1
|
Sola-Leyva A, Pathare ADS, Apostolov A, Aleksejeva E, Kask K, Tammiste T, Ruiz-Durán S, Risal S, Acharya G, Salumets A. The hidden impact of GLP-1 receptor agonists on endometrial receptivity and implantation. Acta Obstet Gynecol Scand 2024. [PMID: 39696822 DOI: 10.1111/aogs.15010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
Increasing infertility rates represent a growing medical challenge in modern societies resulting from a complex interplay of sociocultural trends, lifestyle factors, exposure to environmental toxins, and underlying health problems. Women's fertility is particularly vulnerable to these shifts. The obesogenic lifestyle not only accelerates weight gain, but also disrupts ovulation driving the rise in infertility. Among several medications used for treating obesity and type 2 diabetes, glucagon-like peptide-1 receptor agonists (GLP-1RAs) show promising improvement in female fertility most likely by stimulating ovulation. However, the effects of GLP-1RAs on the endometrium remain unclear. Further studies are needed to investigate the impact of GLP-1RAs on endometrial receptivity and embryo implantation and early development. The aim of this study is to address the knowledge gap regarding the effects of GLP-1RAs on human reproduction, with special focus on the endometrium. Understanding these mechanisms may help to develop new strategies for improving fertility treatment, reduce implantation failure and address potential safety concerns regarding teratogenicity and adverse developmental outcomes for children born to women conceiving during or soon after GLP-1RA treatment.
Collapse
Affiliation(s)
- Alberto Sola-Leyva
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Celvia CC, Competence Centre on Health Technologies, Tartu, Estonia
| | | | - Apostol Apostolov
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Celvia CC, Competence Centre on Health Technologies, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Elina Aleksejeva
- Celvia CC, Competence Centre on Health Technologies, Tartu, Estonia
| | - Keiu Kask
- Celvia CC, Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Triin Tammiste
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- West Tallinn Central Hospital, Women's Clinic, Tallinn, Estonia
| | - Susana Ruiz-Durán
- Department of Obstetrics and Gynecology, Virgen de las Nieves University Hospital, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Sanjiv Risal
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ganesh Acharya
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Center for Fetal Medicine Karolinska, University Hospital, Stockholm, Sweden
- Women's Health and Perinatology Research Group, Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Andres Salumets
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Celvia CC, Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
2
|
Liu C, Liu WH, Yang W, Chen L, Xue Y, Chen XY. GLP-1 modulated the firing activity of nigral dopaminergic neurons in both normal and parkinsonian mice. Neuropharmacology 2024; 252:109946. [PMID: 38599494 DOI: 10.1016/j.neuropharm.2024.109946] [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: 01/26/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
The spontaneous firing activity of nigral dopaminergic neurons is associated with some important roles including modulation of dopamine release, expression of tyrosine hydroxylase (TH), as well as neuronal survival. The decreased neuroactivity of nigral dopaminergic neurons has been revealed in Parkinson's disease. Central glucagon-like peptide-1 (GLP-1) functions as a neurotransmitter or neuromodulator to exert multiple brain functions. Although morphological studies revealed the expression of GLP-1 receptors (GLP-1Rs) in the substantia nigra pars compacta, the possible modulation of GLP-1 on spontaneous firing activity of nigral dopaminergic neurons is unknown. The present extracellular in vivo single unit recordings revealed that GLP-1R agonist exendin-4 significantly increased the spontaneous firing rate and decreased the firing regularity of partial nigral dopaminergic neurons of adult male C57BL/6 mice. Blockade of GLP-1Rs by exendin (9-39) decreased the firing rate of nigral dopaminergic neurons suggesting the involvement of endogenous GLP-1 in the modulation of firing activity. Furthermore, the PKA and the transient receptor potential canonical (TRPC) 4/5 channels are involved in activation of GLP-1Rs-induced excitatory effects of nigral dopaminergic neurons. Under parkinsonian state, both the exogenous and endogenous GLP-1 could still induce excitatory effects on the surviving nigral dopaminergic neurons. As the mild excitatory stimuli exert neuroprotective effects on nigral dopaminergic neurons, the present GLP-1-induced excitatory effects may partially contribute to its antiparkinsonian effects.
Collapse
Affiliation(s)
- Cui Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China; Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wen-Hong Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wu Yang
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Xue
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xin-Yi Chen
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
3
|
Ruska Y, Peterfi Z, Szilvásy-Szabó A, Kővári D, Hrabovszky E, Dorogházi B, Gereben B, Tóth B, Matziari M, Wittmann G, Fekete C. GLP-1 Receptor Signaling Has Different Effects on the Perikarya and Axons of the Hypophysiotropic Thyrotropin-Releasing Hormone Synthesizing Neurons in Male Mice. Thyroid 2024; 34:252-260. [PMID: 38062754 DOI: 10.1089/thy.2023.0284] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Background: Glucagon-like peptide 1 (GLP-1) is involved in the regulation of energy and glucose homeostasis. As GLP-1 has similar effects on the energy homeostasis as the hypophysiotropic thyrotropin-releasing hormone (TRH) neurons that regulate the hypothalamic-pituitary-thyroid (HPT) axis, we raised the possibility that the TRH neurons are involved in the mediation of the effects of GLP-1. Therefore, the relationship and interaction of the GLP-1 system and the TRH neurons of the hypothalamic paraventricular nucleus (PVN) were studied. Methods: To examine the anatomical and functional relationship of TRH neurons and the GLP-1 system in the PVN, immunocytochemistry, in situ hybridization, in vitro patch-clamp electrophysiology, metabolic phenotyping, and explant experiments were performed. Results: Our data demonstrate that the TRH neurons of the PVN are innervated by GLP-1 producing neurons and express the GLP-1 receptor (GLP-1R). However, not only do the GLP-1-innervated TRH neurons express GLP-1R but the receptor is also present in the axons of the hypophysiotropic TRH neurons in the blood-brain barrier free median eminence (ME) suggesting that peripherally derived GLP-1 may also influence the TRH neurons. In vitro, GLP-1 increased the firing rate of TRH neurons and depolarized them. In addition, GLP-1 directly stimulated the GABAergic input of a population of TRH neurons. Furthermore, GLP-1 inhibited the release of TRH from the hypophysiotropic axons in the ME. In vivo, peripheral GLP-1R agonist administration markedly inhibited the food intake and the energy expenditure, but had no effect on the TRH expression in the PVN and resulted in lower circulating free T4 levels. Conclusions: Our results indicate that GLP-1R activation has a direct stimulatory effect on TRH neurons in the PVN, but the activation of GLP-1R may also inhibit TRH neurons by facilitating their inhibitory inputs or by inhibiting the axon terminals of these cells in the ME. The innervation of TRH neurons by GLP-1 neurons suggests that TRH neurons might be influenced by both circulating GLP-1 and by GLP-1 neurons of the nucleus tractus solitarii. The lack of GLP-1R agonist-induced regulation of TRH neurons in vivo suggests that the HPT axis does not mediate the GLP-1R agonist-induced weight loss.
Collapse
Affiliation(s)
- Yvette Ruska
- Laboratory of Integrative Neuroendocrinology; Budapest, Hungary
| | - Zoltan Peterfi
- Laboratory of Integrative Neuroendocrinology; Budapest, Hungary
| | | | - Dóra Kővári
- Laboratory of Integrative Neuroendocrinology; Budapest, Hungary
| | | | - Beáta Dorogházi
- Laboratory of Molecular Cell Metabolism; HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism; HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Magdalini Matziari
- Department of Chemistry, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Gábor Wittmann
- Laboratory of Integrative Neuroendocrinology; Budapest, Hungary
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology; Budapest, Hungary
| |
Collapse
|
4
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
5
|
Stathori G, Tzounakou AM, Mastorakos G, Vlahos NF, Charmandari E, Valsamakis G. Alterations in Appetite-Regulating Hormones in Girls with Central Early or Precocious Puberty. Nutrients 2023; 15:4306. [PMID: 37836591 PMCID: PMC10574110 DOI: 10.3390/nu15194306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
The prevalence of central precocious puberty (CPP) in girls has increased worldwide and is often associated with obesity in childhood as well as high fat/high glycemic index diets. Evidence suggests that subjects with obesity present with alterations in appetite-regulating hormones. The arcuate and paraventricular nuclei of the hypothalamus are the centers of action of appetite hormones, as well as the location of gonadotropin-releasing hormone (GnRH) neurons, the activation of which results in the onset of puberty. This anatomical proximity raises the question of possible alterations in appetite-regulating hormones in patients with CPP. Furthermore, diet-induced hypothalamic inflammation constitutes a probable mechanism of the pathophysiology of CPP, as well as alterations in appetite-regulating hormones in young children. In this article, we summarize the evidence investigating whether girls with CPP present with alterations in appetite-regulating hormones. We present evidence that leptin concentrations are elevated in girls with CPP, ghrelin concentrations are lower in girls with CPP, nesfatin-1 and orexin-A concentrations are elevated among girls with premature thelarche, and insulin concentrations are increased in girls with early menarche.
