its role in reproduction. Kisspeptin-Kissr systems have been investigated in various vertebrates, and a conserved sequence of kisspeptin-Kissr has been identified in most vertebrate species except in the avian linage. In addition, multiple paralogs of kisspeptin sequences have been identified in the non-mammalian vertebrates. The allegedly conserved role of kisspeptin-Kissr in reproduction became debatable when kiss/kissr genes-deficient zebrafish and medaka showed no apparent effect on the onset of puberty, sexual development, maturation and reproductive capacity. Therefore, it is questionable whether the role of kisspeptin in reproduction is conserved among vertebrate species. Here we discuss from a comparative and evolutional aspect the diverse functions of kisspeptin and its receptor in vertebrates. Primarily this review focuses on the role of hypothalamic kisspeptin in reproductive and non-reproductive functions that are conserved in vertebrate species.
Collapse
Affiliation(s)
- Mageswary Sivalingam
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia
| | - Satoshi Ogawa
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Ishwar S Parhar
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia.
| |
Collapse
|
35
|
Pan A, Sun XM, Huang FQ, Liu JF, Cai YY, Wu X, Alolga RN, Li P, Liu BL, Liu Q, Qi LW. The mitochondrial β-oxidation enzyme HADHA restrains hepatic glucagon response by promoting β-hydroxybutyrate production. Nat Commun 2022; 13:386. [PMID: 35046401 PMCID: PMC8770464 DOI: 10.1038/s41467-022-28044-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 01/06/2022] [Indexed: 11/09/2022] Open
Abstract
Disordered hepatic glucagon response contributes to hyperglycemia in diabetes. The regulators involved in glucagon response are less understood. This work aims to investigate the roles of mitochondrial β-oxidation enzyme HADHA and its downstream ketone bodies in hepatic glucagon response. Here we show that glucagon challenge impairs expression of HADHA. Liver-specific HADHA overexpression reversed hepatic gluconeogenesis in mice, while HADHA knockdown augmented glucagon response. Stable isotope tracing shows that HADHA promotes ketone body production via β-oxidation. The ketone body β-hydroxybutyrate (BHB) but not acetoacetate suppresses gluconeogenesis by selectively inhibiting HDAC7 activity via interaction with Glu543 site to facilitate FOXO1 nuclear exclusion. In HFD-fed mice, HADHA overexpression improved metabolic disorders, and these effects are abrogated by knockdown of BHB-producing enzyme. In conclusion, BHB is responsible for the inhibitory effect of HADHA on hepatic glucagon response, suggesting that HADHA activation or BHB elevation by pharmacological intervention hold promise in treating diabetes.
Collapse
Affiliation(s)
- An Pan
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiao-Meng Sun
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng-Qing Huang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jin-Feng Liu
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuan-Yuan Cai
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin Wu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Raphael N Alolga
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Bao-Lin Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qun Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China.
| | - Lian-Wen Qi
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
36
|
Evans RM, Wei Z. Interorgan crosstalk in pancreatic islet function and pathology. FEBS Lett 2022; 596:607-619. [PMID: 35014695 DOI: 10.1002/1873-3468.14282] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022]
Abstract
Pancreatic β cells secrete insulin in response to glucose, a process that is regulated at multiple levels, including a network of input signals from other organ systems. Impaired islet function contributes to the pathogenesis of type 2 diabetes mellitus (T2DM), and targeting inter-organ communications, such as GLP-1 signalling, to enhance β-cell function has been proven to be a successful therapeutic strategy in the last decade. In this review, we will discuss recent advances in inter-organ communication from the metabolic, immune and neural system to pancreatic islets, their biological implication in normal pancreas endocrine function and their role in the (mal)adaptive responses of islet to nutrition-induced stress.
