1
|
Giannakopoulos A, Chrysis D. Reversibility of disturbed pituitary function in pediatric conditions with psychological stressors: implications for clinical practice. Hormones (Athens) 2024; 23:709-716. [PMID: 38421589 DOI: 10.1007/s42000-024-00536-z] [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: 12/04/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
The complex communication network between the central nervous system and the hypothalamic-pituitary axis forms the basis of endocrine functional plasticity, which facilitates adaptation to changing internal and external conditions, but also makes it vulnerable to the negative effects of stressful psychological factors. Herein, clinical conditions such as functional hypothalamic amenorrhea, eating disorders, growth faltering, post-traumatic stress disorder, and pubertal disorders that may emerge during childhood or adolescence, their origin possibly including psychological stressors, are analyzed regarding their genetic susceptibility and reversibility of endocrine function. A discussion on the optimization of therapeutic management defined by managing stress and maximizing the degree and rate of reversibility follows.
Collapse
Affiliation(s)
- Aristeidis Giannakopoulos
- Division of Pediatric Endocrinology, Department of Pediatrics, Medical School of Patras, University Hospital, Rio, 26504, Patras, Greece.
| | - Dionisios Chrysis
- Division of Pediatric Endocrinology, Department of Pediatrics, Medical School of Patras, University Hospital, Rio, 26504, Patras, Greece
| |
Collapse
|
2
|
Liao C, Walters BW, DiStasio M, Lesch BJ. Human-specific epigenomic states in spermatogenesis. Comput Struct Biotechnol J 2024; 23:577-588. [PMID: 38274996 PMCID: PMC10809009 DOI: 10.1016/j.csbj.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/23/2023] [Accepted: 12/23/2023] [Indexed: 01/27/2024] Open
Abstract
Infertility is becoming increasingly common, affecting one in six people globally. Half of these cases can be attributed to male factors, many driven by abnormalities in the process of sperm development. Emerging evidence from genome-wide association studies, genetic screening of patient cohorts, and animal models highlights an important genetic contribution to spermatogenic defects, but comprehensive identification and characterization of the genes critical for male fertility remain lacking. High divergence of gene regulation in spermatogenic cells across species poses challenges for delineating the genetic pathways required for human spermatogenesis using common model organisms. In this study, we leveraged post-translational histone modification and gene transcription data for 15,491 genes in four mammalian species (human, rhesus macaque, mouse, and opossum), to identify human-specific patterns of gene regulation during spermatogenesis. We combined H3K27me3 ChIP-seq, H3K4me3 ChIP-seq, and RNA-seq data to define epigenetic states for each gene at two stages of spermatogenesis, pachytene spermatocytes and round spermatids, in each species. We identified 239 genes that are uniquely active, poised, or dynamically regulated in human spermatogenic cells distinct from the other three species. While some of these genes have been implicated in reproductive functions, many more have not yet been associated with human infertility and may be candidates for further molecular and epidemiologic studies. Our analysis offers an example of the opportunities provided by evolutionary and epigenomic data for broadly screening candidate genes implicated in reproduction, which might lead to discoveries of novel genetic targets for diagnosis and management of male infertility and male contraception.
Collapse
Affiliation(s)
- Caiyun Liao
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | | | - Marcello DiStasio
- Department of Pathology, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Department of Opthamology & Visual Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Bluma J. Lesch
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Department of Genetics, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
- Yale Cancer Center, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
3
|
Karaman V, Karakilic-Ozturan E, Poyrazoglu S, Gelmez MY, Bas F, Darendeliler F, Uyguner ZO. Novel variants ensued genomic imprinting in familial central precocious puberty. J Endocrinol Invest 2024; 47:2041-2052. [PMID: 38367171 PMCID: PMC11266277 DOI: 10.1007/s40618-023-02300-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/29/2023] [Indexed: 02/19/2024]
Abstract
INTRODUCTION Central precocious puberty (CPP) is characterized by the early onset of puberty and is associated with the critical processes involved in the pubertal switch. The puberty-related gene pool in the human genome is considerably large though few have been described in CPP. Within those genes, the genomic imprinting features of the MKRN3 and DLK1 genes add additional complexity to the understanding of the pathologic pathways. This study aimed to investigate the molecular etiology in the CPP cohort. METHODS Eighteen familial CPP cases were investigated by Sanger sequencing for five CPP-related genes; DLK1, KISS1, KISS1R, MKRN3, and PROKR2. Segregation analysis was performed in all patients with pathogenic variants. Using an ELISA test, the functional pathogenicity of novel variants was also investigated in conjunction with serum delta-like 1 homolog (DLK1) concentrations. RESULTS In three probands, a known variant in the MKRN3 gene (c.982C>T/p.(Arg328Cys)) and two novel variants in the DLK1 gene (c.357C>G/p.(Tyr119Ter) and c.67+78C>T) were identified. All three were inherited from the paternal allele. The individuals carrying the DLK1 variants had low detectable DLK1 levels in their serum. CONCLUSIONS The frequencies were 5.5% (1/18) for MKRN3 11% (2/18) for DLK1, and none for either KISS1, KISS1R, and PROKR2. Low serum DLK1 levels in affected individuals supported the relationship between here described novel DLK1 gene variants with CPP. Nonsense nature of c.357C>G/p.(Tyr119Ter) and an alteration in the evolutionarily conserved nucleotide c.67+78C>T suggested the disruptive nature of the variant's compatibility with CPP.
Collapse
Affiliation(s)
- V Karaman
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Millet Cad. Çapa/Fatih, 34096, Istanbul, Turkey.
| | - E Karakilic-Ozturan
- Department of Pediatric Endocrinology and Diabetes, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - S Poyrazoglu
- Department of Pediatric Endocrinology and Diabetes, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - M Y Gelmez
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - F Bas
- Department of Pediatric Endocrinology and Diabetes, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - F Darendeliler
- Department of Pediatric Endocrinology and Diabetes, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Z O Uyguner
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Millet Cad. Çapa/Fatih, 34096, Istanbul, Turkey
| |
Collapse
|
4
|
Rohayem J, Alexander EC, Heger S, Nordenström A, Howard SR. Mini-Puberty, Physiological and Disordered: Consequences, and Potential for Therapeutic Replacement. Endocr Rev 2024; 45:460-492. [PMID: 38436980 PMCID: PMC11244267 DOI: 10.1210/endrev/bnae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Indexed: 03/05/2024]
Abstract
There are 3 physiological waves of central hypothalamic-pituitary-gonadal (HPG) axis activity over the lifetime. The first occurs during fetal life, the second-termed "mini-puberty"-in the first months after birth, and the third at puberty. After adolescence, the axis remains active all through adulthood. Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by a deficiency in hypothalamic gonadotropin-releasing hormone (GnRH) secretion or action. In cases of severe CHH, all 3 waves of GnRH pulsatility are absent. The absence of fetal HPG axis activation manifests in around 50% of male newborns with micropenis and/or undescended testes (cryptorchidism). In these boys, the lack of the mini-puberty phase accentuates testicular immaturity. This is characterized by a low number of Sertoli cells, which are important for future reproductive capacity. Thus, absent mini-puberty will have detrimental effects on later fertility in these males. The diagnosis of CHH is often missed in infants, and even if recognized, there is no consensus on optimal therapeutic management. Here we review physiological mini-puberty and consequences of central HPG axis disorders; provide a diagnostic approach to allow for early identification of these conditions; and review current treatment options for replacement of mini-puberty in male infants with CHH. There is evidence from small case series that replacement with gonadotropins to mimic "mini-puberty" in males could have beneficial outcomes not only regarding testis descent, but also normalization of testis and penile sizes. Moreover, such therapeutic replacement regimens in disordered mini-puberty could address both reproductive and nonreproductive implications.
Collapse
Affiliation(s)
- Julia Rohayem
- Department of Pediatric Endocrinology and Diabetology, Children's Hospital of Eastern Switzerland, 9006 St. Gallen, Switzerland
- University of Muenster, 48149 Muenster, Germany
| | - Emma C Alexander
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sabine Heger
- Department of Pediatric Endocrinology, Children's Hospital Auf der Bult, 30173 Hannover, Germany
| | - Anna Nordenström
- Pediatric Endocrinology, Karolinska Institutet, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
- Department of Paediatric Endocrinology, Royal London Children's Hospital, Barts Health NHS Trust, London E1 1FR, UK
| |
Collapse
|
5
|
Canton APM, Seraphim CE, Montenegro LR, Krepischi ACV, Mendonca BB, Latronico AC, Brito VN. The genetic etiology is a relevant cause of central precocious puberty. Eur J Endocrinol 2024; 190:479-488. [PMID: 38857188 DOI: 10.1093/ejendo/lvae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/14/2024] [Accepted: 06/08/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVES The etiology of central precocious puberty (CPP) has expanded with identification of new genetic causes, including the monogenic deficiency of Makorin-Ring-Finger-Protein-3 (MKRN3). We aimed to assess the prevalence of CPP causes and the predictors of genetic involvement in this phenotype. DESIGN A retrospective cohort study for an etiological survey of patients with CPP from a single academic center. METHODS All patients with CPP had detailed medical history, phenotyping, and brain magnetic resonance imaging (MRI); those with negative brain MRI (apparently idiopathic) were submitted to genetic studies, mainly DNA sequencing studies, genomic microarray, and methylation analysis. RESULTS We assessed 270 patients with CPP: 50 (18.5%) had CPP-related brain lesions (34 [68%] congenital lesions), whereas 220 had negative brain MRI. Of the latter, 174 (165 girls) were included for genetic studies. Genetic etiologies were identified in 22 patients (20 girls), indicating an overall frequency of genetic CPP of 12.6% (22.2% in boys and 12.1% in girls). The most common genetic defects were MKRN3, Delta-Like-Non-Canonical-Notch-Ligand-1 (DLK1), and Methyl-CpG-Binding-Protein-2 (MECP2) loss-of-function mutations, followed by 14q32.2 defects (Temple syndrome). Univariate logistic regression identified family history (odds ratio [OR] 3.3; 95% CI 1.3-8.3; P = .01) and neurodevelopmental disorders (OR 4.1; 95% CI 1.3-13.5; P = .02) as potential clinical predictors of genetic CPP. CONCLUSIONS Distinct genetic causes were identified in 12.6% patients with apparently idiopathic CPP, revealing the genetic etiology as a relevant cause of CPP in both sexes. Family history and neurodevelopmental disorders were suggested as predictors of genetic CPP. We originally proposed an algorithm to investigate the etiology of CPP including genetic studies.
Collapse
Affiliation(s)
- Ana Pinheiro Machado Canton
- Developmental Endocrinology Unit, Hormones and Molecular Genetics Laboratory LIM/42, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 05403-000 Sao Paulo, Brazil
- Cellular and Molecular Endocrinology Laboratory LIM/25, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 01246-903 Sao Paulo, Brazil
| | - Carlos Eduardo Seraphim
- Developmental Endocrinology Unit, Hormones and Molecular Genetics Laboratory LIM/42, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 05403-000 Sao Paulo, Brazil
- Cellular and Molecular Endocrinology Laboratory LIM/25, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 01246-903 Sao Paulo, Brazil
| | - Luciana Ribeiro Montenegro
- Developmental Endocrinology Unit, Hormones and Molecular Genetics Laboratory LIM/42, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 05403-000 Sao Paulo, Brazil
- Cellular and Molecular Endocrinology Laboratory LIM/25, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 01246-903 Sao Paulo, Brazil
| | | | - Berenice Bilharinho Mendonca
- Developmental Endocrinology Unit, Hormones and Molecular Genetics Laboratory LIM/42, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 05403-000 Sao Paulo, Brazil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo, 01246-903 Sao Paulo, Brazil
| | - Ana Claudia Latronico
- Developmental Endocrinology Unit, Hormones and Molecular Genetics Laboratory LIM/42, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 05403-000 Sao Paulo, Brazil
- Cellular and Molecular Endocrinology Laboratory LIM/25, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 01246-903 Sao Paulo, Brazil
| | - Vinicius Nahime Brito
- Developmental Endocrinology Unit, Hormones and Molecular Genetics Laboratory LIM/42, Clinicas Hospital, Discipline of Endocrinology and Metabolism, School of Medicine, University of Sao Paulo, 05403-000 Sao Paulo, Brazil
| |
Collapse
|
6
|
Robilliard R, Lee PA, Swartz Topor L. Diagnosis, Treatment, and Outcomes of Males with Central Precocious Puberty. Endocrinol Metab Clin North Am 2024; 53:239-250. [PMID: 38677867 DOI: 10.1016/j.ecl.2024.01.005] [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] [Indexed: 04/29/2024]
Abstract
Central precocious puberty (CPP) among males is less frequent than among females but more likely to have an underlying pathologic cause. Diagnosis of CPP is often straightforward among males because increased testicular volume, the first sign of puberty, can be verified although careful central nervous system (CNS) assessment is generally necessary. Treatment with gonadotropin-releasing hormone agonist (GnRHa) is indicated, given in conjunction with any therapy needed for CNS lesions. Monitoring of treatment usually can consist of evaluating growth and physical puberty and with testosterone levels as the only lab data. Short-term and long-term outcome data indicate efficacy and safety, although data are limited. Such data need to be reported.