Collapse
Affiliation(s)
- Galateia Stathori
- Center for the Prevention and Management of Overweight and Obesity, Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (G.S.); (A.-M.T.)
| | - Anastasia-Maria Tzounakou
- Center for the Prevention and Management of Overweight and Obesity, Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (G.S.); (A.-M.T.)
| | - George Mastorakos
- Second Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, ‘Aretaieion’ University Hospital, 11528 Athens, Greece; (G.M.); (N.F.V.); (G.V.)
| | - Nikolaos F. Vlahos
- Second Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, ‘Aretaieion’ University Hospital, 11528 Athens, Greece; (G.M.); (N.F.V.); (G.V.)
| | - Evangelia Charmandari
- Center for the Prevention and Management of Overweight and Obesity, Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (G.S.); (A.-M.T.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Georgios Valsamakis
- Second Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, ‘Aretaieion’ University Hospital, 11528 Athens, Greece; (G.M.); (N.F.V.); (G.V.)
| |
Collapse
|
6
|
Simpson EM, Clarke IJ, Scott CJ, Stephen CP, Rao A, Gunn AJ. The GLP-1 agonist, exendin-4, stimulates LH secretion in female sheep. J Endocrinol 2023; 259:e230105. [PMID: 37466202 PMCID: PMC10448581 DOI: 10.1530/joe-23-0105] [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: 03/29/2023] [Accepted: 07/18/2023] [Indexed: 07/20/2023]
Abstract
Our previous studies showed that microinjection into the median eminence of the sheep of glucagon-like peptide- 1 (GLP-1) or its receptor agonist exendin-4 stimulates luteinising hormone (LH) secretion, but it is unknown whether the same effect may be obtained by systemic administration of the same. The present study measured the response in terms of plasma LH concentrations to intravenous (iv) infusion of exendin-4. A preliminary study showed that infusion of 2 mg exendin-4 into ewes produced a greater LH response in the follicular phase of the oestrous cycle than the luteal phase. Accordingly, the main study monitored plasma LH levels in response to either 0.5 mg or 2 mg exendin-4 or vehicle (normal saline) delivered by jugular infusion for 1 h in the follicular phase of the oestrous cycle. Blood samples were collected at 10 min intervals before, during and after infusion. Both doses of exendin-4 increased mean plasma LH concentrations and increased LH peripheral pulse amplitude. There was no effect on inter-pulse interval or timing of the preovulatory LH surge. These doses of exendin-4 did not alter plasma insulin or glucose concentrations. Quantitative PCR of the gastrointestinal tract samples from a population of ewes confirmed the expression of the preproglucagon gene (GCG). Expression increased aborally and was greatest in the rectum. It is concluded that endogenous GLP-1, most likely derived from the hindgut, may act systemically to stimulate LH secretion. The present data suggest that this effect may be obtained with levels of agonist that are lower than those functioning as an incretin.
Collapse
Affiliation(s)
- Elizabeth M Simpson
- School of Agricultural, Environmental and Veterinary Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
- Gulbali Institute, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Iain J Clarke
- School of Agriculture Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Christopher J Scott
- School of Dentistry and Medical Science, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Cyril P Stephen
- School of Agricultural, Environmental and Veterinary Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
- Gulbali Institute, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Alexandra Rao
- School of Agriculture Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Allan J Gunn
- School of Agricultural, Environmental and Veterinary Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
- Gulbali Institute, Charles Sturt University, Wagga Wagga, NSW, Australia
| |
Collapse
|
7
|
Ruiz-Cruz M, Torres-Granados C, Tena-Sempere M, Roa J. Central and peripheral mechanisms involved in the control of GnRH neuronal function by metabolic factors. Curr Opin Pharmacol 2023; 71:102382. [PMID: 37307655 DOI: 10.1016/j.coph.2023.102382] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are the final output pathway for the brain control of reproduction. The activity of this neuronal population, mainly located at the preoptic area of the hypothalamus, is controlled by a plethora of metabolic signals. However, it has been documented that most of these signal impact on GnRH neurons through indirect neuronal circuits, Kiss1, proopiomelanocortin, and neuropeptide Y/agouti-related peptide neurons being some of the most prominent mediators. In this context, compelling evidence has been gathered in recent years on the role of a large range of neuropeptides and energy sensors in the regulation of GnRH neuronal activity through both direct and indirect mechanisms. The present review summarizes some of the most prominent recent advances in our understanding of the peripheral factors and central mechanisms involved in the metabolic control of GnRH neurons.
Collapse
Affiliation(s)
- Miguel Ruiz-Cruz
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain
| | - Carmen Torres-Granados
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Juan Roa
- Instituto Maimónides de Investigación Biomédica de Córdoba, Department of Cell Biology, Physiology and Immunology, University of Córdoba; Hospital Universitario Reina Sofia (IMIBIC/HURS), 14004 Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Córdoba, Spain.
| |
Collapse
|
8
|
Yazıcı D, Yapıcı Eser H, Kıyıcı S, Sancak S, Sezer H, Uygur M, Yumuk V. Clinical Impact of Glucagon-Like Peptide-1 Receptor Analogs on the Complications of Obesity. Obes Facts 2023; 16:149-163. [PMID: 36349778 PMCID: PMC10028372 DOI: 10.1159/000526808] [Citation(s) in RCA: 6] [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: 05/04/2022] [Accepted: 08/04/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Obesity is a chronic disease associated with increased morbidity and mortality due to its complications. The aims of obesity treatment are primarily to accomplish weight loss, and prevention or treatment of its complications. Lifestyle changes along with behavioral therapy constitute the first-line treatment of obesity followed by pharmacotherapy. Glucagon-like peptide receptor analogs (GLP-1 RAs) are among the approved pharmacotherapy options. Their central effect on suppressing appetite results in considerable weight loss. However, their effect on the complications of obesity has not been very well recognized. This review aims to analyze the effects of GLP-1 RAs on the complications of obesity, as diabetes mellitus, hypertension, nonalcoholic steatohepatitis (NASH), cardiovascular diseases, polycystic ovary syndrome, infertility, obstructive sleep apnea (OSA), osteoarthritis, cancer and central nervous system problems. SUMMARY Data from preclinical studies and clinical trials have been thoroughly evaluated. Effects regarding the complications as far as the scope of this review have covered can be summarized as blood glucose lowering, blood pressure lowering, resolution of NASH, improving major cardiovascular events, improving fertility and sex hormone levels, and improvement in OSA symptoms and in cognitive scores. Although the mechanisms are not fully elucidated, it is clear that the effects are not solely due to weight loss, but some pleiotropic effects like decreased inflammation, oxidative stress, and fibrosis also play a role in some of the complications. KEY MESSAGES Treating obesity is not only enabling weight loss but ameliorating complications related to obesity. Thus, any antiobesity medication has to have some favorable effects on the complications. As far as the GLP-RA's analogs are concerned, there seems to be an improvement in many of the complications regardless of the weight loss effect of these medications.