Collapse
Affiliation(s)
- Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zong Wei
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, USA
| |
Collapse
|
37
|
Fernández-Millán E, Guillén C. Multi-Organ Crosstalk with Endocrine Pancreas: A Focus on How Gut Microbiota Shapes Pancreatic Beta-Cells. Biomolecules 2022; 12:biom12010104. [PMID: 35053251 PMCID: PMC8773909 DOI: 10.3390/biom12010104] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes (T2D) results from impaired beta-cell function and insufficient beta-cell mass compensation in the setting of insulin resistance. Current therapeutic strategies focus their efforts on promoting the maintenance of functional beta-cell mass to ensure appropriate glycemic control. Thus, understanding how beta-cells communicate with metabolic and non-metabolic tissues provides a novel area for investigation and implicates the importance of inter-organ communication in the pathology of metabolic diseases such as T2D. In this review, we provide an overview of secreted factors from diverse organs and tissues that have been shown to impact beta-cell biology. Specifically, we discuss experimental and clinical evidence in support for a role of gut to beta-cell crosstalk, paying particular attention to bacteria-derived factors including short-chain fatty acids, lipopolysaccharide, and factors contained within extracellular vesicles that influence the function and/or the survival of beta cells under normal or diabetogenic conditions.
Collapse
Affiliation(s)
- Elisa Fernández-Millán
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
38
|
Dana P, Hayati Roodbari N, Yaghmaei P, Hajebrahimi Z. Effects of empagliflozin on the expression of kisspeptin gene and reproductive system function in streptozotocin-induced diabetic male rats. Front Endocrinol (Lausanne) 2022; 13:1059942. [PMID: 36479221 PMCID: PMC9719967 DOI: 10.3389/fendo.2022.1059942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/25/2022] [Indexed: 11/22/2022] Open
Abstract
One of the main health concerns of diabetes is testicular dysfunction and impairment of reproductive function and sperm quality which can cause male infertility. kisspeptin is a hypothalamic neuropeptide hormone that is involved in the regulation of energy metabolism, gonadotrophin-releasing hormone (GnRH), and reproductive function. In the present study, the therapeutic effects of empagliflozin (sodium-glucose co-transporter 2 inhibitors) on kisspeptin expression along with reproductive function were investigated in diabetic male Wistar rats. Diabetes was induced by a single dose injection of 60 mg/kg streptozotocin. Empagliflozin in doses of 10 and 25 mg/kg body weight was used for 8 weeks. Serum samples, testis, epididymis, and pancreas tissues were collected at the end of the experiments. Lipid profiles, oxidative stress markers, blood hormones, expression of kisspeptin along with pathological alterations of the testis were assayed using real-time PCR, biochemical, and histological technics. Data have shown that empagliflozin improved hyperglycemia, reproductive impairment, oxidative stress condition, and histopathological alterations of pancreatic and testis tissues in diabetic animals. It improved the serum levels of sex hormones, insulin, leptin, and the expression of kisspeptin in the testes tissues. Spermatogenesis is also improved in treated animals. Data indicated that the administration of empagliflozin can ameliorate symptoms of diabetes. It probably has promising antidiabetic potential and may improve the male infertility of diabetic subjects. To our knowledge, this is the first experimental evidence for the potential impact of empagliflozin on kisspeptin expression in diabetic male rats.
Collapse
Affiliation(s)
- Parisa Dana
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nasim Hayati Roodbari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- *Correspondence: Nasim Hayati Roodbari,
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Hajebrahimi
- A&S Research Institute, Ministry of Science Research and Technology, Tehran, Iran
| |
Collapse
|
39
|
Kirsch P, Kunadia J, Shah S, Agrawal N. Metabolic effects of prolactin and the role of dopamine agonists: A review. Front Endocrinol (Lausanne) 2022; 13:1002320. [PMID: 36246929 PMCID: PMC9562454 DOI: 10.3389/fendo.2022.1002320] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Prolactin is a polypeptide hormone that is well known for its role in reproductive physiology. Recent studies highlight its role in neurohormonal appetite regulation and metabolism. Elevated prolactin levels are widely associated with worsening metabolic disease, but it appears that low prolactin levels could also be metabolically unfavorable. This review discusses the pathophysiology of prolactin related metabolic changes, and the less commonly recognized effects of prolactin on adipose tissue, pancreas, liver, and small bowel. Furthermore, the effect of dopamine agonists on the metabolic profiles of patients with hyperprolactinemia are discussed as well.