Collapse
Affiliation(s)
- Renée Robilliard
- Division of Pediatric Endocrinology and Diabetes, Hasbro Children's Hospital, Providence, RI, USA; Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter A Lee
- Division of Pediatric Endocrinology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA.
| | - Lisa Swartz Topor
- Division of Pediatric Endocrinology and Diabetes, Hasbro Children's Hospital, Providence, RI, USA; Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
7
|
Salmeri N, Viganò P, Cavoretto P, Marci R, Candiani M. The kisspeptin system in and beyond reproduction: exploring intricate pathways and potential links between endometriosis and polycystic ovary syndrome. Rev Endocr Metab Disord 2024; 25:239-257. [PMID: 37505370 DOI: 10.1007/s11154-023-09826-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Endometriosis and polycystic ovary syndrome (PCOS) are two common female reproductive disorders with a significant impact on the health and quality of life of women affected. A novel hypothesis by evolutionary biologists suggested that these two diseases are inversely related to one another, representing a pair of diametrical diseases in terms of opposite alterations in reproductive physiological processes but also contrasting phenotypic traits. However, to fully explain the phenotypic features observed in women with these conditions, we need to establish a potential nexus system between the reproductive system and general biological functions. The recent discovery of kisspeptin as pivotal mediator of internal and external inputs on the hypothalamic-pituitary-gonadal axis has led to a new understanding of the neuroendocrine upstream regulation of the human reproductive system. In this review, we summarize the current knowledge on the physiological roles of kisspeptin in human reproduction, as well as its involvement in complex biological functions such as metabolism, inflammation and pain sensitivity. Importantly, these functions are known to be dysregulated in both PCOS and endometriosis. Within the evolving scientific field of "kisspeptinology", we critically discuss the clinical relevance of these discoveries and their potential translational applications in endometriosis and PCOS. By exploring the possibilities of manipulating this complex signaling system, we aim to pave the way for novel targeted therapies in these reproductive diseases.
Collapse
Affiliation(s)
- Noemi Salmeri
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Paola Viganò
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via M. Fanti 6, 20122, Milan, Italy.
| | - Paolo Cavoretto
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Roberto Marci
- Gynecology & Obstetrics, University of Ferrara, 44121, Ferrara, Italy
| | - Massimo Candiani
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| |
Collapse
|
8
|
Wei SM, Gregory MD, Nash T, de Abreu e Gouvêa A, Mervis CB, Cole KM, Garvey MH, Kippenhan JS, Eisenberg DP, Kolachana B, Schmidt PJ, Berman KF. Altered pubertal timing in 7q11.23 copy number variations and associated genetic mechanisms. iScience 2024; 27:109113. [PMID: 38375233 PMCID: PMC10875153 DOI: 10.1016/j.isci.2024.109113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Accepted: 01/31/2024] [Indexed: 02/21/2024] Open
Abstract
Pubertal timing, including age at menarche (AAM), is a heritable trait linked to lifetime health outcomes. Here, we investigate genetic mechanisms underlying AAM by combining genome-wide association study (GWAS) data with investigations of two rare genetic conditions clinically associated with altered AAM: Williams syndrome (WS), a 7q11.23 hemideletion characterized by early puberty; and duplication of the same genes (7q11.23 Duplication syndrome [Dup7]) characterized by delayed puberty. First, we confirm that AAM-derived polygenic scores in typically developing children (TD) explain a modest amount of variance in AAM (R2 = 0.09; p = 0.04). Next, we demonstrate that 7q11.23 copy number impacts AAM (WS < TD < Dup7; p = 1.2x10-8, η2 = 0.45) and pituitary volume (WS < TD < Dup7; p = 3x10-5, ηp2 = 0.2) with greater effect sizes. Finally, we relate an AAM-GWAS signal in 7q11.23 to altered expression in postmortem brains of STAG3L2 (p = 1.7x10-17), a gene we also find differentially expressed with 7q11.23 copy number (p = 0.03). Collectively, these data explicate the role of 7q11.23 in pubertal onset, with STAG3L2 and pituitary development as potential mediators.
Collapse
Affiliation(s)
- Shau-Ming Wei
- Behavioral Endocrinology Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Michael D. Gregory
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany Nash
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Andrea de Abreu e Gouvêa
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Carolyn B. Mervis
- Neurodevelopmental Sciences Laboratory, Department of Psychological and Brain Sciences, University of Louisville, Louisville, KY, USA
| | - Katherine M. Cole
- Behavioral Endocrinology Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Madeline H. Garvey
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - J. Shane Kippenhan
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Daniel P. Eisenberg
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Bhaskar Kolachana
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Peter J. Schmidt
- Behavioral Endocrinology Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Karen F. Berman
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Banerjee AA, Bhanarkar SR, Keshwani R, Pande S, Modi DN, Mehta A, Bombe S, Pathak BR, Joshi B, Tandon D, Patil A, Begum S, Chauhan S, Mahale SD, Rao S, Surve SV. Relevance of augmented kisspeptin signaling through H 364 KISS1R in central precocious puberty. Gene 2024; 895:148016. [PMID: 37981083 DOI: 10.1016/j.gene.2023.148016] [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: 08/03/2023] [Revised: 10/28/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
Understanding the pathophysiology of idiopathic central precocious puberty (ICPP) is essential, in view of its consequences on reproductive health and metabolic disorders in later life. Towards this, estimation of circulating levels of the neuropeptides, viz; Kisspeptin (Kp-10), Neurokinin B (NKB) and Neuropeptide Y (NPY), acting upstream to Gonadotropin-Releasing Hormone (GnRH), has shown promise. Insights can also be gained from functional studies on genetic variations implicated in ICPP. This study investigated the pathophysiology of ICPP in a girl by exploring the therapeutic relevance of the circulating levels of Kp-10, NKB, NPY and characterizing the nonsynonymous KISS1R variant, L364H, that she harbours, in a homozygous condition. Plasma levels of Kp-10, NKB and NPY before and after GnRH analog (GnRHa) treatment, were determined by ELISA. It was observed that GnRHa treatment resulted in suppression of circulating levels of Kp-10, NKB and NPY. Further, the H364 variant in KISS1R was generated by site directed mutagenesis. Post transient transfection of either L364 or H364 KISS1R variant in CHO cells, receptor expression was ascertained by western blotting, indirect immunofluorescence and flow cytometry. Kp-10 stimulated signalling response was also determined by phospho-ERK and inositol phosphate production. Structure-function studies revealed that, although the receptor expression in H364 KISS1R was comparable to L364 KISS1R, there was an enhanced signalling response through this variant at high doses of Kp-10. Thus, elevated levels of Kp-10, acting through H364 KISS1R, contributed to the manifestation of ICPP, providing further evidence that dysregulation of Kp-10/KISS1R axis impacts the onset of puberty.
Collapse
Affiliation(s)
- Antara A Banerjee
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Shital R Bhanarkar
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Rachna Keshwani
- Bai Jerbai Wadia Hospital For Children, Acharya Donde Marg, Parel, Mumbai 400 012, India
| | - Shailesh Pande
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Deepak N Modi
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Amrita Mehta
- Bai Jerbai Wadia Hospital For Children, Acharya Donde Marg, Parel, Mumbai 400 012, India
| | - Shweta Bombe
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Bhakti R Pathak
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Beena Joshi
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Deepti Tandon
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Anushree Patil
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Shahina Begum
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sanjay Chauhan
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Smita D Mahale
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sudha Rao
- Bai Jerbai Wadia Hospital For Children, Acharya Donde Marg, Parel, Mumbai 400 012, India.
| | - Suchitra V Surve
- ICMR-National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, Mumbai 400 012, India.
| |
Collapse
|
10
|
Patel B, Koysombat K, Mills EG, Tsoutsouki J, Comninos AN, Abbara A, Dhillo WS. The Emerging Therapeutic Potential of Kisspeptin and Neurokinin B. Endocr Rev 2024; 45:30-68. [PMID: 37467734 PMCID: PMC10765167 DOI: 10.1210/endrev/bnad023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Kisspeptin (KP) and neurokinin B (NKB) are neuropeptides that govern the reproductive endocrine axis through regulating hypothalamic gonadotropin-releasing hormone (GnRH) neuronal activity and pulsatile GnRH secretion. Their critical role in reproductive health was first identified after inactivating variants in genes encoding for KP or NKB signaling were shown to result in congenital hypogonadotropic hypogonadism and a failure of pubertal development. Over the past 2 decades since their discovery, a wealth of evidence from both basic and translational research has laid the foundation for potential therapeutic applications. Beyond KP's function in the hypothalamus, it is also expressed in the placenta, liver, pancreas, adipose tissue, bone, and limbic regions, giving rise to several avenues of research for use in the diagnosis and treatment of pregnancy, metabolic, liver, bone, and behavioral disorders. The role played by NKB in stimulating the hypothalamic thermoregulatory center to mediate menopausal hot flashes has led to the development of medications that antagonize its action as a novel nonsteroidal therapeutic agent for this indication. Furthermore, the ability of NKB antagonism to partially suppress (but not abolish) the reproductive endocrine axis has supported its potential use for the treatment of various reproductive disorders including polycystic ovary syndrome, uterine fibroids, and endometriosis. This review will provide a comprehensive up-to-date overview of the preclinical and clinical data that have paved the way for the development of diagnostic and therapeutic applications of KP and NKB.
Collapse
Affiliation(s)
- Bijal Patel
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Kanyada Koysombat
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Edouard G Mills
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Jovanna Tsoutsouki
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Alexander N Comninos
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Ali Abbara
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| |
Collapse
|
11
|
Nisa KU, Tarfeen N, Mir SA, Waza AA, Ahmad MB, Ganai BA. Molecular Mechanisms in the Etiology of Polycystic Ovary Syndrome (PCOS): A Multifaceted Hypothesis Towards the Disease with Potential Therapeutics. Indian J Clin Biochem 2024; 39:18-36. [PMID: 38223007 PMCID: PMC10784448 DOI: 10.1007/s12291-023-01130-7] [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: 11/02/2022] [Accepted: 03/01/2023] [Indexed: 03/28/2023]
Abstract
Among the premenopausal women, Polycystic Ovary Syndrome (PCOS) is the most prevalent endocrinopathy affecting the reproductive system and metabolic rhythms leading to disrupted menstrual cycle. Being heterogeneous in nature it is characterized by complex symptomology of oligomennorhoea, excess of androgens triggering masculine phenotypic appearance and/or multiple follicular ovaries. The etiology of this complex disorder remains somewhat doubtful and the researchers hypothesize multisystem links in the pathogenesis of this disease. In this review, we attempt to present several hypotheses that tend to contribute to the etiology of PCOS. Metabolic inflexibility, aberrant pattern of gonadotropin signaling along with the evolutionary, genetic and environmental factors have been discussed. Considered a lifelong endocrinological implication, no universal treatment is available for PCOS so far however; multiple drug therapy is often advised along with simple life style intervention is mainly advised to manage its cardinal symptoms. Here we aimed to present a summarized view of pathophysiological links of PCOS with potential therapeutic strategies.