Collapse
Affiliation(s)
- Dilek Yazıcı
- Koç University Medical School Section of Endocrinology and Metabolism, Istanbul, Turkey
- *Dilek Yazıcı,
| | - Hale Yapıcı Eser
- Koç University Medical School Department of Psychiatry, Istanbul, Turkey
| | - Sinem Kıyıcı
- Health Sciences University Bursa Yİ Education and Research Hospital, Bursa, Turkey
| | - Seda Sancak
- Health Sciences University Fatih Sultan Mehmet Education and Research Hospital, Istanbul, Turkey
| | - Havva Sezer
- Koç University Medical School Section of Endocrinology and Metabolism, Istanbul, Turkey
| | - Melin Uygur
- Marmara University Medical School, Section of Endocrinology and Metabolism, Istanbul, Turkey
| | - Volkan Yumuk
- Cerrahpaşa U. Medical School Section of Endocrinology and Metabolism, Istanbul, Turkey
| |
Collapse
|
9
|
Liu J, Liu Z, Sun W, Luo L, An X, Yu D, Wang W. Role of sex hormones in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1135530. [PMID: 37143724 PMCID: PMC10151816 DOI: 10.3389/fendo.2023.1135530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
Diabetic nephropathy (DN) is the most common microvascular complication in diabetes and one of the leading causes of end-stage renal disease. The standard treatments for patients with classic DN focus on blood glucose and blood pressure control, but these treatments can only slow the progression of DN instead of stopping or reversing the disease. In recent years, new drugs targeting the pathological mechanisms of DN (e.g., blocking oxidative stress or inflammation) have emerged, and new therapeutic strategies targeting pathological mechanisms are gaining increasing attention. A growing number of epidemiological and clinical studies suggest that sex hormones play an important role in the onset and progression of DN. Testosterone is the main sex hormone in males and is thought to accelerate the occurrence and progression of DN. Estrogen is the main sex hormone in females and is thought to have renoprotective effects. However, the underlying molecular mechanism by which sex hormones regulate DN has not been fully elucidated and summarized. This review aims to summarize the correlation between sex hormones and DN and evaluate the value of hormonotherapy in DN.
Collapse
Affiliation(s)
- Jiahui Liu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhe Liu
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Weixia Sun
- Nephrology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Ling Luo
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingna An
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Public Research Platform, First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Dehai Yu, ; Wanning Wang,
| | - Wanning Wang
- Nephrology Department, First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Dehai Yu, ; Wanning Wang,
| |
Collapse
|
10
|
Khan D, Ojo OO, Woodward ORM, Lewis JE, Sridhar A, Gribble FM, Reimann F, Flatt PR, Moffett RC. Evidence for Involvement of GIP and GLP-1 Receptors and the Gut-Gonadal Axis in Regulating Female Reproductive Function in Mice. Biomolecules 2022; 12:1736. [PMID: 36551163 PMCID: PMC9775379 DOI: 10.3390/biom12121736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/11/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Substantial evidence suggests crosstalk between reproductive and gut-axis but mechanisms linking metabolism and reproduction are still unclear. The present study evaluated the possible role of glucose-dependent-insulinotropic-polypeptide (GIP) and glucagon-like-peptide-1 (GLP-1) in reproductive function by examining receptor distribution and the effects of global GIPR and GLP-1R deletion on estrous cycling and reproductive outcomes in mice. GIPR and GLP-1R gene expression were readily detected by PCR in female reproductive tissues including pituitary, ovaries and uterine horn. Protein expression was confirmed with histological visualisation of incretin receptors using GIPR-Cre and GLP1R-Cre mice in which the incretin receptor expressing cells were fluorescently tagged. Functional studies revealed that female GIPR-/- and GLP-1R-/- null mice exhibited significantly (p < 0.05 and p < 0.01) deranged estrous cycling compared to wild-type controls, indicative of reduced fertility. Furthermore, only 50% and 16% of female GIPR-/- and GLP-1R-/- mice, respectively produced litters with wild-type males across three breeding cycles. Consistent with a physiological role of incretin receptors in pregnancy outcome, litter size was significantly (p < 0.001-p < 0.05) decreased in GIPR-/- and GLP-1R-/- mice. Treatment with oral metformin (300 mg/kg body-weight), an agent used clinically for treatment of PCOS, for a further two breeding periods showed no amelioration of pregnancy outcome except that litter size in the GIPR-/- group was approximately 2 times greater in the second breeding cycle. These data highlight the significance of incretin receptors in modulation of female reproductive function which may provide future targets for pharmacological intervention in reproductive disorders.
Collapse
Affiliation(s)
- Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
| | - Opeolu O. Ojo
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
- Department of Biology, Chemistry & Forensic Science, School of Sciences, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Orla RM Woodward
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Jo Edward Lewis
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Ananyaa Sridhar
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
| | - Fiona M. Gribble
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Frank Reimann
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Peter R. Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
| | - R. Charlotte Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK
| |
Collapse
|
11
|
Shao S, Zhang X, Xu Q, Pan R, Chen Y. Emerging roles of Glucagon like peptide-1 in the management of autoimmune diseases and diabetes-associated comorbidities. Pharmacol Ther 2022; 239:108270. [PMID: 36002078 DOI: 10.1016/j.pharmthera.2022.108270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/26/2022]
|
12
|
Jensterle M, Herman R, Janež A. Therapeutic Potential of Glucagon-like Peptide-1 Agonists in Polycystic Ovary Syndrome: From Current Clinical Evidence to Future Perspectives. Biomedicines 2022; 10:1989. [PMID: 36009535 PMCID: PMC9405922 DOI: 10.3390/biomedicines10081989] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/30/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the continuous effort to understand the pathophysiology and determine potential therapeutic targets, PCOS treatment largely depends on lifestyle intervention and symptomatic management of individual signs and symptoms. International guidelines recognize the importance of weight reduction as a cornerstone for the achievement of better metabolic, reproductive, and cardiovascular outcomes in PCOS women who are overweight or obese. With its profound weight loss potential in patients with or without diabetes, the administration of GLP-1 receptor agonists has been investigated in overweight/obese women with PCOS in several single-center randomized control trials with considerable variation in the dosing regimen, follow-up duration, and outcome measurements over recent years. Most trials reported superior weight loss effects of GLP-1 receptor agonists compared to lifestyle changes or metformin, with additional metabolic, reproductive, and cardiovascular benefits in this population. However, their use is currently not widely accepted by the clinical community that treats this population. The major concern is how to balance the reproductive and metabolic treatment strategies since the use of GLP-1 receptor agonists requires effective contraception while on therapy and a washout period before pregnancy. Both approaches are not mutually exclusive, yet the best choice requires a careful assessment of the clinical context. Knowing a patient's individual circumstances, precise clinical sub-phenotyping, and regular monitoring are crucial components for the safe and effective use of these new tools. In the present narrative review, we explore the current clinical evidence and provide the future perspectives and challenges for their implementation in PCOS management.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Rok Herman
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
13
|
Jamieson BB, Piet R. Kisspeptin neuron electrophysiology: Intrinsic properties, hormonal modulation, and regulation of homeostatic circuits. Front Neuroendocrinol 2022; 66:101006. [PMID: 35640722 DOI: 10.1016/j.yfrne.2022.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/04/2022]
Abstract
The obligatory role of kisspeptin (KISS1) and its receptor (KISS1R) in regulating the hypothalamic-pituitary-gonadal axis, puberty and fertility was uncovered in 2003. In the few years that followed, an impressive body of work undertaken in many species established that neurons producing kisspeptin orchestrate gonadotropin-releasing hormone (GnRH) neuron activity and subsequent GnRH and gonadotropin hormone secretory patterns, through kisspeptin-KISS1R signaling, and mediate many aspects of gonadal steroid hormone feedback regulation of GnRH neurons. Here, we review knowledge accrued over the past decade, mainly in genetically modified mouse models, of the electrophysiological properties of kisspeptin neurons and their regulation by hormonal feedback. We also discuss recent progress in our understanding of the role of these cells within neuronal circuits that control GnRH neuron activity and GnRH secretion, energy balance and, potentially, other homeostatic and reproductive functions.