Collapse
Affiliation(s)
- Polly Kirsch
- New York University (NYU) Grossman School of Medicine, NYU Langone Health, New York, NY, United States
| | - Jessica Kunadia
- Department of Medicine, NYU Langone Health, New York, NY, United States
| | - Shruti Shah
- New York University (NYU) Grossman School of Medicine, NYU Langone Health, New York, NY, United States
| | - Nidhi Agrawal
- Department of Medicine, NYU Langone Health, New York, NY, United States
- *Correspondence: Nidhi Agrawal,
| |
Collapse
|
40
|
Ku EJ, Lee DH, Jeon HJ, Oh TK. Long-term effectiveness and safety of quadruple combination therapy with empagliflozin versus dapagliflozin in patients with type 2 diabetes: 3-year prospective observational study. Diabetes Res Clin Pract 2021; 182:109123. [PMID: 34740742 DOI: 10.1016/j.diabres.2021.109123] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/18/2021] [Accepted: 10/31/2021] [Indexed: 10/19/2022]
Abstract
AIMS To investigate the long-term effectiveness and safety of two distinct sodium-glucose co-transporter 2 (SGLT2) inhibitors, empagliflozin and dapagliflozin, in inadequately controlled type 2 diabetes (T2D) despite a combined administration of metformin, glimepiride and dipeptidyl peptidase-4 inhibitor. METHODS A total of 362 patients with T2D were enrolled for this 3-year open-label, prospective observational study. Empagliflozin (25 mg/day, n = 185) or dapagliflozin (10 mg/day, n = 177) was added to the existing triple drug regimen. HbA1c, fasting plasma glucose (FPG), body weight, and other cardiometabolic variables and adverse events were evaluated. RESULTS At 3 years, changes in HbA1c and FPG were -1.7% (standard error [SE] 0.10) and -60.0 mg/dL(2.2), and -1.1%(0.12) and -48.1 mg/dL(3.6), for empagliflozin and dapagliflozin group, respectively (P = 0.001 and P = 0.055). Empagliflozin group showed significantly greater body weight reduction (-4.5 kg [SE 0.35] vs. -1.0 kg [SE 0.40], P = 0.024) and had beneficial effects on HDL cholesterol and LDL cholesterol (both P < 0.05). The overall incidence of adverse events, cardiovascular events and mortality did not differ between the two groups. CONCLUSIONS Quadruple combination therapy with either empagliflozin or dapagliflozin showed a positive long-term effect in the glycemic control and body weight reduction with generally well tolerance. In general, the use of empagliflozin performed better than dapagliflozin. Clinical Trial Number NCT03748810 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Eu Jeong Ku
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Dong-Hwa Lee
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Hyun Jeong Jeon
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Tae Keun Oh
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Republic of Korea.
| |
Collapse
|
41
|
Deletion of Stim1 in Hypothalamic Arcuate Nucleus Kiss1 Neurons Potentiates Synchronous GCaMP Activity and Protects against Diet-Induced Obesity. J Neurosci 2021; 41:9688-9701. [PMID: 34654752 DOI: 10.1523/jneurosci.0622-21.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/29/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Kisspeptin (Kiss1) neurons are essential for reproduction, but their role in the control of energy balance and other homeostatic functions remains unclear. High-frequency firing of hypothalamic arcuate Kiss1 (Kiss1ARH) neurons releases kisspeptin into the median eminence, and neurokinin B (NKB) and dynorphin onto neighboring Kiss1ARH neurons to generate a slow EPSP mediated by TRPC5 channels that entrains intermittent, synchronous firing of Kiss1ARH neurons. High-frequency optogenetic stimulation of Kiss1ARH neurons also releases glutamate to excite the anorexigenic proopiomelanocortin (POMC) neurons and inhibit the orexigenic neuropeptide Y/agouti-related peptide (AgRP) neurons via metabotropic glutamate receptors. At the molecular level, the endoplasmic reticulum (ER) calcium-sensing protein stromal interaction molecule 1 (STIM1) is critically involved in the regulation of neuronal Ca2+ signaling and neuronal excitability through its interaction with plasma membrane (PM) calcium (e.g., TRPC) channels. Therefore, we hypothesized that deletion of Stim1 in Kiss1ARH neurons would increase neuronal excitability and their synchronous firing, which ultimately would affect energy homeostasis. Using optogenetics in combination with whole-cell recording and GCaMP6 imaging in slices, we discovered that deletion of Stim1 in Kiss1 neurons significantly increased the amplitude and duration of the slow EPSP and augmented synchronous [Ca2+]i oscillations in Kiss1ARH neurons. Deletion of Stim1 in Kiss1ARH neurons