Collapse
Affiliation(s)
- Khair Ul Nisa
- Department of Environmental Science, University of Kashmir, Srinagar, 190006 India
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, 190006 India
| | - Najeebul Tarfeen
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, 190006 India
| | - Shahnaz Ahmad Mir
- Department of Endocrinology, Government Medical College, Shireen Bagh, Srinagar, 190010 India
| | - Ajaz Ahmad Waza
- Multidisciplinary Research Unit (MRU), Government Medical Collage (GMC), Srinagar, 190010 India
| | - Mir Bilal Ahmad
- Department of Biochemistry, University of Kashmir, Srinagar, 190006 India
| | - Bashir Ahmad Ganai
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, 190006 India
| |
Collapse
|
12
|
Kumar V, Doshi G. Revolutionizing Infertility Management through Novel Peptide-based Targets. Curr Protein Pept Sci 2024; 25:738-752. [PMID: 38778605 DOI: 10.2174/0113892037304433240430144106] [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: 01/15/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
Around 48 million couples and 186 million people worldwide have infertility; of these, approximately 85% have an identifiable cause, the most common being ovulatory dysfunctions, male infertility, polycystic ovary syndrome, and tubule disease. The remaining 15% have infertility for unknown reasons, including lifestyle and environmental factors. The regulation of the hypothalamic- pituitary-adrenal axis (HPA) is crucial for the secretion of gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH), and follicle-stimulating hormone (FSH), which are essential for female reproductive functions. GnRH is the primary reproductive axis regulator. The pattern of GnRH, FSH, and LH release is determined by its pulsatile secretion, which in turn controls endocrine function and gamete maturation in the gonads. Peptides called Kisspeptin (KP), Neurokinin-B (NKB), and Orexin influence both positive and negative feedback modulation of GnRH, FSH, and LH secretion in reproduction. This review article mainly focuses on the historical perspective, isoform, and signaling pathways of KP, NKB, and Orexin novel peptide-based targets including clinical and preclinical studies and having a promising effect in the management of infertility.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400 056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400 056, India
| |
Collapse
|
13
|
Song J, Choi SY. Arcuate Nucleus of the Hypothalamus: Anatomy, Physiology, and Diseases. Exp Neurobiol 2023; 32:371-386. [PMID: 38196133 PMCID: PMC10789173 DOI: 10.5607/en23040] [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: 12/10/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024] Open
Abstract
The hypothalamus is part of the diencephalon and has several nuclei, one of which is the arcuate nucleus. The arcuate nucleus of hypothalamus (ARH) consists of neuroendocrine neurons and centrally-projecting neurons. The ARH is the center where the homeostasis of nutrition/metabolism and reproduction are maintained. As such, dysfunction of the ARH can lead to disorders of nutrition/metabolism and reproduction. Here, we review various types of neurons in the ARH and several genetic disorders caused by mutations in the ARH.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea
| |
Collapse
|
14
|
Zevin EL, Eugster EA. Central precocious puberty: a review of diagnosis, treatment, and outcomes. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:886-896. [PMID: 37973253 DOI: 10.1016/s2352-4642(23)00237-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/23/2023] [Accepted: 09/08/2023] [Indexed: 11/19/2023]
Abstract
Central precocious puberty (CPP) refers to early activation of the hypothalamic-pituitary-gonadal (HPG) axis and is manifested by breast development in girls or testicular enlargement in boys before the normal physiological age ranges. CPP can be precipitated by intracranial pathology, exposure to high levels of sex steroids, or environmental risk factors, but most cases are idiopathic. Monogenic causes have also been identified. In this Review, we summarise pathophysiology, risk factors, diagnosis, and management of CPP. Concern for CPP should prompt referral to paediatric endocrinology where diagnosis is confirmed by clinical, biochemical, radiological, and genetic testing. CPP is treated with a gonadotropin-releasing hormone analogue, the primary aims of which are to increase adult height and postpone development of secondary sexual characteristics to an age that is more commensurate with peers. Although long-term outcomes of treatment with gonadotropin-releasing hormone analogues are reassuring, additional research on the psychological effect of CPP is needed.
Collapse
Affiliation(s)
- Erika L Zevin
- Division of Pediatric Endocrinology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Erica A Eugster
- Division of Pediatric Endocrinology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
15
|
Shi H, Yan Z, Du H, Tang Y, Song K, Yang Q, Huang X, Wang P, Gao X, Yang J, Gun S. Regulatory Effects of the Kiss1 Gene in the Testis on Puberty and Reproduction in Hezuo and Landrance Boars. Int J Mol Sci 2023; 24:16700. [PMID: 38069021 PMCID: PMC10705963 DOI: 10.3390/ijms242316700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Kisspeptin, a neuropeptide encoded by the Kiss1 gene, combines with its receptor Kiss1R to regulate the onset of puberty and male fertility by the hypothalamic-pituitary-gonadal axis. However, little is known regarding the expression signatures and molecular functions of Kiss1 in the testis. H&E staining revealed that well-arranged spermatogonia, spermatocytes, round and elongated spermatids, and spermatozoa, were observed in 4-, 6-, and 8-month-old testes compared to 1- and 3-month-old testes of Hezuo pigs; however, these were not observed in Landrance until 6 months. The diameter, perimeter, and cross-sectional area of seminiferous tubules and the perimeter and area of the tubular lumen increased gradually with age in both pigs. Still, Hezuo pigs grew faster than Landrance. The cloning results suggested that the Hezuo pigs' Kiss1 CDS region is 417 bp in length, encodes 138 amino acids, and is highly conserved in the kisspeptin-10 region. qRT-PCR and Western blot indicated that the expression trends of Kiss1 mRNA and protein were essentially identical, with higher expression levels at post-pubertal stages. Immunohistochemistry demonstrated that the Kiss1 protein was mainly located in Leydig cells and post-pubertal spermatogenic cells, ranging from round spermatids to spermatozoa. These studies suggest that Kiss1 is an essential regulator in the onset of puberty and spermatogenesis of boars.
Collapse
Affiliation(s)
- Haixia Shi
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Hong Du
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuran Tang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Kelin Song
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xiaoli Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiaojiao Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Research Center for Swine Production Engineering and Technology, Lanzhou 730070, China
| |
Collapse
|
16
|
Okafor IA, Mbagwu SI, Okafor US, Nweke JO, Ibeabuchi KC, Ogbonna SN. The methanolic extract of Hibiscus sabdariffa downregulates the relative expression of Kiss1 gene in the hypothalamus of Wistar rats: A preliminary report. AVICENNA JOURNAL OF PHYTOMEDICINE 2023; 13:575-584. [PMID: 38106630 PMCID: PMC10719723 DOI: 10.22038/ajp.2023.22692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/19/2023]
Abstract
Objective Kiss1 gene expression in the rat hypothalamus was investigated following administration of methanolic extract of Hibiscus sabdariffa (MEHS) to provide mechanistic evidence for the reproductive effect of the MEHS as a potential regulator of Kiss1 gene (which directly controls the hypogonadal axis). Materials and Methods This experiment was done using fifteen (15) male rats with average weight of 148 g, randomly grouped into three (3) groups (A-C). Group A was the control group and received no treatment. Group B and C were orally administered with 200 mg/kg and 400 mg/kg of MEHS, respectively. The animals received the extract once a day for twenty-one (21) days. The hypothalamus was harvested on the last day of administration to investigate antioxidant levels, histopathology, and Kiss1 gene expression. Results The relative expression of Kiss1 gene in the group C was downregulated compared to the control group (p=0.023). No significant changes were seen in the antioxidant levels of the groups treated with MEHS when compared to the control. MEHS had no histopathological effects in the hypothalamus at both low (200 mg/kg) and high (400 mg/kg) doses. Conclusion High-dose MEHS lowers the expression of the Kiss1 gene in the hypothalamus. However, this effect could not be explained by the oxidative profile or histology of the hypothalamus.
Collapse
Affiliation(s)
- Izuchukwu Azuka Okafor
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus, PMB 5001, Nnewi, Nigeria
- Department of Obstetrics and Gynaecology, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Pan African University of Life and Earth Science Institute (Including Health and Agriculture), PAULESI, University of Ibadan, Ibadan, Nigeria
| | - Smart Ikechukwu Mbagwu
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus, PMB 5001, Nnewi, Nigeria
- Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, Massachusetts
| | | | - Johnson Okwudili Nweke
- Diagnostic Laboratory Unit, Medical Centre, Michael Okpara University of Agriculture, Umudike, Abia State, Nigeria
| | | | - Samuel Nduka Ogbonna
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus, PMB 5001, Nnewi, Nigeria
| |
Collapse
|
17
|
Alghamdi A. Precocious Puberty: Types, Pathogenesis and Updated Management. Cureus 2023; 15:e47485. [PMID: 38021712 PMCID: PMC10663169 DOI: 10.7759/cureus.47485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2023] [Indexed: 12/01/2023] Open
Abstract
Precocious puberty (PP) means the appearance of secondary sexual characters before the age of eight years in girls and nine years in boys. Puberty is indicated in girls by the enlargement of the breasts (thelarche) in girls and in boys by the enlargement of the testes in either volume or length (testicular volume = 4 mL, testicular length = 25 mm, or both). Two types of PP are recognized - namely central PP (CPP) and peripheral PP (PPP). This paper aims to describe the clinical findings and laboratory workup of PP and to illustrate the new trends in the management of precocious sexual maturation. Gonadotropin-releasing hormone (GnRH)-independent type (PPP) refers to the development of early pubertal maturation not related to the central activation of the hypothalamic-pituitary-gonadal (HPG) axis. It is classified into genetic or acquired disorders. The most common forms of congenital or genetic causes involve McCune-Albright syndrome (MAS), familial male-limited PP, and congenital adrenal hyperplasia. The acquired causes include exogenous exposure to androgens, functioning tumors or cysts, and the pseudo-PP of profound primary hypothyroidism. On the other hand, CPP is the most common and it is a gonadotropin-dependent form. It is due to premature maturation of the HPG axis. CPP may occur as genetic alterations, such as MKRN3, DLK1, or KISS1;as a part of mutations in the epigenetic factors that regulate the HPG axis, such as Lin28b and let-7; or as a part of syndromes, central lesions such as hypothalamic hamartoma, and others. A full, detailed history and physical examination should be taken. Furthermore, several investigations should be conducted for both types of PP, including the estimation of serum gonadotropins such as luteinizing and follicle-stimulating hormones and sex steroids, in addition to a radiographic workup and thyroid function tests. Treatment depends on the type of PP: Long-acting GnRHa, either intramuscularly or implanted, is the norm of care for CPP management, while in PPP, especially in congenital adrenal hyperplasia, the goal of management is to suppress adrenal androgen secretion by glucocorticoids. In addition, anastrozole and letrozole - third-generation aromatase inhibitors - are more potent for MAS.
Collapse
Affiliation(s)
- Ahmed Alghamdi
- Pediatric Endocrinology, Faculty of Medicine, Al Baha University, Al Baha, SAU
| |
Collapse
|
18
|
Nikolettos K, Nikolettos N, Vlahos N, Pagonopoulou O, Asimakopoulos B. Role of leptin, adiponectin, and kisspeptin in polycystic ovarian syndrome pathogenesis. Minerva Obstet Gynecol 2023; 75:460-467. [PMID: 36255161 DOI: 10.23736/s2724-606x.22.05139-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
INTRODUCTION Polycystic ovarian syndrome (PCOS) affects 5-20% of females and is the most common cause of anovulatory infertility. Leptin seems to have an important role in reproduction. Many reproductive pathologies such as preeclampsia, PCOS, and endometriosis are associated to plasma adiponectin levels. Kisspeptin levels are increased in PCOS women. EVIDENCE ACQUISITION A review of the literature was completed through the PubMed database aiming to find articles regarding leptin, adiponectin and kisspeptin and if they are related to PCOS pathogenesis. EVIDENCE SYNTHESIS Even today it is not clear what is the role of leptin in women with PCOS, although most of the researchers found increased levels of leptin as well as leptin resistance in PCOS (both obese and lean individuals). Many more longitudinal studies should be done to discover the usefulness of measuring adiponectin in prepubertal women who apparently have a possibility to develop PCOS to find out if they finally develop PCOS. Most of the researchers found that PCOS women have decreased levels of adiponectin unrelated to BMI levels. Nevertheless, not all studies had the same result. Moreover, it is necessary more studies to be made to investigate the connection between kisspeptin and other metabolic factors such as LH and insulin resistance. CONCLUSIONS In general, it remains inconclusive whether leptin, adiponectin, and kisspeptin can be used as clinical and/or biochemical markers of PCOS. Therefore, it is essential to review the current data with regards to the association between PCOS and circulating leptin, adiponectin, and kisspeptin in women with PCOS.