Collapse
Affiliation(s)
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
14
|
Abdullah DM, Alsemeh AE, Khamis T. Semaglutide early intervention attenuated testicular dysfunction by targeting the GLP-1-PPAR-α-Kisspeptin-Steroidogenesis signaling pathway in a testicular ischemia-reperfusion rat model. Peptides 2022; 149:170711. [PMID: 34920048 DOI: 10.1016/j.peptides.2021.170711] [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: 08/18/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022]
Abstract
Testicular torsion is a serious emergency and a well-known cause of male infertility. It represents 10 %-15 % of scrotal diseases in children. Kisspeptin (KISS1) is a hormone secreted from the hypothalamic nuclei and testis, but its role in testis is not fully understood. Semaglutide is a novel antidiabetic glucagon-like peptide 1 (GLP-1) analog. Hence, we designed the current study to elucidate the possible ameliorative effect of semaglutide on ischemia/reperfusion-induced testicular dysfunction in rats and highlight the role of the testicular GLP-1/PCG-1α-PPAR-α-KISS1 signaling pathway. We randomly divided 50 male Sprague Dawley into five equal groups (10 rats each): SHAM, exendin 9-39 -treated (EX), testicular torsion/detorsion (T/D), testicular torsion/detorsion and semaglutide-treated (SEM + T/D), and testicular torsion/detorsion, exendin, and semaglutide-treated (EX + SEM + T/D). We quantified serum follicle-stimulating hormone, luteinizing hormone, total testosterone, testicular oxidative stress markers, testicular gene expression of GLP-1/KISS1 pathway-related genes (KISS1, KISS1R, GLP-1, GLP-1R, PGC-1α, PPAR-α), steroidogenesis pathway-related genes (STAR, CYP11A1, CYP17A1, HSD17B3, CYP19A1), HO-1, Nrf-2, and testicular protein expression of HIF-1α, TNF-α, NF-κβ, Caspase-3, FAS, proliferating cell nuclear antigen, and KISS1 through testicular histopathology and immunohistochemistry assays. Testicular torsion/detorsion markedly elevated proapoptotic, proinflammatory, and oxidative stress marker levels, noticeably downregulating the expression of GLP-1/KISS1 and steroidogenesis pathway-related proteins. Semaglutide administration significantly ameliorated all these deleterious effects. Nevertheless, injecting exendin, a GLP1-R antagonist, before semaglutide abolished all the documented improvements. We concluded that semaglutide ameliorated ischemia/reperfusion-induced testicular dysfunction by modulating the GLP-1/PGC-1α-PPAR-α/KISS1/steroidogenesis signaling pathway, improving testicular oxidative state, and suppressing testicular inflammation and apoptosis.
Collapse
Affiliation(s)
- Doaa M Abdullah
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, 44519 Zagazig, Egypt
| | - Amira Ebrahim Alsemeh
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, 44519 Zagazig, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, 44519 Zagazig, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, 44519 Zagazig, Egypt.
| |
Collapse
|
15
|
Xing C, Zhang J, Zhao H, He B. Effect of Sex Hormone-Binding Globulin on Polycystic Ovary Syndrome: Mechanisms, Manifestations, Genetics, and Treatment. Int J Womens Health 2022; 14:91-105. [PMID: 35140526 PMCID: PMC8818772 DOI: 10.2147/ijwh.s344542] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/19/2022] [Indexed: 12/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine diseases causing infertility in women of childbearing age. It is characterized by hyperandrogenemia (HA), chronic anovulation, and polycystic ovary morphology (PCOM). Most women with PCOS have metabolic abnormalities. Sex hormone-binding globulin (SHBG), a transport carrier that binds estrogen and androgens and regulates their biological activity, is usually used as an indicator of hyperandrogenism in women with PCOS. Low serum SHBG levels are considered a biomarker of metabolic abnormalities and are associated with insulin resistance (IR), HA, and abnormal glucose and lipid metabolism in PCOS patients. SHBG is also related to the long-term prognosis of PCOS, whereas SHBG gene polymorphism is associated with PCOS risk. In addition, the administration of metformin (MET), glucagon-like peptide-1 receptor agonists (GLP-1 RAs), thiazolidinediones (TZDs), compound oral contraceptives (COCs), as well as nutrient supplements such as inositol (MI), vitamin D, and synbiotics can regulate SHBG levels to ameliorate PCOS complications and improve prognosis. This review focuses on the interaction between SHBG and various PCOS complications as well as the regulation of SHBG by various drugs and nutrients and its therapeutic effects on PCOS.
Collapse
Affiliation(s)
- Chuan Xing
- The First Department of Endocrine, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Jiaqi Zhang
- The First Department of Endocrine, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Han Zhao
- The First Department of Endocrine, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Bing He
- The First Department of Endocrine, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| |
Collapse
|
16
|
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
|
17
|
Chen XY, Chen L, Yang W, Xie AM. GLP-1 Suppresses Feeding Behaviors and Modulates Neuronal Electrophysiological Properties in Multiple Brain Regions. Front Mol Neurosci 2022; 14:793004. [PMID: 34975402 PMCID: PMC8718614 DOI: 10.3389/fnmol.2021.793004] [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] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022] Open
Abstract
The glucagon-like peptide-1 (GLP-1) plays important roles in the regulation of food intake and energy metabolism. Peripheral or central GLP-1 suppresses food intake and reduces body weight. The electrophysiological properties of neurons in the mammalian central nervous system reflect the neuronal excitability and the functional organization of the brain. Recent studies focus on elucidating GLP-1-induced suppression of feeding behaviors and modulation of neuronal electrophysiological properties in several brain regions. Here, we summarize that activation of GLP-1 receptor (GLP-1R) suppresses food intake and induces postsynaptic depolarization of membrane potential and/or presynaptic modulation of glutamatergic or GABAergic neurotransmission in brain nuclei located within the medulla oblongata, pons, mesencephalon, diencephalon, and telencephalon. This review may provide a background to guide future research about the cellular mechanisms of GLP-1-induced feeding inhibition.
Collapse
Affiliation(s)
- Xin-Yi Chen
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wu Yang
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, China
| | - An-Mu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
18
|
Constantin S, Pizano K, Matson K, Shan Y, Reynolds D, Wray S. An Inhibitory Circuit From Brainstem to GnRH Neurons in Male Mice: A New Role for the RFRP Receptor. Endocrinology 2021; 162:6132086. [PMID: 33564881 PMCID: PMC8016070 DOI: 10.1210/endocr/bqab030] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 12/31/2022]
Abstract
RFamide-related peptides (RFRPs, mammalian orthologs of gonadotropin-inhibitory hormone) convey circadian, seasonal, and social cues to the reproductive system. They regulate gonadotropin secretion by modulating gonadotropin-releasing hormone (GnRH) neurons via the RFRP receptor. Mice lacking this receptor are fertile but exhibit abnormal gonadotropin responses during metabolic challenges, such as acute fasting, when the normal drop in gonadotropin levels is delayed. Although it is known that these food intake signals to the reproductive circuit originate in the nucleus tractus solitarius (NTS) in the brainstem, the phenotype of the neurons conveying the signal remains unknown. Given that neuropeptide FF (NPFF), another RFamide peptide, resides in the NTS and can bind to the RFRP receptor, we hypothesized that NPFF may regulate GnRH neurons. To address this question, we used a combination of techniques: cell-attached electrophysiology on GnRH-driven green fluorescent protein-tagged neurons in acute brain slices; calcium imaging on cultured GnRH neurons; and immunostaining on adult brain tissue. We found (1) NPFF inhibits GnRH neuron excitability via the RFRP receptor and its canonical signaling pathway (Gi/o protein and G protein-coupled inwardly rectifying potassium channels), (2) NPFF-like fibers in the vicinity of GnRH neurons coexpress neuropeptide Y, (3) the majority of NPFF-like cell bodies in the NTS also coexpress neuropeptide Y, and (4) acute fasting increased NPFF-like immunoreactivity in the NTS. Together these data indicate that NPFF neurons within the NTS inhibit GnRH neurons, and thus reproduction, during fasting but prior to the energy deficit.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Katherine Pizano
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Kaya Matson
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Yufei Shan
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Daniel Reynolds
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
- Correspondence: Dr. Susan Wray, Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive MSC 3703, Building 35, Room 3A1012, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Abdalla MA, Deshmukh H, Atkin S, Sathyapalan T. The potential role of incretin-based therapies for polycystic ovary syndrome: a narrative review of the current evidence. Ther Adv Endocrinol Metab 2021; 12:2042018821989238. [PMID: 33552465 PMCID: PMC7844452 DOI: 10.1177/2042018821989238] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/03/2021] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age. Metabolic consequences associated with PCOS include, but are not limited to, insulin resistance (IR), type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). This narrative review aims to provide a comprehensive overview of the potential therapeutic roles of the incretin-based therapies in the management of PCOS. METHODS We performed a systematic search of databases including PubMed, MEDLINE and EMBASE up to 1 October 2020. We developed a search string of medical subject headings (MeSH) including the terms PCOS, incretin mimetics, glucagon-like peptide-1 (GLP-1), glucagon-like peptide-1 receptor antagonists (GLP-1 RAs), liraglutide, exenatide, semaglutide, dipeptidyl peptidase-4 (DPP-4) inhibitors, combined with IR, testosterone and sex hormone-binding globulin (SHBG). RESULTS We identified 854 relevant articles and, after the initial screening, eight interventional animal studies, one observational animal study, 14 interventional human studies, two case-control studies and one systematic review were included. These studies showed the potential significant roles of GLP-1 RAs and DPP-4 inhibitors in the management of PCOS, with significant improvements in the metabolic parameters, including substantial weight reduction and improved insulin sensitivity. These agents also improved the hormonal parameters through decreased free androgen and increased SHBG. Moreover, they improved menstrual regularity, increased fertility with enhanced ovulation and pregnancy in obese women with PCOS. CONCLUSION GLP-1 RAs and DPP-4 inhibitors have a promising therapeutic role in PCOS; however, larger clinical trials are needed to establish the role of incretin-based therapies in the management of PCOS.