amplified the actions of NKB and protected ovariectomized female mice from developing obesity and glucose intolerance with high-fat dieting (HFD). Therefore, STIM1 appears to play a critical role in regulating synchronous firing of Kiss1ARH neurons, which ultimately affects the coordination between energy homeostasis and reproduction.SIGNIFICANCE STATEMENT Hypothalamic arcuate kisspeptin (Kiss1ARH) neurons are essential for stimulating the pulsatile release of gonadotropin-releasing hormone (GnRH) and maintaining fertility. However, Kiss1ARH neurons appear to be a key player in coordinating energy balance with reproduction. The regulation of calcium channels and hence calcium signaling is critically dependent on the endoplasmic reticulum (ER) calcium-sensing protein stromal interaction molecule 1 (STIM1), which interacts with the plasma membrane (PM) calcium channels. We have conditionally deleted Stim1 in Kiss1ARH neurons and found that it significantly increased the excitability of Kiss1ARH neurons and protected ovariectomized female mice from developing obesity and glucose intolerance with high-fat dieting (HFD).
Collapse
|
42
|
Rong B, Wu Q, Saeed M, Sun C. Gut microbiota-a positive contributor in the process of intermittent fasting-mediated obesity control. ACTA ACUST UNITED AC 2021; 7:1283-1295. [PMID: 34786501 PMCID: PMC8567329 DOI: 10.1016/j.aninu.2021.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/12/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022]
Abstract
Historically, intermittent fasting (IF) has been considered as an effective strategy for controlling the weight of athletes before competition. Along with excellent insight into its application in various spaces by numerous studies, increasing IF-mediated positive effects have been reported, including anti-aging, neuroprotection, especially obesity control. Recently, the gut microbiota has been considered as an essential manipulator for host energy metabolism and its structure has been reported to be sensitive to dietary structure and habits, indicating that there is a potential and strong association between IF and gut microbiota. In this paper, we focus on the crosstalk between these symbionts and energy metabolism during IF which hold the promise to optimize host energy metabolism at various physical positions, including adipose tissue, liver and intestines, and further improve milieu internal homeostasis. Moreover, this paper also discusses the positive function of a potential recommendatory strain (Akkermansia muciniphila) based on the observational data for IF-mediated alternated pattern of gut microbiota and a hopefully regulatory pathway (circadian rhythm) for gut microbiota in IF-involved improvement on host energy metabolism. Finally, this review addresses the limitation and perspective originating from these studies, such as the association with tissue-specific bio-clock and single strain research, which may continuously reveal novel viewpoints and mechanisms to understand the energy metabolism and develop new strategies for treating obesity, diabetes, and metabolic disorders.
Collapse
Affiliation(s)
- Bohan Rong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Qiong Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.,Department of Pathophysiology, Qinghai University Medical College, Xining, Qinghai, China
| | - Muhammad Saeed
- Faculty of Animal Production & Technology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63100, Pakistan
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
43
|
Menghoum N, Oriot P, Hermans MP. Clinical and biochemical characteristics and analysis of risk factors for euglycaemic diabetic ketoacidosis in type 2 diabetic individuals treated with SGLT2 inhibitors: A review of 72 cases over a 4.5-year period. Diabetes Metab Syndr 2021; 15:102275. [PMID: 34562870 DOI: 10.1016/j.dsx.2021.102275] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS To study euglycemic diabetic ketoacidosis (euDKA) outcomes associated with sodium-glucose co-transporter 2 inhibitors (SGLT2is) METHODS: Review of 72 euDKA cases in T2DM between September 2015 and January 2020 (PUBMED). RESULTS euDKA could occur at any time during SGLT2is treatment, with nausea, abdominal pain and vomiting as main symptoms. Hyperglycemia did not correlate with pH and β-hydroxybutyrates. Low pH and high β-hydroxybutyrates were significantly associated with euDKA. In biguanides users, acidosis was unrelated to lactic acidosis. euDKA occurred during fasting, surgery, acute infection, insulin deprivation (endogenous or exogenous). CONCLUSIONS These data support avoidance of euDKA risk states in SGLT2i users.