Collapse
Affiliation(s)
- Konstantinos Nikolettos
- Obstetric and Gynecologic Clinic, Medical School, Democritus University of Thrace, Alexandroupolis, Greece -
| | - Nikos Nikolettos
- Obstetric and Gynecologic Clinic, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Nikolaos Vlahos
- Second Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, School of Medicine, Aretaieion Hospital, Athens, Greece
| | - Olga Pagonopoulou
- Laboratory of Physiology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Byron Asimakopoulos
- Laboratory of Physiology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
19
|
Klein KO, Mauras N, Nayak S, Sunil B, Martinez-Placencia BM, Dragnic S, Ballina M, Zhou Q, Kansra AR. Efficacy and Safety of Leuprolide Acetate 6-Month Depot for the Treatment of Central Precocious Puberty: A Phase 3 Study. J Endocr Soc 2023; 7:bvad071. [PMID: 37334213 PMCID: PMC10274571 DOI: 10.1210/jendso/bvad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Indexed: 06/20/2023] Open
Abstract
Context Treatment options for central precocious puberty (CPP) are important for individualization of therapy. Objective We evaluated the efficacy and safety of 6-month 45-mg leuprolide acetate (LA) depot with intramuscular administration. Methods LA depot was administered at weeks 0 and 24 to treatment-naïve (n = 27) or previously treated (n = 18) children with CPP in a phase 3, multicenter, single-arm, open-label study (NCT03695237). Week 24 peak-stimulated luteinizing hormone (LH) suppression (<4 mIU/mL) was the primary outcome. Secondary/other outcomes included basal sex hormone suppression (girls, estradiol <20 pg/mL; boys, testosterone <30 ng/dL), suppression of physical signs, height velocity, bone age, patient/parent-reported outcomes, and adverse events. Results All patients (age, 7.8 ± 1.27 years) received both scheduled study doses. At 24 weeks, 39/45 patients (86.7%) had LH suppressed. Six were counted as unsuppressed; 2 because of missing data, 3 with LH of 4.35-5.30 mIU/mL and 1 with LH of 21.07 mIU/mL. Through 48 weeks, LH, estradiol, and testosterone suppression was achieved in ≥86.7%, ≥97.4%, and 100%, respectively (as early as week 4 for LH and estradiol and week 12 for testosterone). Physical signs were suppressed at week 48 (girls, 90.2%; boys, 75.0%). Mean height velocity ranged 5.0 to 5.3 cm/year post-baseline in previously treated patients and declined from 10.1 to 6.5 cm/year at week 20 in treatment-naïve patients. Mean bone age advanced slower than chronological age. Patient/parent-reported outcomes remained stable. No new safety signals were identified. No adverse event led to treatment discontinuation. Conclusion Six-month intramuscular LA depot demonstrated 48-week efficacy with a safety profile consistent with other GnRH agonist formulations.
Collapse
Affiliation(s)
- Karen O Klein
- University of California, San Diego, and Rady Children's Hospital San Diego, San Diego, CA 92121, USA
| | - Nelly Mauras
- Nemours Children's Health, Jacksonville, FL 32207, USA
| | - Sunil Nayak
- Pediatric Endocrine Associates, Greenwood Village, CO 80111, USA
| | - Bhuvana Sunil
- Mary Bridge Children's Hospital, Tacoma, WA 98403, USA
| | | | | | | | - Qing Zhou
- AbbVie, Inc., North Chicago, IL 60064, USA
| | - Alvina R Kansra
- Correspondence: Alvina R. Kansra, MD, Associate Medical Director, TAMD, Therapeutic Area, AbbVie, Inc., 1 North Waukegan Road, Bldg. AP31, North Chicago, IL 60064, USA.
| |
Collapse
|
20
|
Khabibullina DA, Kolodkina AA, Vizerov TV, Zubkova NA, Bezlepkina OB. [Gonadotropin-dependent precocious puberty: genetic and clinical characteristics]. PROBLEMY ENDOKRINOLOGII 2023; 69:58-66. [PMID: 37448272 DOI: 10.14341/probl13215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/12/2023] [Accepted: 01/22/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND In 90% cases of girls and 25-60% cases of boys the cause of gonadotropin-dependent precocious puberty (PP) is unclear. Up to 25-27.5% of gonadotropin-dependent PP cases are monogenic and suggest autosomal-dominant inheritance with incomplete sex-dependent penetrance. To date, mutations in genes KISS1, KISS1R, MKRN3, DLK1 have been described as causal variants leading to precocious hypothalamic-pituitary axis activation in childhood. Genetic testing in patients with hereditary forms of PP can expand our knowledge of underlying molecular mechanisms of the disease and it is also necessary for genetic counselling. AIM To study clinical features and genetic characteristics of patients with idiopathic gonadotropin-dependent precocious puberty. MATERIALS AND METHODS A group of patients with idiopathic gonadotropin-dependent precocious puberty and positive family history (early or precocious puberty) was examined. Laboratory and instrumental diagnostic tests, full-exome sequencing (NGS, next-generation sequencing) were provided for all patients. RESULTS The study included 30 patients (29 girls, 1 boy) with idiopathic gonadotropin-dependent precocious puberty. The median of patients age at the time of the examination was 7,2 years [6,5; 7,7]. Positive family history presented in all cases: in 40% of patients on father's side, in 37% - on mother's side, in 23% of patients PP was diagnosed in siblings. The fullexome sequencing was conducted to 21 patients: in 61,9% of cases (95% CI [40;79]) nucleotide variants were identified in genes, associated with gonadotropin-dependent precocious puberty. MKRN3 gene defect was detected in most cases (77% cases (95% CI [49; 92]), which consistent with international data on its highest prevalence in the monogenic forms of PP. In 23% of cases (95% CI [7; 50]) nucleotide variants were identified in other candidate genes associated with neuroontogenesis and neuroendocrine regulation mechanisms of hypothalamic-pituitary axis. CONCLUSION Our study confirms that detailed family history data in children with PP provides a rational approach to molecular-genetic testing. Data of inheritance pattern and clinical manifestations will simplify the diagnosis of hereditary forms of disease and enhance genetic counselling of families, followed by timely examination and administration of pathogenetic therapy.
Collapse
|
21
|
Brito VN, Canton APM, Seraphim CE, Abreu AP, Macedo DB, Mendonca BB, Kaiser UB, Argente J, Latronico AC. The Congenital and Acquired Mechanisms Implicated in the Etiology of Central Precocious Puberty. Endocr Rev 2023; 44:193-221. [PMID: 35930274 PMCID: PMC9985412 DOI: 10.1210/endrev/bnac020] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Indexed: 01/20/2023]
Abstract
The etiology of central precocious puberty (CPP) is multiple and heterogeneous, including congenital and acquired causes that can be associated with structural or functional brain alterations. All causes of CPP culminate in the premature pulsatile secretion of hypothalamic GnRH and, consequently, in the premature reactivation of hypothalamic-pituitary-gonadal axis. The activation of excitatory factors or suppression of inhibitory factors during childhood represent the 2 major mechanisms of CPP, revealing a delicate balance of these opposing neuronal pathways. Hypothalamic hamartoma (HH) is the most well-known congenital cause of CPP with central nervous system abnormalities. Several mechanisms by which hamartoma causes CPP have been proposed, including an anatomical connection to the anterior hypothalamus, autonomous neuroendocrine activity in GnRH neurons, trophic factors secreted by HH, and mechanical pressure applied to the hypothalamus. The importance of genetic and/or epigenetic factors in the underlying mechanisms of CPP has grown significantly in the last decade, as demonstrated by the evidence of genetic abnormalities in hypothalamic structural lesions (eg, hamartomas, gliomas), syndromic disorders associated with CPP (Temple, Prader-Willi, Silver-Russell, and Rett syndromes), and isolated CPP from monogenic defects (MKRN3 and DLK1 loss-of-function mutations). Genetic and epigenetic discoveries involving the etiology of CPP have had influence on the diagnosis and familial counseling providing bases for potential prevention of premature sexual development and new treatment targets in the future. Global preventive actions inducing healthy lifestyle habits and less exposure to endocrine-disrupting chemicals during the lifespan are desirable because they are potentially associated with CPP.
Collapse
Affiliation(s)
- Vinicius N Brito
- Discipline of Endocrinology & Metabolism, Department of Internal
Medicine, University of Sao Paulo Medical School, University of Sao
Paulo, Sao Paulo 01246 903, Brazil
| | - Ana P M Canton
- Discipline of Endocrinology & Metabolism, Department of Internal
Medicine, University of Sao Paulo Medical School, University of Sao
Paulo, Sao Paulo 01246 903, Brazil
| | - Carlos Eduardo Seraphim
- Discipline of Endocrinology & Metabolism, Department of Internal
Medicine, University of Sao Paulo Medical School, University of Sao
Paulo, Sao Paulo 01246 903, Brazil
| | - Ana Paula Abreu
- Division of Endocrinology, Diabetes and Hypertension, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School,
Boston, MA 02115, USA
| | - Delanie B Macedo
- Discipline of Endocrinology & Metabolism, Department of Internal
Medicine, University of Sao Paulo Medical School, University of Sao
Paulo, Sao Paulo 01246 903, Brazil
- Division of Endocrinology, Diabetes and Hypertension, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School,
Boston, MA 02115, USA
- Núcleo de Atenção Médica Integrada, Centro de Ciências da Saúde,
Universidade de Fortaleza, Fortaleza 60811 905,
Brazil
| | - Berenice B Mendonca
- Discipline of Endocrinology & Metabolism, Department of Internal
Medicine, University of Sao Paulo Medical School, University of Sao
Paulo, Sao Paulo 01246 903, Brazil
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School,
Boston, MA 02115, USA
| | - Jesús Argente
- Hospital Infantil Universitario Niño Jesús, Department of Endocrinology and
Department of Pediatrics, Universidad Autónoma de Madrid, Spanish PUBERE Registry,
CIBER of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, IMDEA
Institute, Madrid 28009, Spain
| | - Ana Claudia Latronico
- Discipline of Endocrinology & Metabolism, Department of Internal
Medicine, University of Sao Paulo Medical School, University of Sao
Paulo, Sao Paulo 01246 903, Brazil
| |
Collapse
|
22
|
Argente J, Dunkel L, Kaiser UB, Latronico AC, Lomniczi A, Soriano-Guillén L, Tena-Sempere M. Molecular basis of normal and pathological puberty: from basic mechanisms to clinical implications. Lancet Diabetes Endocrinol 2023; 11:203-216. [PMID: 36620967 PMCID: PMC10198266 DOI: 10.1016/s2213-8587(22)00339-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 01/07/2023]
Abstract
Puberty is a major maturational event; its mechanisms and timing are driven by genetic determinants, but also controlled by endogenous and environmental cues. Substantial progress towards elucidation of the neuroendocrine networks governing puberty has taken place. However, key aspects of the mechanisms responsible for the precise timing of puberty and its alterations have only recently begun to be deciphered, propelled by epidemiological data suggesting that pubertal timing is changing in humans, via mechanisms that are not yet understood. By integrating basic and clinical data, we provide a comprehensive overview of current advances on the physiological basis of puberty, with a particular focus on the roles of kisspeptins and other central transmitters, the underlying molecular and endocrine mechanisms, and the pathways involved in pubertal modulation by nutritional and metabolic cues. Additionally, we have summarised molecular features of precocious and delayed puberty in both sexes, as revealed by clinical and genetic studies. This Review is a synoptic up-to-date view of how puberty is controlled and of the pathogenesis of major pubertal alterations, from both a clinical and translational perspective. We also highlight unsolved challenges that will seemingly concentrate future research efforts in this active domain of endocrinology.