Collapse
Affiliation(s)
- Mohammed Altigani Abdalla
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Harshal Deshmukh
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Stephen Atkin
- School of Postgraduate Studies and Research, RCSI Medical University of Bahrain, Kingdom of Bahrain
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| |
Collapse
|
20
|
Liao B, Qiao J, Pang Y. Central Regulation of PCOS: Abnormal Neuronal-Reproductive-Metabolic Circuits in PCOS Pathophysiology. Front Endocrinol (Lausanne) 2021; 12:667422. [PMID: 34122341 PMCID: PMC8194358 DOI: 10.3389/fendo.2021.667422] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease. PCOS patients are characterized by hyperandrogenemia, anovulation, and metabolic dysfunction. Hypothalamus-pituitary-ovary axis imbalance is considered as an important pathophysiology underlying PCOS, indicating that central modulation, especially the abnormal activation of hypothalamic GnRH neurons plays a vital role in PCOS development. Increased GnRH pulse frequency can promote LH secretion, leading to ovarian dysfunction and abnormal sex steroids synthesis. By contrast, peripheral sex steroids can modulate the action of GnRH neurons through a feedback effect, which is impaired in PCOS, thus forming a vicious cycle. Additionally, hypothalamic GnRH neurons not only serve as the final output pathway of central control of reproductive axis, but also as the central connection point where reproductive function and metabolic state inter-regulate with each other. Metabolic factors, such as insulin resistance and obesity in PCOS patients can regulate GnRH neurons activity, and ultimately regulate reproductive function. Besides, gut hormones act on both brain and peripheral organs to modify metabolic state. Gut microbiota disturbance is also related to many metabolic diseases and has been reported to play an essential part in PCOS development. This review concludes with the mechanism of central modulation and the interaction between neuroendocrine factors and reproductive or metabolic disorders in PCOS development. Furthermore, the role of the gut microenvironment as an important part involved in the abnormal neuronal-reproductive-metabolic circuits that contribute to PCOS is discussed, thus offering possible central and peripheral therapeutic targets for PCOS patients.
Collapse
Affiliation(s)
- Baoying Liao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Yanli Pang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
- *Correspondence: Yanli Pang,
| |
Collapse
|
21
|
Vastagh C, Farkas I, Scott MM, Liposits Z. Networking of glucagon-like peptide-1 axons with GnRH neurons in the basal forebrain of male mice revealed by 3DISCO-based immunocytochemistry and optogenetics. Brain Struct Funct 2020; 226:105-120. [PMID: 33169188 PMCID: PMC7817561 DOI: 10.1007/s00429-020-02167-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/17/2020] [Indexed: 12/25/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) regulates reproduction centrally, although, the neuroanatomical basis of the process is unknown. Therefore, the putative networking of the central GLP-1 and gonadotropin-releasing hormone (GnRH) systems was addressed in male mice using whole mount immunocytochemistry and optogenetics. Enhanced antibody penetration and optical clearing procedures applied to 500–1000 µm thick basal forebrain slices allowed the simultaneous visualization of the two distinct systems in the basal forebrain. Beaded GLP-1-IR axons innervated about a quarter of GnRH neurons (23.2 ± 1.4%) forming either single or multiple contacts. GnRH dendrites received a more intense GLP-1 innervation (64.6 ± 0.03%) than perikarya (35.4 ± 0.03%). The physiological significance of the innervation was examined by optogenetic activation of channelrhodopsin-2 (ChR2)-expressing axons of preproglucagon (GCG) neurons upon the firing of GnRH neurons by patch clamp electrophysiology in acute brain slices of triple transgenic mice (Gcg-cre/ChR2/GFP-GnRH). High-frequency laser beam stimulation (20 Hz, 10 ms pulse width, 3 mW laser power) of ChR2-expressing GCG axons in the mPOA increased the firing rate of GnRH neurons (by 75 ± 17.3%, p = 0.0007). Application of the GLP-1 receptor antagonist, Exendin-3-(9-39) (1 μM), prior to the photo-stimulation, abolished the facilitatory effect. In contrast, low-frequency trains of laser pulses (0.2 Hz, 60 pulses) had no effect on the spontaneous postsynaptic currents of GnRH neurons. The findings indicate a direct wiring of GLP-1 neurons with GnRH cells which route is excitatory for the GnRH system. The pathway may relay metabolic signals to GnRH neurons and synchronize metabolism with reproduction.
Collapse
Affiliation(s)
- Csaba Vastagh
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, 1083, Budapest, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Michael M Scott
- Department of Pharmacology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, 1083, Budapest, Hungary.
- Department of Neuroscience, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
| |
Collapse
|
22
|
Abstract
Polycystic ovary syndrome is a complex and heterogenous disorder involving multiple organ systems and different molecular pathways. It is tightly associated with obesity and especially abdominal obesity. As body weight reduction is the main modifiable risk factor for polycystic ovary syndrome, therapeutic approaches in overweight or obese women with polycystic ovary syndrome have been developed. Liraglutide is a glucagon-like peptide-1 receptor agonist that promotes sustained weight loss, as well as abdominal fat reduction, in individuals with obesity, prediabetes, and type 2 diabetes mellitus. The majority of current clinical studies have demonstrated that liraglutide therapy achieved significant reductions in body weight, body mass index, and abdominal circumference in overweight and obese women with polycystic ovary syndrome. Liraglutide therapy promoted significant improvements in free testosterone and sex hormone-binding globulin levels in some studies. Important metabolic and hormonal improvements were also reported after the combination of liraglutide with metformin. Increased menstrual frequency, as well as potential positive effects in reproduction, were described. However, the small number of participants, short duration, and low daily liraglutide dose are some of the main limitations of these studies. Larger and longer, multi-centred, double-blind, placebo-controlled trials of liraglutide monotherapy or combination therapy, with prolonged post-interventional monitoring, are crucially anticipated. Metabolic, hormonal, and reproductive primary outcomes should be uniformly addressed, to tailor future targeted treatment approaches, according to the patient phenotype and needs. This will improve long-term therapeutic outcomes in this population.