Collapse
Affiliation(s)
- N Menghoum
- Department of Internal Medicine, Mouscron Hospital Center, Avenue de Fécamp 49, 7700, Mouscron, Belgium; Pole of Cardiovascular Research, Institute of Experimental and Clinical Research, Catholic University of Louvain, 1200, Brussels, Belgium
| | - P Oriot
- Department of Internal Medicine, Mouscron Hospital Center, Avenue de Fécamp 49, 7700, Mouscron, Belgium; Department of Diabetology, Mouscron Hospital Centre, Avenue de Fécamp 49, 7700, Mouscron, Belgium.
| | - M P Hermans
- Endocrinology & Nutrition Unit, Cliniques Universitaires St-Luc, and Pole of Cardiovascular Research, Institute of Experimental and Clinical Research, Catholic University of Louvain, Avenue Hippocrate 55, 1200, Brussels, Belgium
| |
Collapse
|
44
|
Fang P, She Y, Zhao J, Yan J, Yu X, Jin Y, Wei Q, Zhang Z, Shang W. Emerging roles of kisspeptin/galanin in age-related metabolic disease. Mech Ageing Dev 2021; 199:111571. [PMID: 34517021 DOI: 10.1016/j.mad.2021.111571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/08/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022]
Abstract
Age is a major risk factor for developing metabolic diseases such as obesity and diabetes. There is an unprecedented rise in obesity and type 2 diabetes in recent decades. A convincing majority of brain-gut peptides are associated with a higher risk to develop metabolic disorders, and may contribute to the pathophysiology of age-related metabolic diseases. Accumulating basic studies revealed an intriguing role of kisspeptin and galanin involved in the amelioration of insulin resistance in different ways. In patients suffered from obesity and diabetes a significant, sex-related changes in the plasma kisspeptin and galanin levels occurred. Kisspeptin is anorexigenic to prevent obesity, its level is negatively correlative with obesity and insulin resistance. While galanin is appetitive to stimulate food intake and body weight, its level is positively correlative with obesity, HOMA-IR and glucose/triglyceride concentration. In turn, kisspeptin and galanin also distinctly increase glucose uptake and utilization as well as energy expenditure. This article reviews recent evidence dealing with the role of kisspeptin and galanin in the pathophysiology of age-related metabolic diseases. It should be therefore taken into account that the targeted modulation of those peptidergic signaling may be potentially helpful in the future treatment of age-related metabolic diseases.
Collapse
Affiliation(s)
- Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, 225300, China.
| | - Yuqing She
- Department of Endocrinology, Pukou Branch of Jiangsu People's Hospital, Nanjing, 210023, China
| | - Juan Zhao
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xizhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu Jin
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qingbo Wei
- Key Laboratory of Acupuncture and Medicine Research of Minister of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, 225001, China.