Collapse
Affiliation(s)
- Jesús Argente
- Department of Pediatrics & Pediatric Endocrinology, Universidad Autónoma de Madrid, University Hospital Niño Jesús, Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; IMDEA Food Institute, Madrid, Spain.
| | - Leo Dunkel
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London Medical School, London, UK
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana C Latronico
- Developmental Endocrinology Unit, Laboratory of Hormones and Molecular Genetics, LIM42, Department of Endocrinology and Metabolism, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Alejandro Lomniczi
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Leandro Soriano-Guillén
- Service of Pediatrics, University Hospital Fundación Jiménez Díaz, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofia, Córdoba, Spain; Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
23
|
Chen GY, Wang LZ, Cui Y, Liu JC, Wang LQ, Wang LL, Sun JY, Liu C, Tan HL, Li Q, Jin YS, Xu ZC, Yu DJ. Serum metabolomic analysis reveals key metabolites in drug treatment of central precocious puberty in female children. Front Mol Neurosci 2023; 15:972297. [PMID: 36776772 PMCID: PMC9912178 DOI: 10.3389/fnmol.2022.972297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
Precocious puberty (PP) is a common condition among children. According to the pathogenesis and clinical manifestations, PP can be divided into central precocious puberty (CPP, gonadotropin dependent), peripheral precocious puberty (PPP, gonadotropin independent), and incomplete precocious puberty (IPP). Identification of the variations in key metabolites involved in CPP and their underlying biological mechanisms has increased the understanding of the pathological processes of this condition. However, little is known about the role of metabolite variations in the drug treatment of CPP. Moreover, it remains unclear whether the understanding of the crucial metabolites and pathways can help predict disease progression after pharmacological therapy of CPP. In this study, systematic metabolomic analysis was used to examine three groups, namely, healthy control (group N, 30 healthy female children), CPP (group S, 31 female children with CPP), and treatment (group R, 29 female children) groups. A total of 14 pathways (the top two pathways were aminoacyl-tRNA biosynthesis and phenylalanine, tyrosine, and tryptophan biosynthesis) were significantly enriched in children with CPP. In addition, two short peptides (His-Arg-Lys-Glu and Lys-Met-His) were found to play a significant role in CPP. Various metabolites associated with different pathways including amino acids, PE [19:1(9Z)0:0], tumonoic acid I, palmitic amide, and linoleic acid-biotin were investigated in the serum of children in all groups. A total of 45 metabolites were found to interact with a chemical drug [a gonadotropin-releasing hormone (GnRH) analog] and a traditional Chinese medicinal formula (DBYW). This study helps to understand metabolic variations in CPP after drug therapy, and further investigation may help develop individualized treatment approaches for CPP in clinical practice.
Collapse
Affiliation(s)
- Guo-you Chen
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China,College of Pharmacy, Daqing Campus, Harbin Medical University, Daqing, China
| | - Li-zhe Wang
- Heilongjiang Provincial Hospital, Harbin, China
| | - Yue Cui
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China
| | - Jin-cheng Liu
- College of Pharmacy, Daqing Campus, Harbin Medical University, Daqing, China
| | - Li-qiu Wang
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China
| | - Long-long Wang
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China
| | - Jing-yue Sun
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China
| | - Chang Liu
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China
| | - Hai-ling Tan
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China
| | - Qi Li
- College of Pharmacy, Daqing Campus, Harbin Medical University, Daqing, China
| | - Yi-si Jin
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China,Yi-si Jin,
| | - Zhi-chun Xu
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China,Zhi-chun Xu,
| | - De-jun Yu
- The Fifth Affiliated Hospital of Harbin Medical University, Women and Children’s Healthcare Hospital, Daqing, China,*Correspondence: De-jun Yu,
| |
Collapse
|
24
|
Heger S, Reschke F. Genetische und epigenetische Einflüsse auf den Pubertätsverlauf in Bezug auf Pubertas praecox vera und Pubertas tarda. GYNAKOLOGISCHE ENDOKRINOLOGIE 2023. [DOI: 10.1007/s10304-022-00497-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
25
|
Suzuki E, Miyado M, Kuroki Y, Fukami M. Genetic variants of G-protein coupled receptors associated with pubertal disorders. Reprod Med Biol 2023; 22:e12515. [PMID: 37122876 PMCID: PMC10134480 DOI: 10.1002/rmb2.12515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/02/2023] [Accepted: 04/11/2023] [Indexed: 05/02/2023] Open
Abstract
Background The human hypothalamic-pituitary-gonadal (HPG) axis is the regulatory center for pubertal development. This axis involves six G-protein coupled receptors (GPCRs) encoded by KISS1R, TACR3, PROKR2, GNRHR, LHCGR, and FSHR. Methods Previous studies have identified several rare variants of the six GPCR genes in patients with pubertal disorders. In vitro assays and animal studies have provided information on the function of wild-type and variant GPCRs. Main Findings Of the six GPCRs, those encoded by KISS1R and TACR3 are likely to reside at the top of the HPG axis. Several loss-of-function variants in the six genes were shown to cause late/absent puberty. In particular, variants in KISS1R, TACR3, PROKR2, and GNRHR lead to hypogonadotropic hypogonadism in autosomal dominant, recessive, and oligogenic manners. Furthermore, a few gain-of-function variants of KISS1R, PROKR2, and LHCGR have been implicated in precocious puberty. The human HPG axis may contain additional GPCRs. Conclusion The six GPCRs in the HPG axis govern pubertal development through fine-tuning of hormone secretion. Rare sequence variants in these genes jointly account for a certain percentage of genetic causes of pubertal disorders. Still, much remains to be clarified about the molecular network involving the six GPCRs.
Collapse
Affiliation(s)
- Erina Suzuki
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Mami Miyado
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of Food and NutritionBeppu UniversityOitaJapan
| | - Yoko Kuroki
- Department of Genome Medicine, National Center for Child Health and DevelopmentTokyoJapan
- Division of Collaborative Research, National Center for Child Health and DevelopmentTokyoJapan
- Division of Diversity ResearchNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Maki Fukami
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Division of Diversity ResearchNational Research Institute for Child Health and DevelopmentTokyoJapan
| |
Collapse
|
26
|
Palumbo S, Cirillo G, Sanchez G, Aiello F, Fachin A, Baldo F, Pellegrin MC, Cassio A, Salerno M, Maghnie M, Faienza MF, Wasniewska M, Fintini D, Giacomozzi C, Ciccone S, Miraglia Del Giudice E, Tornese G, Grandone A. A new DLK1 defect in a family with idiopathic central precocious puberty: elucidation of the male phenotype. J Endocrinol Invest 2022; 46:1233-1240. [PMID: 36577869 DOI: 10.1007/s40618-022-01997-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE We aimed to investigate a cohort of female and male patients with idiopathic central precocious puberty (CPP), negative for Makorin Ring Finger Protein 3 (MKRN3) defect, by molecular screening for Delta-like 1 homolog (DLK1) defects. DLK1 is an imprinted gene, whose mutations have been described as a rare cause of CPP in girls and adult women with precocious menarche, obesity and metabolic derangement. METHODS We enrolled 14 girls with familial CPP and 13 boys with familial or sporadic CPP from multiple academic hospital centers. Gene sequencing of DLK1 gene was performed. Circulating levels of DLK1 were measured and clinical and biochemical characteristics were described in those with DLK1 defects. RESULTS A novel heterozygous mutation in DLK1, c.288_289insC (p.Cys97Leufs*16), was identified in a male proband, his sister and their father. Age at onset of puberty was in line with previous reports in the girl and 8 years in the boy. The father with untreated CPP showed short stature. No metabolic derangement was present in the father except hypercholesterolemia. Undetectable Dlk1 serum levels indicated the complete lack of protein production in the three affected patients. CONCLUSION A DLK1 defect has been identified for the first time in a boy, underscoring the importance of genetic testing in males with idiopathic or sporadic CPP. The short stature reported by his untreated father suggests the need for timely diagnosis and treatment of subjects with DLK1 defects.
Collapse
Affiliation(s)
- S Palumbo
- Department of Child, Women, General and Specialized Surgery, University of Campania, "L. Vanvitelli", Vico L. De Crecchio n° 2, 80138, Naples, Italy
| | - G Cirillo
- Department of Child, Women, General and Specialized Surgery, University of Campania, "L. Vanvitelli", Vico L. De Crecchio n° 2, 80138, Naples, Italy
| | - G Sanchez
- Department of Child, Women, General and Specialized Surgery, University of Campania, "L. Vanvitelli", Vico L. De Crecchio n° 2, 80138, Naples, Italy
| | - F Aiello
- Department of Child, Women, General and Specialized Surgery, University of Campania, "L. Vanvitelli", Vico L. De Crecchio n° 2, 80138, Naples, Italy
| | - A Fachin
- University of Trieste, Trieste, Italy
| | - F Baldo
- University of Trieste, Trieste, Italy
| | - M C Pellegrin
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - A Cassio
- Pediatric Endocrine Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - M Salerno
- Pediatric Endocrine Unit, Department of Translational Medical Sciences, University Federico II, Naples, Italy
| | - M Maghnie
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genoa, Italy
| | - M F Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari A. Moro, Bari, Italy
| | - M Wasniewska
- Unit of Paediatrics, Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - D Fintini
- Endocrinology Unit, University-Hospital Pediatric Department, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - C Giacomozzi
- Unit of Pediatrics, Department of Maternal and Child Health, Carlo Poma Hospital, ASST-Mantova, Mantua, Italy
| | - S Ciccone
- Pediatric Unit-"M. Bufalini" Hospital - Cesena, Cesena, Italy
| | - E Miraglia Del Giudice
- Department of Child, Women, General and Specialized Surgery, University of Campania, "L. Vanvitelli", Vico L. De Crecchio n° 2, 80138, Naples, Italy
| | - G Tornese
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - A Grandone
- Department of Child, Women, General and Specialized Surgery, University of Campania, "L. Vanvitelli", Vico L. De Crecchio n° 2, 80138, Naples, Italy.
| |
Collapse
|
27
|
Gui Z, Lv M, Han M, Li S, Mo Z. Effect of CPP-related genes on GnRH secretion and Notch signaling pathway during puberty. Biomed J 2022; 46:100575. [PMID: 36528337 DOI: 10.1016/j.bj.2022.12.003] [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: 08/25/2021] [Revised: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Puberty is a complex biological process of sexual development, influenced by genetic, metabolic-nutritional, environmental and socioeconomic factors, characterized by the development of secondary sexual characteristics, maturation of the gonads, leading to the acquisition of reproductive capacity. The onset of central precocious puberty (CPP) is mainly associated with the early activation of the hypothalamic-pituitary-gonadal (HPG) axis and increased secretion of gonadotropin-releasing hormone (GnRH), leading to increased pituitary secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) and activation of gonadal function. Due to the expense and invasiveness of current diagnostic testing and drug therapies for CPP, it would be helpful to find serum and genetic markers to facilitate diagnosis. In this paper, we summarized the related factors that may affect the expression of GnRH1 gene and the secretion and action pathway of GnRH and related sex hormones, and found several potential targets, such as MKRN3, DLK1 and KISS1. Although, the specific mechanism still needs to be further studied, we would be encouraged if the insights from this review could provide new insights for future research and clinical diagnosis and treatment of CPP.