Collapse
|
23
|
Kisspeptin-52 partially rescues the activity of the hypothalamus-pituitary-gonadal axis in underweight male rats dosed with an anti-obesity compound. Toxicol Appl Pharmacol 2020; 404:115152. [PMID: 32726590 DOI: 10.1016/j.taap.2020.115152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/09/2020] [Accepted: 07/16/2020] [Indexed: 11/21/2022]
Abstract
Energy metabolism and reproduction are closely linked and reciprocally regulated. The detrimental effect of underweight on reproduction complicates the safety evaluation of anti-obesity drugs, making it challenging to distinguish pathological changes mediated through the intended drug-induced weight loss from direct drug effects on reproductive organs. Four-weeks dosing of normal weight Sprague Dawley rats with a glucagon-like peptide 1 (GLP-1)/glucagon receptor co-agonist induced a robust weight loss, accompanied by histological findings in prostate, seminal vesicles, mammary glands, uterus/cervix and vagina. Characterization of the hypothalamus-pituitary-gonadal (HPG) axis in male rats revealed reduced hypothalamic Kiss1 mRNA levels and decreased serum luteinizing hormone (LH) and testosterone concentrations following co-agonist dosing. These alterations resemble hypogonadotropic hypogonadism typically seen in adverse energy deprived conditions, like chronic food restriction. Concomitant daily administration of kisspeptin-52 from day 21 to the end of the four-week co-agonist dosing period evoked LH and testosterone responses without normalizing histological findings. This incomplete rescue by kisspeptin-52 may be due to the rather short kisspeptin-52 treatment period combined with a desensitization observed on testosterone responses. Concomitant leptin treatment from day 21 did not reverse co-agonist induced changes in HPG axis activity. Furthermore, a single co-agonist injection in male rats slightly elevated LH levels but left testosterone unperturbed, thereby excluding a direct acute inhibitory effect on the HPG axis. Our data suggest that the reproductive phenotype after repeated co-agonist administration was driven by the intended weight loss, however, we cannot exclude a direct organ related effect in chronically treated rats.
Collapse
|
24
|
Jensterle M, Janez A, Fliers E, DeVries JH, Vrtacnik-Bokal E, Siegelaar SE. The role of glucagon-like peptide-1 in reproduction: from physiology to therapeutic perspective. Hum Reprod Update 2020; 25:504-517. [PMID: 31260047 DOI: 10.1093/humupd/dmz019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/09/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1 RAs) have become firmly established in the treatment of type 2 diabetes and obesity, disorders frequently associated with diminished reproductive health. Understanding of the role of GLP-1 and GLP-1 RAs in reproduction is currently limited and largely unaddressed in clinical studies. OBJECTIVE AND RATIONALE The purpose of this narrative review is to provide a comprehensive overview of the role of GLP-1 in reproduction and to address a therapeutic perspective that can be derived from these findings. SEARCH METHODS We performed a series of PubMed database systemic searches, last updated on 1 February 2019, supplemented by the authors' knowledge and research experience in the field. A search algorithm was developed incorporating the terms glucagon-like peptide-1, GLP-1, glucagon-like peptide-1 receptor, GLP-1R, or incretins, and this was combined with terms related to reproductive health. The PICO (Population, Intervention, Comparison, Outcome) framework was used to identify interventional studies including GLP-1 RAs and dipeptidyl peptidase-4 (DPP-4) inhibitors, which prevent the degradation of endogenously released GLP-1. We identified 983 potentially relevant references. At the end of the screening process, we included 6 observational (3 preclinical and 3 human) studies, 24 interventional (9 preclinical and 15 human) studies, 4 case reports, and 1 systematic and 2 narrative reviews. OUTCOMES The anatomical distribution of GLP-1 receptor throughout the reproductive system and observed effects of GLP-1 in preclinical models and in a few clinical studies indicate that GLP-1 might be one of the important modulating signals connecting the reproductive and metabolic system. The outcomes show that there is mostly stimulating role of GLP-1 and its mimetics in mammalian reproduction that goes beyond mere weight reduction. In addition, GLP-1 seems to have anti-inflammatory and anti-fibrotic effects in the gonads and the endometrium affected by obesity, diabetes, and polycystic ovary syndrome (PCOS). It also seems that GLP-1 RAs and DPP-4 inhibitors can reverse polycystic ovary morphology in preclinical models and decrease serum concentrations of androgens and their bioavailability in women with PCOS. Preliminary data from interventional clinical studies suggest improved menstrual regularity as well as increased fertility rates in overweight and/or obese women with PCOS treated with GLP-1 RAs in the preconception period. WIDER IMPLICATIONS GLP-1 RAs and DPP-4 inhibitors show promise in the treatment of diabetes and obesity-related subfertility. Larger interventional studies are needed to establish the role of preconception intervention with GLP-1 based therapies, assessing fertility outcomes in obesity, PCOS, and diabetes-related fertility problems. The potential impact of the dose- and exposure time-response of different GLP-1 RAs need further exploration. Future research should also investigate sex-specific variability of GLP-1 on reproductive outcomes, in particular on the gonads where the observations in males are most conflicting.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, Ljubljana, Slovenia
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, Ljubljana, Slovenia
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - J Hans DeVries
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Eda Vrtacnik-Bokal
- Department of Human Reproduction, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Slajmerjeva ulica 03, Ljubljana, Slovenia
| | - Sarah E Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
25
|
Izzi-Engbeaya C, Jones S, Crustna Y, Machenahalli PC, Papadopoulou D, Modi M, Panayi C, Starikova J, Eng PC, Phylactou M, Mills E, Yang L, Ratnasabapathy R, Sykes M, Plumptre I, Coumbe B, Wing VC, Pacuszka E, Bech P, Minnion J, Tharakan G, Tan T, Veldhuis J, Abbara A, Comninos AN, Dhillo WS. Effects of Glucagon-like Peptide-1 on the Reproductive Axis in Healthy Men. J Clin Endocrinol Metab 2020; 105:5735220. [PMID: 32052032 PMCID: PMC7082082 DOI: 10.1210/clinem/dgaa072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/10/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Glucagon-like peptide-1 (GLP-1) potently reduces food intake and augments glucose-stimulated insulin secretion. Recent animal data suggest that GLP-1 may also influence reproduction. As GLP-1 receptor agonists are currently widely used in clinical practice to treat obesity/type 2 diabetes, it is necessary to determine the effects of GLP-1 on the reproductive system in humans. OBJECTIVE To investigate the effects of GLP-1 administration on the reproductive axis in humans. DESIGN Single-blind, randomized, placebo-controlled crossover study. SETTING Clinical Research Facility, Imperial College Healthcare NHS Trust. PARTICIPANTS Eighteen healthy men (mean age 24.7 ± 0.1years, mean BMI 22.1 ± 0.4kg/m2). INTERVENTION Eight-hour intravenous infusion of 0.8 pmol/kg/min GLP-1 or rate-matched vehicle infusion. MAIN OUTCOME MEASURES Number of luteinizing hormone (LH) pulses, LH, follicle-stimulating hormone (FSH), and testosterone levels. RESULTS The number of LH pulses (number of LH pulses/500 min: vehicle 4.2 ± 0.4, GLP-1 4.5 ± 0.3, P = 0.46), LH area under the curve (AUC) (vehicle 1518 ± 88min.IU/L, GLP-1 1524 ± 101min.IU/L, P = 0.95), follicle-stimulating hormone AUC (vehicle 1210 ± 112 min IU/L, GLP-1 1216 ± 112 min IU/L, P = 0.86), and testosterone AUC (vehicle 10893 ± 615 min nmol/L, GLP-1 11088 ± 792 min nmol/L, P = 0.77) did not significantly differ during vehicle and GLP-1 administration. Glucagon-like peptide-1 significantly reduced food intake (vehicle 15.7 ± 1.3 kcal/kg, GLP-1 13.4 ± 1.3 kcal/kg, P = 0.01). CONCLUSIONS In contrast to the animal literature, our data demonstrate that acute GLP-1 administration does not affect reproductive hormone secretion in healthy men.