| | - Wenbin Shang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
45
|
Schaefer J, Vilos AG, Vilos GA, Bhattacharya M, Babwah AV. Uterine kisspeptin receptor critically regulates epithelial estrogen receptor α transcriptional activity at the time of embryo implantation in a mouse model. Mol Hum Reprod 2021; 27:gaab060. [PMID: 34524460 PMCID: PMC8786495 DOI: 10.1093/molehr/gaab060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/07/2021] [Indexed: 12/14/2022] Open
Abstract
Embryo implantation failure is a major cause of infertility in women of reproductive age and a better understanding of uterine factors that regulate implantation is required for developing effective treatments for female infertility. This study investigated the role of the uterine kisspeptin receptor (KISS1R) in the molecular regulation of implantation in a mouse model. To conduct this study, a conditional uterine knockout (KO) of Kiss1r was created using the Pgr-Cre (progesterone receptor-CRE recombinase) driver. Reproductive profiling revealed that while KO females exhibited normal ovarian function and mated successfully to stud males, they exhibited significantly fewer implantation sites, reduced litter size and increased neonatal mortality demonstrating that uterine KISS1R is required for embryo implantation and a healthy pregnancy. Strikingly, in the uterus of Kiss1r KO mice on day 4 (D4) of pregnancy, the day of embryo implantation, KO females exhibited aberrantly elevated epithelial ERα (estrogen receptor α) transcriptional activity. This led to the temporal misexpression of several epithelial genes [Cftr (Cystic fibrosis transmembrane conductance regulator), Aqp5 (aquaporin 5), Aqp8 (aquaporin 8) and Cldn7 (claudin 7)] that mediate luminal fluid secretion and luminal opening. As a result, on D4 of pregnancy, the lumen remained open disrupting the final acquisition of endometrial receptivity and likely accounting for the reduction in implantation events. Our data clearly show that uterine KISS1R negatively regulates ERα signaling at the time of implantation, in part by inhibiting ERα overexpression and preventing detrimentally high ERα activity. To date, there are no reports on the regulation of ERα by KISS1R; therefore, this study has uncovered an important and powerful regulator of uterine ERα during early pregnancy.
Collapse
Affiliation(s)
- Jennifer Schaefer
- Laboratory of Human Growth and Reproductive Development, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- School of Graduate Studies, Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Angelos G Vilos
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - George A Vilos
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Moshmi Bhattacharya
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, USA
| | - Andy V Babwah
- Laboratory of Human Growth and Reproductive Development, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- School of Graduate Studies, Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
46
|
Calcaterra V, Nappi RE, Pelizzo G, De Silvestri A, Albertini R, De Amici M, Tenuta E, Vinci F, Mameli C, Zuccotti G. Insulin resistance and potential modulators of ovarian reserve in young reproductive-aged women with obesity and type 1 diabetes. Gynecol Endocrinol 2021; 37:823-830. [PMID: 34137355 DOI: 10.1080/09513590.2021.1940127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Both obesity and diabetes play a significant role in reproductive disorders in women and insulin resistance (IR) is a confirmed trait d'union. We evaluated the relationship between IR and an established ovarian reserve biomarker such as anti-mullerian hormone (AMH) together with other potential modulators of ovarian physiology (adiponectin and kisspeptin) in young reproductive-aged group women with obesity and type 1 diabetes (T1D). PATIENTS AND METHODS We recruited 32 female youths: 14 of them presented with T1D (14.6 ± 2.6 years) and 18 with obesity (15.1 ± 2.6 years). The control group included 20 age-matched normal weight females. Each patient underwent physical examination and hormonal assessment. AMH, kisspeptin and adiponectin levels were also measured. IR was calculated as the homeostasis model assessment for insulin resistance (HOMA-IR) and the glucose disposal rate (eGDR) in patients with obesity and with T1D, respectively. RESULTS adiponectin and kisspeptin levels were significantly different into groups (p ≤ .001), whereas AMH levels were not. Adiponectin values were higher in controls compared to patients with obesity (p < .001) and T1D (p = .02). Kisspeptin levels were lower in controls compared to patients with obesity (p = .001), without reaching statistical significance when compared to T1D (p = .06). IR was associated with lower adiponectin and higher kisspeptin levels (p < .001 and p = .02, respectively), but not with AMH. CONCLUSIONS IR displays a relationship with adiponectin and kisspeptin in young reproductive-aged women with obesity and T1D. Interventions to correct IR in adolescents could be part of an early approach to prevent reproductive disorders and to promote factors associated with longevity in adult women.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Pediatric and Adolescent Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