Collapse
Affiliation(s)
- Zihao Gui
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China; Clinical Medicine of Hengyang Medical School, University of South China, Hengyang, China
| | - Mei Lv
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China; Anshun City People's Hospital, Anshun, Guizhou, China
| | - Min Han
- Clinical Medicine of Hengyang Medical School, University of South China, Hengyang, China
| | - Shan Li
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China
| | - Zhongcheng Mo
- Guangxi Provincial Postgraduate Co-training Base for Collaborative Innovation in Basic Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi, China; Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.
| |
Collapse
|
28
|
Shi X, Zhuang Y, Chen Z, Xu M, Kuang J, Sun XL, Gao L, Kuang X, Zhang H, Li W, Wong SZH, Liu C, Liu L, Jiang D, Pei D, Lin Y, Wu QF. Hierarchical deployment of Tbx3 dictates the identity of hypothalamic KNDy neurons to control puberty onset. SCIENCE ADVANCES 2022; 8:eabq2987. [PMID: 36383654 PMCID: PMC9668310 DOI: 10.1126/sciadv.abq2987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/23/2022] [Indexed: 05/17/2023]
Abstract
The neuroendocrine system consists of a heterogeneous collection of neuropeptidergic neurons in the brain, among which hypothalamic KNDy neurons represent an indispensable cell subtype controlling puberty onset. Although neural progenitors and neuronal precursors along the cell lineage hierarchy adopt a cascade diversification strategy to generate hypothalamic neuronal heterogeneity, the cellular logic operating within the lineage to specify a subtype of neuroendocrine neurons remains unclear. As human genetic studies have recently established a link between TBX3 mutations and delayed puberty onset, we systematically studied Tbx3-derived neuronal lineage and Tbx3-dependent neuronal specification and found that Tbx3 hierarchically established and maintained the identity of KNDy neurons for triggering puberty. Apart from the well-established lineage-dependent fate determination, we uncovered rules of interlineage interaction and intralineage retention operating through neuronal differentiation in the absence of Tbx3. Moreover, we revealed that human TBX3 mutations disturbed the phase separation of encoded proteins and impaired transcriptional regulation of key neuropeptides, providing a pathological mechanism underlying TBX3-associated puberty disorders.
Collapse
Affiliation(s)
- Xiang Shi
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yanrong Zhuang
- IDG/McGovern Institute for Brain Research, Tsinghua–Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Mingrui Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Junqi Kuang
- University of Chinese Academy of Sciences, Beijing 100101, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xue-Lian Sun
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Lisen Gao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xia Kuang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huairen Zhang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Samuel Zheng Hao Wong
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chuanyu Liu
- BGI-ShenZhen, Shenzhen 518103, China
- Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Longqi Liu
- BGI-ShenZhen, Shenzhen 518103, China
- Shenzhen Bay Laboratory, Shenzhen 518000, China
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Danhua Jiang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Yi Lin
- IDG/McGovern Institute for Brain Research, Tsinghua–Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Corresponding author. (Q.-F.W.); (Y.L.)
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China
- Chinese Institute for Brain Research, Beijing 102206, China
- Beijing Children’s Hospital, Capital Medical University, Beijing 100045, China
- Corresponding author. (Q.-F.W.); (Y.L.)
| |
Collapse
|
29
|
Roberts SA, Naulé L, Chouman S, Johnson T, Johnson M, Carroll RS, Navarro VM, Kaiser UB. Hypothalamic Overexpression of Makorin Ring Finger Protein 3 Results in Delayed Puberty in Female Mice. Endocrinology 2022; 163:bqac132. [PMID: 35974456 PMCID: PMC10233297 DOI: 10.1210/endocr/bqac132] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Indexed: 11/19/2022]
Abstract
Makorin ring finger protein 3 (MKRN3) is an important neuroendocrine player in the control of pubertal timing and upstream inhibitor of gonadotropin-releasing hormone secretion. In mice, expression of Mkrn3 in the hypothalamic arcuate and anteroventral periventricular nucleus is high early in life and declines before the onset of puberty. Therefore, we aimed to explore if the persistence of hypothalamic Mkrn3 expression peripubertally would result in delayed puberty. Female mice that received neonatal bilateral intracerebroventricular injections of a recombinant adeno-associated virus expressing Mkrn3 had delayed vaginal opening and first estrus compared with animals injected with control virus. Subsequent estrous cycles and fertility were normal. Interestingly, male mice treated similarly did not exhibit delayed puberty onset. Kiss1, Tac2, and Pdyn mRNA levels were increased in the mediobasal hypothalamus in females at postnatal day 28, whereas kisspeptin and neurokinin B protein levels in the arcuate nucleus were decreased, following Mkrn3 overexpression, compared to controls. Cumulatively, these data suggest that Mkrn3 may directly or indirectly target neuropeptides of Kiss1 neurons to degradation pathways. This mouse model suggests that MKRN3 may be a potential contributor to delayed onset of puberty, in addition to its well-established roles in central precocious puberty and the timing of menarche.
Collapse
Affiliation(s)
- Stephanie A Roberts
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Lydie Naulé
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Soukayna Chouman
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Tatyana Johnson
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Marciana Johnson
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Victor M Navarro
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
30
|
Saengkaew T, Howard SR. Genetics of pubertal delay. Clin Endocrinol (Oxf) 2022; 97:473-482. [PMID: 34617615 PMCID: PMC9543006 DOI: 10.1111/cen.14606] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
The timing of pubertal development is strongly influenced by the genetic background, and clinical presentations of delayed puberty are often found within families with clear patterns of inheritance. The discovery of the underlying genetic regulators of such conditions, in recent years through next generation sequencing, has advanced the understanding of the pathogenesis of disorders of pubertal timing and the potential for genetic testing to assist diagnosis for patients with these conditions. This review covers the significant advances in the understanding of the biological mechanisms of delayed puberty that have occurred in the last two decades.
Collapse
Affiliation(s)
- Tansit Saengkaew
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Endocrinology Unit, Department of Paediatrics, Faculty of MedicinePrince of Songkla UniversitySongkhlaThailand
| | - Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
31
|
The Role of Genetics in Central Precocious Puberty: Confirmed and Potential Neuroendocrine Genetic and Epigenetic Contributors and Their Interactions with Endocrine Disrupting Chemicals (EDCs). ENDOCRINES 2022. [DOI: 10.3390/endocrines3030035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Despite the growing prevalence of central precocious puberty (CPP), most cases are still diagnosed as “idiopathic” due to the lack of identifiable findings of other diagnostic etiology. We are gaining greater insight into some key genes affecting neurotransmitters and receptors and how they stimulate or inhibit gonadotropin-releasing hormone (GnRH) secretion, as well as transcriptional and epigenetic influences. Although the genetic contributions to pubertal regulation are more established in the hypogonadotropic hypogonadism (HH) literature, cases of CPP have provided the opportunity to learn more about its own genetic influences. There have been clinically confirmed cases of CPP associated with gene mutations in kisspeptin and its receptor (KISS1, KISS1R), Delta-like noncanonical Notch ligand 1 (DLK1), and the now most commonly identified genetic cause of CPP, makorin ring finger protein (MKRN3). In addition to these proven genetic causes, a number of other candidates continue to be evaluated. After reviewing the basic clinical aspects of puberty, we summarize what is known about the various genetic and epigenetic causes of CPP as well as discuss some of the potential effects of endocrine disrupting chemicals (EDCs) on some of these processes.
Collapse
|
32
|
Yu T, Yu Y, Li X, Xue P, Yu X, Chen Y, Kong H, Lin C, Wang X, Mei H, Wang D, Liu S. Effects of childhood obesity and related genetic factors on precocious puberty: protocol for a multi-center prospective cohort study. BMC Pediatr 2022; 22:310. [PMID: 35624438 PMCID: PMC9135982 DOI: 10.1186/s12887-022-03350-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 05/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Childhood obesity has important effects on the onset and development of puberty. Although a number of studies have confirmed the relationship between obesity and precocious puberty, little is known about the pleiotropic genes of obesity and precocious puberty and the interaction between genes and environment. There are four objectives: (1) to analyze the incidence of precocious puberty in the general population in China; (2) to verify the direct effect of obesity on children's precocious puberty using a variety of methods; (3) to verify the effect of obesity and its risk gene polymorphism on precocious puberty in a prospective cohort study; and (4) to analyze the interaction effect of genes and environment on pubertal development. METHODS We will conduct a multi-center prospective cohort study in three cities, which are selected in southern, central, and northern China, respectively. Primary schools in these cities will be selected by a stratified cluster random sampling method. Primary school students from grade 1 to grade 3 (6 to 10 years old) will be selected for the cohort with extensive baseline data collection, including assessment of pubertal development, family demographic information, early development, sleep pattern, dietary pattern, and physical activity. Participants will be followed up for at least three years, and long-term follow-up will depend on future funding. DISCUSSION The findings of this multicenter prospective population-based cohort study may expand previous related puberty development research as well as provide important information on the mechanism of early puberty. Targeted interventions can also be developed to improve adolescent health problems related to puberty development based on the available evidence. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04113070 , prospectively registered on October 2, 2019.
Collapse
Affiliation(s)
- Tingting Yu
- Sanya Women and Children's Hospital, managed by Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 339 Yingbin Road, Sanya, 572022, China.,Office of Hospital Infection Management, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ying Yu
- Sanya Women and Children's Hospital, managed by Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 339 Yingbin Road, Sanya, 572022, China
| | - Xiaoqing Li
- Sanya Women and Children's Hospital, managed by Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 339 Yingbin Road, Sanya, 572022, China.,School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Xue
- Sanya Women and Children's Hospital, managed by Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 339 Yingbin Road, Sanya, 572022, China.,School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodan Yu
- Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Chen
- Department of Endocrinology and Genetic Metabolism, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huijun Kong
- Department of Pediatrics, Qu Fu People's Hospital, Qufu, Shandong, China
| | - Cuilan Lin
- Boai Hospital of Zhongshan, Southern Medical University, Zhongshan, Guangdong, China
| | - Xiumin Wang
- Department of Endocrinology and Genetic Metabolism, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Mei
- Department of Data Science, School of Population Health, University of Mississippi Medical Center, Jackson, MS, USA
| | - Dan Wang
- Sanya Women and Children's Hospital, managed by Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 339 Yingbin Road, Sanya, 572022, China.
| | - Shijian Liu
- Sanya Women and Children's Hospital, managed by Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 339 Yingbin Road, Sanya, 572022, China. .,School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
33
|
Hu KL, Chen Z, Li X, Cai E, Yang H, Chen Y, Wang C, Ju L, Deng W, Mu L. Advances in clinical applications of kisspeptin-GnRH pathway in female reproduction. Reprod Biol Endocrinol 2022; 20:81. [PMID: 35606759 PMCID: PMC9125910 DOI: 10.1186/s12958-022-00953-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/30/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Kisspeptin is the leading upstream regulator of pulsatile and surge Gonadotrophin-Releasing Hormone secretion (GnRH) in the hypothalamus, which acts as the key governor of the hypothalamic-pituitary-ovary axis. MAIN TEXT Exogenous kisspeptin or its receptor agonist can stimulate GnRH release and subsequent physiological gonadotropin secretion in humans. Based on the role of kisspeptin in the hypothalamus, a broad application of kisspeptin and its receptor agonist has been recently uncovered in humans, including central control of ovulation, oocyte maturation (particularly in women at a high risk of ovarian hyperstimulation syndrome), test for GnRH neuronal function, and gatekeepers of puberty onset. In addition, the kisspeptin analogs, such as TAK-448, showed promising agonistic activity in healthy women as well as in women with hypothalamic amenorrhoea or polycystic ovary syndrome. CONCLUSION More clinical trials should focus on the therapeutic effect of kisspeptin, its receptor agonist and antagonist in women with reproductive disorders, such as hypothalamic amenorrhoea, polycystic ovary syndrome, and endometriosis.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Center for Reproductive Medicine, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, People's Republic of China, 100191
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Zimiao Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Xiaoxue Li
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Enci Cai
- Department of Nutrition and Food Science, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, Twin Cities, Minneapolis, MN, 55455, USA
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Yi Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Congying Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Liping Ju
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Wenhai Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China, 325006.
| | - Liangshan Mu
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200.
| |
Collapse
|
34
|
Lin Y, He Y, Sun W, Wang Y, Yu J. Recent advances on the relationship between the DLK1 system and central precocious puberty. Biol Reprod 2022; 107:679-683. [PMID: 35594453 DOI: 10.1093/biolre/ioac106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/14/2022] Open
Abstract
Precocious puberty, as a common pediatric endocrine disease, can be divided into central precocious puberty (CPP) and peripheral precocious puberty (PPP), even though most cases of precocious puberty are diagnosed as CPP. According to its etiology, CPP can be further divided into organic and idiopathic CPP. However, the mechanisms of idiopathic CPP have not yet been fully elucidated. Currently, four genes, including the kisspeptin gene (KISS1), the kisspeptin receptor gene (KISS1R), the makorin ring finger protein 3 (MKRN3), and the Delta-like non-canonical Notch ligand 1 (DLK1), have been implicated in CPP cases, of which DLK1 has been determined to represent a key, recently found CPP-related gene. In this review, we will not only highlight the latest discoveries on the relationship between the DLK1 system and CPP but also explore the involvement of the system as well as the Notch signaling pathway in CPP occurrence.