Collapse
Affiliation(s)
- Chioma Izzi-Engbeaya
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Sophie Jones
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Yoshibye Crustna
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Pratibha C Machenahalli
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Deborah Papadopoulou
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Manish Modi
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Christos Panayi
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Jessica Starikova
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Pei Chia Eng
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Maria Phylactou
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Edouard Mills
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Lisa Yang
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Risheka Ratnasabapathy
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Mark Sykes
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Isabella Plumptre
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Ben Coumbe
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Victoria C Wing
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Ewa Pacuszka
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - Paul Bech
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - James Minnion
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
| | - George Tharakan
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
- Department of Acute Medicine, Imperial College Healthcare NHS Trust, London, UK
| | - Tricia Tan
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | | | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Department of Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
- Correspondence and Reprint Requests: Waljit S. Dhillo, MBBS, PhD, Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 0NN, UK. E-mail:
| |
Collapse
|
26
|
Smith NK, Hackett TA, Galli A, Flynn CR. GLP-1: Molecular mechanisms and outcomes of a complex signaling system. Neurochem Int 2019; 128:94-105. [PMID: 31002893 PMCID: PMC7081944 DOI: 10.1016/j.neuint.2019.04.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/26/2019] [Accepted: 04/15/2019] [Indexed: 12/15/2022]
Abstract
Meal ingestion provokes the release of hormones and transmitters, which in turn regulate energy homeostasis and feeding behavior. One such hormone, glucagon-like peptide-1 (GLP-1), has received significant attention in the treatment of obesity and diabetes due to its potent incretin effect. In addition to the peripheral actions of GLP-1, this hormone is able to alter behavior through the modulation of multiple neural circuits. Recent work that focused on elucidating the mechanisms and outcomes of GLP-1 neuromodulation led to the discovery of an impressive array of GLP-1 actions. Here, we summarize the many levels at which the GLP-1 signal adapts to different systems, with the goal being to provide a background against which to guide future research.
Collapse
Affiliation(s)
- Nicholas K Smith
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Troy A Hackett
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aurelio Galli
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Charles R Flynn
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
27
|
Hultman K, Scarlett JM, Baquero AF, Cornea A, Zhang Y, Salinas CBG, Brown J, Morton GJ, Whalen EJ, Grove KL, Koegler FH, Schwartz MW, Mercer AJ. The central fibroblast growth factor receptor/beta klotho system: Comprehensive mapping in Mus musculus and comparisons to nonhuman primate and human samples using an automated in situ hybridization platform. J Comp Neurol 2019; 527:2069-2085. [PMID: 30809795 DOI: 10.1002/cne.24668] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/25/2022]
Abstract
Central activation of fibroblast growth factor (FGF) receptors regulates peripheral glucose homeostasis and reduces food intake in preclinical models of obesity and diabetes. The current work was undertaken to advance our understanding of the receptor expression, as sites of ligand action by FGF19, FGF21, and FGF1 in the mammalian brain remains unresolved. Recent advances in automated RNAscope in situ hybridization and droplet digital PCR (ddPCR) technology allowed us to interrogate central FGFR/beta klotho (Klb) system at the cellular level in the mouse, with relevant comparisons to nonhuman primate and human brain. FGFR1-3 gene expression was broadly distributed throughout the CNS in Mus musculus, with FGFR1 exhibiting the greatest heterogeneity. FGFR4 expression localized only in the medial habenula and subcommissural organ of mice. Likewise, Klb mRNA was restricted to the suprachiasmatic nucleus (SCh) and select midbrain and hindbrain nuclei. ddPCR in the rodent hypothalamus confirmed that, although expression levels are indeed low for Klb, there is nonetheless a bonafide subpopulation of Klb+ cells in the hypothalamus. In NHP and human midbrain and hindbrain, Klb + cells are quite rare, as is expression of FGFR4. Collectively, these data provide the most robust central map of the FGFR/Klb system to date and highlight central regions that may be of critical importance to assess central ligand effects with pharmacological dosing, such as the putative interactions between the endocrine FGFs and FGFR1/Klb, or FGF19 with FGFR4.
Collapse
Affiliation(s)
| | - Jarrad M Scarlett
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington.,Department of Pediatric Gastroenterology & Hepatology, Seattle Children's Hospital, Seattle, Washington
| | - Arian F Baquero
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Anda Cornea
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Yu Zhang
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | | | - Jenny Brown
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Gregory J Morton
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Erin J Whalen
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Kevin L Grove
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Frank H Koegler
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Michael W Schwartz
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Aaron J Mercer
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| |
Collapse
|
28
|
Hill JW, Elias CF. Neuroanatomical Framework of the Metabolic Control of Reproduction. Physiol Rev 2019; 98:2349-2380. [PMID: 30109817 DOI: 10.1152/physrev.00033.2017] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A minimum amount of energy is required for basic physiological processes, such as protein biosynthesis, thermoregulation, locomotion, cardiovascular function, and digestion. However, for reproductive function and survival of the species, extra energy stores are necessary. Production of sex hormones and gametes, pubertal development, pregnancy, lactation, and parental care all require energy reserves. Thus the physiological systems that control energy homeostasis and reproductive function coevolved in mammals to support both individual health and species subsistence. In this review, we aim to gather scientific knowledge produced by laboratories around the world on the role of the brain in integrating metabolism and reproduction. We describe essential neuronal networks, highlighting key nodes and potential downstream targets. Novel animal models and genetic tools have produced substantial advances, but critical gaps remain. In times of soaring worldwide obesity and metabolic dysfunction, understanding the mechanisms by which metabolic stress alters reproductive physiology has become crucial for human health.
Collapse
Affiliation(s)
- Jennifer W Hill
- Center for Diabetes and Endocrine Research, Departments of Physiology and Pharmacology and of Obstetrics and Gynecology, University of Toledo College of Medicine , Toledo, Ohio ; and Departments of Molecular and Integrative Physiology and of Obstetrics and Gynecology, University of Michigan , Ann Arbor, Michigan
| | - Carol F Elias
- Center for Diabetes and Endocrine Research, Departments of Physiology and Pharmacology and of Obstetrics and Gynecology, University of Toledo College of Medicine , Toledo, Ohio ; and Departments of Molecular and Integrative Physiology and of Obstetrics and Gynecology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
29
|
Verma RK, Sriramaneni R, Pandey M, Chaudhury H, Gorain B, Gupta G. Current updates on pharmacological roles of glucagon-like peptide 1 in obesity. Panminerva Med 2018; 60:224-225. [DOI: 10.23736/s0031-0808.18.03479-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
30
|
Izzi‐Engbeaya C, Comninos AN, Clarke SA, Jomard A, Yang L, Jones S, Abbara A, Narayanaswamy S, Eng PC, Papadopoulou D, Prague JK, Bech P, Godsland IF, Bassett P, Sands C, Camuzeaux S, Gomez‐Romero M, Pearce JTM, Lewis MR, Holmes E, Nicholson JK, Tan T, Ratnasabapathy R, Hu M, Carrat G, Piemonti L, Bugliani M, Marchetti P, Johnson PR, Hughes SJ, James Shapiro AM, Rutter GA, Dhillo WS. The effects of kisspeptin on β-cell function, serum metabolites and appetite in humans. Diabetes Obes Metab 2018; 20:2800-2810. [PMID: 29974637 PMCID: PMC6282711 DOI: 10.1111/dom.13460] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/22/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023]
Abstract
AIMS To investigate the effect of kisspeptin on glucose-stimulated insulin secretion and appetite in humans. MATERIALS AND METHODS In 15 healthy men (age: 25.2 ± 1.1 years; BMI: 22.3 ± 0.5 kg m-2 ), we compared the effects of 1 nmol kg-1 h-1 kisspeptin versus vehicle administration on glucose-stimulated insulin secretion, metabolites, gut hormones, appetite and food intake. In addition, we assessed the effect of kisspeptin on glucose-stimulated insulin secretion in vitro in human pancreatic islets and a human β-cell line (EndoC-βH1 cells). RESULTS Kisspeptin administration to healthy men enhanced insulin secretion following an intravenous glucose load, and modulated serum metabolites. In keeping with this, kisspeptin increased glucose-stimulated insulin secretion from human islets and a human pancreatic cell line in vitro. In addition, kisspeptin administration did not alter gut hormones, appetite or food intake in healthy men. CONCLUSIONS Collectively, these data demonstrate for the first time a beneficial role for kisspeptin in insulin secretion in humans in vivo. This has important implications for our understanding of the links between reproduction and metabolism in humans, as well as for the ongoing translational development of kisspeptin-based therapies for reproductive and potentially metabolic conditions.