- Department of Pediatrics, "Vittore Buzzi" Children's Hospital, Milano, Italy
| | - Rossella E Nappi
- Research Center for Reproductive Medicine, Gynecological Endocrinology and Menopause, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Gloria Pelizzo
- Pediatric Surgery Department, "Vittore Buzzi" Children's Hospital, Milano, Italy
- Department of Biomedical and Clinical Science "L. Sacco", University of Milano, Milano, Italy
| | - Annalisa De Silvestri
- Biometry and Clinical Epidemiology, Scientific Direction, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Riccardo Albertini
- Laboratory of Clinical Chemistry, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mara De Amici
- Laboratory of Clinical Chemistry, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Pediatric Unit, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Elisavietta Tenuta
- Pediatric and Adolescent Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Federica Vinci
- Pediatric Unit, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Chiara Mameli
- Department of Pediatrics, "Vittore Buzzi" Children's Hospital, Milano, Italy
- Department of Biomedical and Clinical Science "L. Sacco", University of Milano, Milano, Italy
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, "Vittore Buzzi" Children's Hospital, Milano, Italy
- Department of Biomedical and Clinical Science "L. Sacco", University of Milano, Milano, Italy
| |
Collapse
|
47
|
Geronikolou SA, Pavlopoulou A, Cokkinos DV, Bacopoulou F, Chrousos GP. Polycystic οvary syndrome revisited: An interactions network approach. Eur J Clin Invest 2021; 51:e13578. [PMID: 33955010 DOI: 10.1111/eci.13578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/26/2021] [Accepted: 04/19/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND The polycystic ovary syndrome (PCOS) has genetic, epigenetic, metabolic and reproductive aspects, while its complex pathophysiology has not been conclusively deciphered. AIM The goal of this research was to screen the gene/gene products associated with PCOS and to predict any possible interactions with the highest possible fidelity. MATERIALS AND METHODS STRING v10.5 database and a confidence level of 0.7 were used. RESULTS A highly interconnected network of 48 nodes was created, where insulin (INS) appears to be the major hub. INS upstream and downstream defects were analysed and revealed that only the kisspeptin- and glucagon-coding genes were upstream of INS. CONCLUSION A metabolic dominance was inferred and discussed herein with its implications in puberty, obesity, infertility and cardiovascular function. This study, thus, may contribute to the resolution of a scientific conflict between the USA and EU definitions of the syndrome and/or provide a new P4 medicine approach.
Collapse
Affiliation(s)
- Styliani A Geronikolou
- Clinical, Translational & Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,University Research Institute of Maternal & Child Health & Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Dennis V Cokkinos
- Clinical, Translational & Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Flora Bacopoulou
- University Research Institute of Maternal & Child Health & Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Chrousos
- Clinical, Translational & Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,University Research Institute of Maternal & Child Health & Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
48
|
Yeoh HL, Lee M, Pan WJ, Ong HY. Case of sodium-glucose cotransporter-2 inhibitor-associated euglycaemic diabetic ketoacidosis. BMJ Case Rep 2021; 14:e235953. [PMID: 34404640 PMCID: PMC8375747 DOI: 10.1136/bcr-2020-235953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Following non-elective orthopaedic surgery, a 61-year-old man with poorly controlled type 2 diabetes mellitus on empagliflozin developed high anion gap metabolic acidosis in the high-dependency unit. Metabolic acidosis persisted despite intravenous sodium bicarbonate, contributing to tachycardia and a run of non-sustained ventricular tachycardia. He was euglycaemic throughout hospital admission. Investigations revealed elevated urine and capillary ketones, and a diagnosis of sodium-glucose cotransporter-2 inhibitor-associated euglycaemic diabetic ketoacidosis was made. He was treated with an intravenous sliding scale insulin infusion and concurrent dextrose 5% with potassium chloride. Within 24 hours of treatment, his arterial pH, anion gap and serum bicarbonate levels normalised. After a further 12 hours, the intravenous insulin infusion was converted to a basal/bolus regimen of subcutaneous insulin, and he was transferred to the general ward. He was discharged well on subcutaneous insulin 6 days postoperatively.