Collapse
Affiliation(s)
- Yating Lin
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yuanyuan He
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Wen Sun
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Yonghong Wang
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Jian Yu
- Department of Traditional Chinese Medicine, Children's Hospital of Fudan University, Shanghai, 201102, China
| |
Collapse
|
35
|
Abstract
Pubertal onset is known to result from reactivation of the hypothalamic-pituitary-gonadal (HPG) axis, which is controlled by complex interactions of genetic and nongenetic factors. Most cases of precocious puberty (PP) are diagnosed as central PP (CPP), defined as premature activation of the HPG axis. The cause of CPP in most girls is not identifiable and, thus, referred to as idiopathic CPP (ICPP), whereas boys are more likely to have an organic lesion in the brain. ICPP has a genetic background, as supported by studies showing that maternal age at menarche is associated with pubertal timing in their offspring. A gain of expression in the kisspeptin gene (KISS1), gain-of-function mutation in the kisspeptin receptor gene (KISS1R), loss-of-function mutation in makorin ring finger protein 3 (MKRN3), and loss-of-function mutations in the delta-like homolog 1 gene (DLK1) have been associated with ICPP. Other genes, such as gamma-aminobutyric acid receptor subunit alpha-1 (GABRA1), lin-28 homolog B (LIN28B), neuropeptide Y (NPYR), tachykinin 3 (TAC3), and tachykinin receptor 3 (TACR3), have been implicated in the progression of ICPP, although their relationships require elucidation. Environmental and socioeconomic factors may also be correlated with ICPP. In the progression of CPP, epigenetic factors such as DNA methylation, histone posttranslational modifications, and noncoding ribonucleic acids may mediate the relationship between genetic and environmental factors. CPP is correlated with short- and long-term adverse health outcomes, which forms the rationale for research focusing on understanding its genetic and nongenetic factors.
Collapse
Affiliation(s)
- Young Suk Shim
- Department of Pediatrics, Ajou University Hospital, Ajou University School of Medicine, Suwon, Korea
| | - Hae Sang Lee
- Department of Pediatrics, Ajou University Hospital, Ajou University School of Medicine, Suwon, Korea
| | - Jin Soon Hwang
- Department of Pediatrics, Ajou University Hospital, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
36
|
Aberrant Notch Signaling Pathway as a Potential Mechanism of Central Precocious Puberty. Int J Mol Sci 2022; 23:ijms23063332. [PMID: 35328752 PMCID: PMC8950842 DOI: 10.3390/ijms23063332] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023] Open
Abstract
The Notch signaling pathway is highly conserved during evolution. It has been well documented that Notch signaling regulates cell proliferation, migration, and death in the nervous, cardiac, and endocrine systems. The Notch pathway is relatively simple, but its activity is regulated by numerous complex mechanisms. Ligands bind to Notch receptors, inducing their activation and cleavage. Various post-translational processes regulate Notch signaling by affecting the synthesis, secretion, activation, and degradation of Notch pathway-related proteins. Through such post-translational regulatory processes, Notch signaling has versatile effects in many tissues, including the hypothalamus. Recently, several studies have reported that mutations in genes related to the Notch signaling pathway were found in patients with central precocious puberty (CPP). CPP is characterized by the early activation of the hypothalamus–pituitary–gonadal (HPG) axis. Although genetic factors play an important role in CPP development, few associated genetic variants have been identified. Aberrant Notch signaling may be associated with abnormal pubertal development. In this review, we discuss the current knowledge about the role of the Notch signaling pathway in puberty and consider the potential mechanisms underlying CPP.
Collapse
|
37
|
Hudson AD, Kauffman AS. Metabolic actions of kisspeptin signaling: Effects on body weight, energy expenditure, and feeding. Pharmacol Ther 2022; 231:107974. [PMID: 34530008 PMCID: PMC8884343 DOI: 10.1016/j.pharmthera.2021.107974] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022]
Abstract
Kisspeptin (encoded by the Kiss1 gene) and its receptor, KISS1R (encoded by the Kiss1r gene), have well-established roles in stimulating reproduction via central actions on reproductive neural circuits, but recent evidence suggests that kisspeptin signaling also influences metabolism and energy balance. Indeed, both Kiss1 and Kiss1r are expressed in many metabolically-relevant peripheral tissues, including both white and brown adipose tissue, the liver, and the pancreas, suggesting possible actions on these tissues or involvement in their physiology. In addition, there may be central actions of kisspeptin signaling, or factors co-released from kisspeptin neurons, that modulate metabolic, feeding, or thermoregulatory processes. Accumulating data from animal models suggests that kisspeptin signaling regulates a wide variety of metabolic parameters, including body weight and energy expenditure, adiposity and adipose tissue function, food intake, glucose metabolism, respiratory rates, locomotor activity, and thermoregulation. Herein, the current evidence for the involvement of kisspeptin signaling in each of these physiological parameters is reviewed, gaps in knowledge identified, and future avenues of important research highlighted. Collectively, the discussed findings highlight emerging non-reproductive actions of kisspeptin signaling in metabolism and energy balance, in addition to previously documented roles in reproductive control, but also emphasize the need for more research to resolve current controversies and uncover underlying molecular and physiological mechanisms.
Collapse
Affiliation(s)
- Alexandra D Hudson
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Alexander S Kauffman
- Dept. of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| |
Collapse
|
38
|
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: 3.0] [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
|
39
|
Soriano-Guillén L, Tena-Sempere M, Seraphim CE, Latronico AC, Argente J. Precocious sexual maturation: Unravelling the mechanisms of pubertal onset through clinical observations. J Neuroendocrinol 2022; 34:e12979. [PMID: 33904190 DOI: 10.1111/jne.12979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 01/05/2023]
Abstract
Puberty is a crucial biological process normally occurring at a specific time during the lifespan, during which sexual and somatic maturation are completed, and reproductive capacity is reached. Pubertal timing is not only determined by genetics, but also by endogenous and environmental cues, including nutritional and metabolic signals. During the last decade, we have learned much regarding the essential roles of kisspeptins and the neuropeptide pathways that converge on these neurones to modulate kisspeptin signalling, as well as neurokinin B and dynorphin, the co-transmitters of Kiss1 neurones in the arcuate nucleus, and the effects of melanocortins on puberty. Indeed, melanocortins are involved in transmitting the regulatory actions of metabolic cues on pubertal maturation. Intracellular metabolic sensors, such as the AMP-activated protein kinase and the fuel-sensing deacetylase SIRT1, have been shown to contribute to puberty. Further understanding of these signals and regulatory circuits will help uncover the intimacies of the central control of puberty, as well as how alterations in metabolic status, ranging from undernutrition to obesity, affect the pubertal process. Precocious puberty is rare and has a clear female predominance. Central precocious puberty (CPP) is diagnosed when premature activation of the hypothalamic-pituitary axis occurs. Its causes are heterogeneous, with alterations of the central nervous system being of special interest, and with environmental factors also playing a role in some cases. During the last decade, several mutations in different genes (including KISS1, KISS1R, MKRN3 and DLK1) that cause CPP have been discovered. Loss-of-function mutations in MKRN3 are the most common monogenic cause of CPP known to date. Here, we review and update what is known regarding the genotype-phenotype relationship in patients with CPP.
Collapse
Affiliation(s)
- Leandro Soriano-Guillén
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Pediatrics, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
- Instituto de Investigación Fundación Jiménez Díaz, Madrid, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Córdoba, Spain
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Carlos E Seraphim
- Laboratory of Hormones and Molecular Genetics, LIM42, Developmental Endocrinology Unit, Department of Internal Medicine, Discipline Endocrinology and Metabolism, Faculty of Medicine, Clinicas Hospital, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana C Latronico
- Laboratory of Hormones and Molecular Genetics, LIM42, Developmental Endocrinology Unit, Department of Internal Medicine, Discipline Endocrinology and Metabolism, Faculty of Medicine, Clinicas Hospital, University of Sao Paulo, Sao Paulo, Brazil
| | - Jesús Argente
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEIUAM+CSIC, Madrid, Spain
| |
Collapse
|
40
|
Calcaterra V, Rossi V, Massini G, Regalbuto C, Hruby C, Panelli S, Bandi C, Zuccotti G. Precocious puberty and microbiota: The role of the sex hormone-gut microbiome axis. Front Endocrinol (Lausanne) 2022; 13:1000919. [PMID: 36339428 PMCID: PMC9634744 DOI: 10.3389/fendo.2022.1000919] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
Puberty is a critical phase of life associated with physiological changes related to sexual maturation, and represents a complex process regulated by multiple endocrine and genetic controls. Puberty is driven by hormones, and it can impact the gut microbiome (GM). GM differences between sex emerge at puberty onset, confirming a relationship between microbiota and sex hormones. In this narrative review, we present an overview of precocious pubertal development and the changes in the GM in precocious puberty (PP) in order to consider the role of the sex hormone-gut microbiome axis from the perspective of pediatric endocrinology. Bidirectional interactions between the GM and sex hormones have been proposed in different studies. Although the evidence on the interaction between microbiota and sex hormones remains limited in pediatric patients, the evidence that GM alterations may occur in girls with central precocious puberty (CPP) represents an interesting finding for the prediction and prevention of PP. Deepening the understanding of the connection between the sex hormones and the role of microbiota changes can lead to the implementation of microbiota-targeted therapies in pubertal disorders by offering a pediatric endocrinology perspective.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, Milan, Italy
- Department of Internal Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Valeria Calcaterra,
| | - Virginia Rossi
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, Milan, Italy
| | - Giulia Massini
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, Milan, Italy
| | - Corrado Regalbuto
- Pediatric unit , Fondazione Istituto di Ricovero e Cura a Carattere (IRCCS) Policlinico S. Matteo and University of Pavia, Pavia, Italy
| | - Chiara Hruby
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, Milan, Italy
| | - Simona Panelli
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
| | - Claudio Bandi
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Department, “Vittore Buzzi” Children’s Hospital, Milan, Italy
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
| |
Collapse
|
41
|
Fu D, Li T, Zhang Y, Wang H, Wu X, Chen Y, Cao B, Wei H. Analysis of the Incidence and Risk Factors of Precocious Puberty in Girls during the COVID-19 Pandemic. Int J Endocrinol 2022; 2022:9229153. [PMID: 36213197 PMCID: PMC9534639 DOI: 10.1155/2022/9229153] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 08/05/2022] [Accepted: 09/08/2022] [Indexed: 11/26/2022] Open
Abstract
Home quarantine due to the global coronavirus disease 2019 (COVID-19) pandemic has had a significant impact on children. Lifestyle changes have led to an increase in precocious puberty (PP) among girls, and the underlying risk factors for this remain unclear. Thus, we aimed to assess the influence of environmental, genetic, nutritional, and other lifestyle factors on the risk of PP in girls. We evaluated the incidence of new-onset PP in girls during home quarantine for COVID-19 and analyzed the potential risk factors. This was a retrospective questionnaire and medical record-based study involving 22 representative medical units from 13 cities in Henan Province, China. Girls with new-onset PP (central precocious puberty, 58; premature thelarche, 58; age, 5-9 years) between February 2020 and May 2020 were included, along with 124 healthy, age-matched controls. The number of new-onset PP cases reported during the study period was compared with that reported between February and May in 2018 and 2019. Patients' families completed a questionnaire to assess potential risk factors. There was a 5.01- and 3.14-fold increase in the number of new-onset PP cases from 2018 to 2020 and from 2019 to 2020, respectively; the differences were statistically significant (p < 0.01). High-risk factors for PP included longer time spent using electronic devices, decreased exercise time, higher body mass index, vitamin D deficiency, young age (<12 years) of mother during menarche, consumption of fried food and processed meat, residence in rural areas, and consumption of off-season fruits. Thus, we found that lifestyle changes caused due to the COVID-19 pandemic led to a significant increase in PP in girls. Management of the risk factors identified in this study may help in PP prevention.