Collapse
Affiliation(s)
- Chioma Izzi‐Engbeaya
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Alexander N. Comninos
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
- Department of EndocrinologyImperial College Healthcare NHS TrustLondonUK
| | - Sophie A. Clarke
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Anne Jomard
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Lisa Yang
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Sophie Jones
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Shakunthala Narayanaswamy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Pei Chia Eng
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Deborah Papadopoulou
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Julia K. Prague
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Paul Bech
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Ian F. Godsland
- Section of Metabolic Medicine, Department of Medicine, Imperial College LondonSt Mary's HospitalLondonUK
| | | | - Caroline Sands
- The MRC‐NIHR National Phenome Centre and Imperial BRC Clinical Phenotyping Centre, Division of Computational, Systems and Digestive Medicine, Department of Surgery and CancerLondonUK
| | - Stephane Camuzeaux
- The MRC‐NIHR National Phenome Centre and Imperial BRC Clinical Phenotyping Centre, Division of Computational, Systems and Digestive Medicine, Department of Surgery and CancerLondonUK
| | - Maria Gomez‐Romero
- The MRC‐NIHR National Phenome Centre and Imperial BRC Clinical Phenotyping Centre, Division of Computational, Systems and Digestive Medicine, Department of Surgery and CancerLondonUK
| | - Jake T. M. Pearce
- The MRC‐NIHR National Phenome Centre and Imperial BRC Clinical Phenotyping Centre, Division of Computational, Systems and Digestive Medicine, Department of Surgery and CancerLondonUK
| | - Matthew R. Lewis
- The MRC‐NIHR National Phenome Centre and Imperial BRC Clinical Phenotyping Centre, Division of Computational, Systems and Digestive Medicine, Department of Surgery and CancerLondonUK
| | - Elaine Holmes
- The MRC‐NIHR National Phenome Centre and Imperial BRC Clinical Phenotyping Centre, Division of Computational, Systems and Digestive Medicine, Department of Surgery and CancerLondonUK
| | - Jeremy K. Nicholson
- The MRC‐NIHR National Phenome Centre and Imperial BRC Clinical Phenotyping Centre, Division of Computational, Systems and Digestive Medicine, Department of Surgery and CancerLondonUK
| | - Tricia Tan
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Risheka Ratnasabapathy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
- Imperial Pancreatic Islet Biology and Diabetes ConsortiumHammersmith Hospital, Imperial College LondonLondonUK
| | - Gaelle Carrat
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
- Imperial Pancreatic Islet Biology and Diabetes ConsortiumHammersmith Hospital, Imperial College LondonLondonUK
| | - Lorenzo Piemonti
- Diabetes Research Institute (SR‐DRI), IRCCS San Raffaele Scientific InstituteMilanItaly
- Faculty of MedicineVita‐Salute San Raffaele UniversityMilanItaly
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, Islet Cell LaboratoryUniversity of PisaPisaItaly
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell LaboratoryUniversity of PisaPisaItaly
| | - Paul R. Johnson
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
- Oxford Centre for Diabetes, Endocrinology, and MetabolismUniversity of OxfordOxfordUK
- National Institute of Health Research Oxford Biomedical Research Centre, Churchill HospitalOxfordUK
| | - Stephen J. Hughes
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
- Oxford Centre for Diabetes, Endocrinology, and MetabolismUniversity of OxfordOxfordUK
- National Institute of Health Research Oxford Biomedical Research Centre, Churchill HospitalOxfordUK
| | - A. M. James Shapiro
- Clinical Islet Laboratory and Clinical Islet Transplant ProgramUniversity of AlbertaEdmontonCanada
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
- Imperial Pancreatic Islet Biology and Diabetes ConsortiumHammersmith Hospital, Imperial College LondonLondonUK
| | - Waljit S. Dhillo
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of MedicineImperial College LondonLondonUK
| |
Collapse
|
31
|
McEwen HJL, Cognard E, Ladyman SR, Khant-Aung Z, Tups A, Shepherd PR, Grattan DR. Feeding and GLP-1 receptor activation stabilize β-catenin in specific hypothalamic nuclei in male rats. J Neuroendocrinol 2018; 30:e12607. [PMID: 29752762 DOI: 10.1111/jne.12607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/03/2018] [Indexed: 12/21/2022]
Abstract
β-catenin is a multifunctional protein that can act in the canonical Wnt/β-catenin pathway to regulate gene expression but can also bind to cadherin proteins in adherens junctions where it plays a key role in regulating cytoskeleton linked with these junctions. Recently, evidence has been presented indicating an essential role for β-catenin in regulating trafficking of insulin vesicles in β-cells and showing that changes in nutrient levels rapidly alter levels of β-catenin in these cells. Given the importance of neuroendocrine hormone secretion in the regulation of whole body glucose homeostasis, the objective of this study was to investigate whether β-catenin signalling is regulated in the hypothalamus during the normal physiological response to food intake. Rats were subjected to a fasting/re-feeding paradigm, and then samples collected at specific timepoints for analysis of β-catenin expression by immunohistochemistry and Western blotting. Changes in gene expression were assessed by RT-qPCR. Using immunohistochemistry, feeding acutely increased detectable cytoplasmic levels of β-catenin ('stabilized β-catenin') in neurons in specific regions of the hypothalamus involved in metabolic regulation, including the arcuate, dorsomedial and paraventricular nuclei of the hypothalamus. Feeding-induced elevations in β-catenin in these nuclei were associated with increased transcription of several genes that are known to be responsive to Wnt/β-catenin signalling. The effect of feeding was mimicked by administration of the GLP-1 agonist exendin-4, and was characterized by cAMP-dependent phosphorylation of β-catenin at serine residues 552 and 675. The data suggest that β-catenin/TCF signalling is involved in metabolic sensing in the hypothalamus. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hayden J L McEwen
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Emmanuelle Cognard
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Sharon R Ladyman
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Zin Khant-Aung
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Alexander Tups
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Peter R Shepherd
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| |
Collapse
|
32
|
Wahab F, Atika B, Ullah F, Shahab M, Behr R. Metabolic Impact on the Hypothalamic Kisspeptin-Kiss1r Signaling Pathway. Front Endocrinol (Lausanne) 2018; 9:123. [PMID: 29643834 PMCID: PMC5882778 DOI: 10.3389/fendo.2018.00123] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
A large body of data has established the hypothalamic kisspeptin (KP) and its receptor, KISS1R, as major players in the activation of the neuroendocrine reproductive axis at the time of puberty and maintenance of reproductive capacity in the adult. Due to its strategic location, this ligand-receptor pair acts as an integrator of cues from gonadal steroids as well as of circadian and seasonal variation-related information on the reproductive axis. Besides these cues, the activity of the hypothalamic KP signaling is very sensitive to the current metabolic status of the body. In conditions of energy imbalance, either positive or negative, a number of alterations in the hypothalamic KP signaling pathway have been documented in different mammalian models including nonhuman primates and human. Deficiency of metabolic fuels during fasting causes a marked reduction of Kiss1 gene transcript levels in the hypothalamus and, hence, decreases the output of KP-containing neurons. Food intake or exogenous supply of metabolic cues, such as leptin, reverses metabolic insufficiency-related changes in the hypothalamic KP signaling. Likewise, alterations in Kiss1 expression have also been reported in other situations of energy imbalance like diabetes and obesity. Information related to the body's current metabolic status reaches to KP neurons both directly as well as indirectly via a complex network of other neurons. In this review article, we have provided an updated summary of the available literature on the regulation of the hypothalamic KP-Kiss1r signaling by metabolic cues. In particular, the potential mechanisms of metabolic impact on the hypothalamic KP-Kiss1r signaling, in light of available evidence, are discussed.
Collapse
Affiliation(s)
- Fazal Wahab
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- *Correspondence: Fazal Wahab,
| | - Bibi Atika
- Department of Developmental Biology, Faculty of Biology, University of Göttingen, Göttingen, Germany
| | - Farhad Ullah
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quiad-i-Azam University, Islamabad, Pakistan
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|