Collapse
Affiliation(s)
- Huei Li Yeoh
- Pharmacy Department, Mount Elizabeth Novena Hospital, Singapore
| | | | | | | |
Collapse
|
49
|
Palmer BF, Clegg DJ. Euglycemic Ketoacidosis as a Complication of SGLT2 Inhibitor Therapy. Clin J Am Soc Nephrol 2021; 16:1284-1291. [PMID: 33563658 PMCID: PMC8455044 DOI: 10.2215/cjn.17621120] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors are drugs designed to lower plasma glucose concentration by inhibiting Na+-glucose-coupled transport in the proximal tubule. Clinical trials demonstrate these drugs have favorable effects on cardiovascular outcomes to include slowing the progression of CKD. Although most patients tolerate these drugs, a potential complication is development of ketoacidosis, often with a normal or only a minimally elevated plasma glucose concentration. Inhibition of sodium-glucose cotransporter-2 in the proximal tubule alters kidney ATP turnover so that filtered ketoacids are preferentially excreted as Na+ or K+ salts, leading to indirect loss of bicarbonate from the body and systemic acidosis under conditions of increased ketogenesis. Risk factors include reductions in insulin dose, increased insulin demand, metabolic stress, low carbohydrate intake, women, and latent autoimmune diabetes of adulthood. The lack of hyperglycemia and nonspecific symptoms of ketoacidosis can lead to delays in diagnosis. Treatment strategies and various precautions are discussed that can decrease the likelihood of this complication.
Collapse
Affiliation(s)
- Biff F. Palmer
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Deborah J. Clegg
- Associate Dean for Research, College of Nursing and Health Professionals, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
50
|
Gorkem U, Kan O, Bostanci MO, Taskiran D, Inal HA. Kisspeptin and Hematologic Parameters as Predictive Biomarkers for First-Trimester Abortions. Medeni Med J 2021; 36:98-105. [PMID: 34239761 PMCID: PMC8226412 DOI: 10.5222/mmj.2021.32549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/04/2021] [Indexed: 11/28/2022] Open
Abstract
Objective Spontaneous abortion is the most common complication of early pregnancy, affecting up to 20% of recognized pregnancies. Kisspeptin is predominantly released by placental syncytiotrophoblasts, and regulates their placental invasion into the uterine matrices. We aimed to establish an association of serum kisspeptin levels with pregnancy outcomes during the early gestational stage of the first trimester. Method In this prospective study, 90 pregnant women in their 7 to 8 6/7 gestational weeks were classified into three groups: (i) The control group, consisting of healthy pregnant women (n=30), (ii) the threatened abortion group (n=30), and (iii) the spontaneous abortion group (n=30). The maternal serum samples were analyzed for complete blood count parameters and kisspeptin levels. Results There was no statistical difference regarding body mass index (BMI) and gestational age (p=0.370). Regarding detailed obstetric notations, including gravida, parity, abortion, and living children, socioeconomic levels, and employment rates, all study groups were comparable (p>0.05, for all). No significant association was found regarding the biochemical parameters of complete blood count, including neutrophil, lymphocyte, and platelet concentrations, as well as neutrophil-to-lymphocyte ratios (NLR) and platelet-to-lymphocyte ratios (PLR) (p>0.05, for all). The median serum kisspeptin levels of the study groups did not differ between the groups (p=0.153). Correlation analysis revealed no significant relationship between serum kisspeptin levels and other study parameters in any study groups (p>0.05, for all) Conclusion We found no statistically significant relationship between serum kisspeptin concentrations and pregnancy outcomes in the early gestational stage of the first trimester, and serum kisspeptin concentrations did not seem to be a reliable marker to distinguish abortion status from viable pregnancy.
Collapse
Affiliation(s)
- Umit Gorkem
- Hitit University, Medical Faculty, Department of Obstetric and Gynecology, Corum, Turkey
| | - Ozgur Kan
- Hitit University Medical Faculty, Departmant of Obstetric and Gynecology, Corum, Turkey
| | | | - Deniz Taskiran
- Hitit University Medical Faculty, Departmant of Obstetric and Gynecology, Corum, Turkey
| | - Hasan Ali Inal
- Konya Training and Research Hospital, Departmant of Obstetric and Gynecology, Konya, Turkey
| |
Collapse
|