Collapse
Affiliation(s)
- Dongxia Fu
- Department of Endocrinology and Inborn Error of Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450000, China
| | - Tao Li
- Department of Endocrinology and Inborn Error of Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450000, China
| | - Yingxian Zhang
- Department of Endocrinology and Inborn Error of Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450000, China
| | - Huizhen Wang
- Department of Endocrinology and Inborn Error of Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450000, China
| | - Xue Wu
- Department of Endocrinology and Inborn Error of Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450000, China
| | - Yongxing Chen
- Department of Endocrinology and Inborn Error of Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450000, China
| | - Bingyan Cao
- Department of Endocrinology and Inborn Error of Metabolism, Beijing Children's Hospital Affiliated to Capital Medical University, Beijing 100000, China
| | - Haiyan Wei
- Department of Endocrinology and Inborn Error of Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450000, China
| |
Collapse
|
42
|
Abstract
Puberty marks the end of childhood and is a period when individuals undergo physiological and psychological changes to achieve sexual maturation and fertility. The onset of puberty is first detected as an increase in pulsatile secretion of gonadotropin-releasing hormone (GnRH). Pubertal onset is regulated by genetic, nutritional, environmental, and socio-economic factors. Disturbances affecting pubertal timing result in adverse health conditions later in life. Human genetic studies show that around 50-80% of the variation in pubertal onset is genetically determined. The genetic control of pubertal timing has been a field of active investigation in attempt to better understand the neuroendocrine control of this relevant period of life. Large populational studies and patient cohort-based studies have provided insights into the genetic regulation of pubertal onset. In this review, we discuss these discoveries and discuss potential mechanisms for how implicated genes may affect pubertal timing.
Collapse
Affiliation(s)
- Alessandra Mancini
- Department of Medicine, Harvard Medical School, Division of Endocrinology Diabetes and Hypertension, Brigham and Women's Hospital, Boston, USA.
| | - John C Magnotto
- Department of Medicine, Harvard Medical School, Division of Endocrinology Diabetes and Hypertension, Brigham and Women's Hospital, Boston, USA.
| | - Ana Paula Abreu
- Department of Medicine, Harvard Medical School, Division of Endocrinology Diabetes and Hypertension, Brigham and Women's Hospital, Boston, USA.
| |
Collapse
|
43
|
Xie Q, Kang Y, Zhang C, Xie Y, Wang C, Liu J, Yu C, Zhao H, Huang D. The Role of Kisspeptin in the Control of the Hypothalamic-Pituitary-Gonadal Axis and Reproduction. Front Endocrinol (Lausanne) 2022; 13:925206. [PMID: 35837314 PMCID: PMC9273750 DOI: 10.3389/fendo.2022.925206] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/30/2022] [Indexed: 01/07/2023] Open
Abstract
The discovery of kisspeptin as a critical central regulatory factor of GnRH release has given people a novel understanding of the neuroendocrine regulation in human reproduction. Kisspeptin activates the signaling pathway by binding to its receptor kisspeptin receptor (KISS1R) to promote GnRH secretion, thereby regulating the hypothalamic-pituitary-gonadal axis (HPG) axis. Recent studies have shown that kisspeptin neurons located in arcuate nucleus (ARC) co-express neurokinin B (NKB) and dynorphin (Dyn). Such neurons are called KNDy neurons. KNDy neurons participate in the positive and negative feedback of estrogen to GnRH secretion. In addition, kisspeptin is a key factor in the initiation of puberty, and also regulates the processes of female follicle development, oocyte maturation, and ovulation through the HPG axis. In male reproduction, kisspeptin also plays an important role, getting involved in the regulation of Leydig cells, spermatogenesis, sperm functions and reproductive behaviors. Mutations in the KISS1 gene or disorders of the kisspeptin/KISS1R system may lead to clinical symptoms such as idiopathic hypogonadotropic hypogonadism (iHH), central precocious puberty (CPP) and female infertility. Understanding the influence of kisspeptin on the reproductive axis and related mechanisms will help the future application of kisspeptin in disease diagnosis and treatment. In this review, we critically appraise the role of kisspeptin in the HPG axis, including its signaling pathways, negative and positive feedback mechanisms, and its control on female and male reproduction.
Collapse
Affiliation(s)
- Qinying Xie
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Kang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlu Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Xie
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuxiong Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caiqian Yu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Zhao
- Department of Human Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Donghui Huang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Li M, Lan D, Chen Y. Integrated analysis of proteomics and metabolomics in girls with central precocious puberty. Front Endocrinol (Lausanne) 2022; 13:951552. [PMID: 35966072 PMCID: PMC9365929 DOI: 10.3389/fendo.2022.951552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Central precocious puberty (CPP) is a multifactorial and complex condition. Traditional studies focusing on a single indicator cannot always elucidate this panoramic condition but these may be revealed by using omics techniques. OBJECTIVE Proteomics and metabolomics analysis of girls with CPP were compared to normal controls and the potential biomarkers and pathways involved were explored. METHODS Serum proteins and metabolites from normal girls and those with CPP were compared by LC-MS/MS. Multivariate and univariate statistical analysis were used to identify the differentially expressed proteins (DEPs) and differentially expressed metabolites (DEMs). Functional annotation and pathway enrichment analysis were performed by using GO and KEGG databases, and candidate markers were screened. Finally, bioinformatic analysis was used to integrate the results of proteomics and metabolomics to find the key differential proteins, metabolites and potential biomarkers of CPP. RESULTS 134 DEPs were identified in girls with CPP with 71 up- and 63 down-regulated, respectively. Up-regulated proteins were enriched mainly in the extracellular matrix, cell adhesion and cellular protein metabolic processes, platelet degranulation and skeletal system development. The down-regulated proteins were mainly enriched in the immune response. Candidate proteins including MMP9, TIMP1, SPP1, CDC42, POSTN, COL1A1, COL6A1, COL2A1 and BMP1, were found that may be related to pubertal development. 103 DEMs were identified, including 42 up-regulated and 61 down-regulated metabolites which were mainly enriched in lipid and taurine metabolic pathways. KGML network analysis showed that phosphocholine (16:1(9Z)/16:1(9Z)) was involved in arachidonic acid, glycerophospholipid, linoleic acid and α-linolenic acid metabolism and it may be used as a biomarker of CPP. CONCLUSIONS Our study is the first to integrate proteomics and metabolomics to analyze the serum of girls with CPP and we found some key differential proteins and metabolites as well as a potential biomarker for this condition. Lipid metabolism pathways are involved and these may provide a key direction to further explore the molecular mechanisms and pathogenesis of CPP.
Collapse
Affiliation(s)
| | - Dan Lan
- *Correspondence: Dan Lan, ;;
| | | |
Collapse
|
45
|
Fanis P, Morrou M, Tomazou M, Michailidou K, Spyrou GM, Toumba M, Skordis N, Neocleous V, Phylactou LA. Methylation status of hypothalamic Mkrn3 promoter across puberty. Front Endocrinol (Lausanne) 2022; 13:1075341. [PMID: 36714607 PMCID: PMC9880154 DOI: 10.3389/fendo.2022.1075341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/20/2022] [Indexed: 01/15/2023] Open
Abstract
Makorin RING finger protein 3 (MKRN3) is an important factor located on chromosome 15 in the imprinting region associated with Prader-Willi syndrome. Imprinted MKRN3 is expressed in hypothalamic regions essential for the onset of puberty and mutations in the gene have been found in patients with central precocious puberty. The pubertal process is largely controlled by epigenetic mechanisms that include, among other things, DNA methylation at CpG dinucleotides of puberty-related genes. In the present study, we investigated the methylation status of the Mkrn3 promoter in the hypothalamus of the female mouse before, during and after puberty. Initially, we mapped the 32 CpG dinucleotides in the promoter, the 5'UTR and the first 50 nucleotides of the coding region of the Mkrn3 gene. Moreover, we identified a short CpG island region (CpG islet) located within the promoter. Methylation analysis using bisulfite sequencing revealed that CpG dinucleotides were methylated regardless of developmental stage, with the lowest levels of methylation being found within the CpG islet region. In addition, the CpG islet region showed significantly lower methylation levels at the pre-pubertal stage when compared with the pubertal or post-pubertal stage. Finally, in silico analysis of transcription factor binding sites on the Mkrn3 CpG islet identified the recruitment of 29 transcriptional regulators of which 14 were transcriptional repressors. Our findings demonstrate the characterization and differential methylation of the CpG dinucleotides located in the Mkrn3 promoter that could influence the transcriptional activity in pre-pubertal compared to pubertal or post-pubertal period. Further studies are needed to clarify the possible mechanisms and effects of differential methylation of the Mkrn3 promoter.
Collapse
Affiliation(s)
- Pavlos Fanis
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Maria Morrou
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Marios Tomazou
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Kyriaki Michailidou
- Biostatistics Unit, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George M. Spyrou
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Meropi Toumba
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Child Endocrine Care, Department of Pediatrics, Aretaeio Hospital, Nicosia, Cyprus
| | - Nicos Skordis
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Division of Pediatric Endocrinology, Paedi Center for Specialized Pediatrics, Nicosia, Cyprus
- Medical School, University of Nicosia, Nicosia, Cyprus
| | - Vassos Neocleous
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Leonidas A. Phylactou
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- *Correspondence: Leonidas A. Phylactou,
| |
Collapse
|
46
|
Tajima T. Genetic causes of central precocious puberty. Clin Pediatr Endocrinol 2022; 31:101-109. [PMID: 35928377 PMCID: PMC9297165 DOI: 10.1297/cpe.2022-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/07/2022] [Indexed: 11/12/2022] Open
Abstract
Central precocious puberty (CPP) is a condition in which the
hypothalamus–pituitary–gonadal system is activated earlier than the normal developmental
stage. The etiology includes organic lesions in the brain; however, in the case of
idiopathic diseases, environmental and/or genetic factors are involved in the development
of CPP. A genetic abnormality in KISS1R, that encodes the kisspeptin
receptor, was first reported in 2008 as a cause of idiopathic CPP. Furthermore, genetic
alterations in KISS1, MKRN3, DLK1, and
PROKR2 have been reported in idiopathic and/or familial CPP. Of these,
MKRN3 has the highest frequency of pathological variants associated
with CPP worldwide; but, abnormalities in MKRN3 are rare in patients in
East Asia, including Japan. MKRN3 and DLK1 are maternal
imprinting genes; thus, CPP develops when a pathological variant is inherited from the
father. The mechanism of CPP due to defects in MKRN3 and
DLK1 has not been completely clarified, but it is suggested that both
may negatively control the progression of puberty. CPP due to such a single gene
abnormality is extremely rare, but it is important to understand the mechanisms of puberty
and reproduction. A further development in the genetics of CPP is expected in the
future.
Collapse
Affiliation(s)
- Toshihiro Tajima
- Department of Pediatrics, Jichi Medical University Tochigi Children’ Medical Center, Tochigi, Japan
| |
Collapse
|
47
|
Hypothalamic kisspeptin and kisspeptin receptors: Species variation in reproduction and reproductive behaviours. Front Neuroendocrinol 2022; 64:100951. [PMID: 34757093 DOI: 10.1016/j.yfrne.2021.100951] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/22/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
Kisspeptin, encoded by the KISS1 gene, was first discovered as a potential metastasis suppressor gene. The prepro-kisspeptin precursor is cleaved into shorter mature bioactive peptides of varying sizes that bind to the G protein-coupled receptor GPR54 (=KISS1R). Over the last two decades, multiple types of Kiss and KissR genes have been discovered in mammalian and non-mammalian vertebrate species, but they are remarkably absent in birds. Kiss neuronal populations are distributed mainly in the hypothalamus. The KissRs are widely distributed in the brain, including the hypothalamic and non-hypothalamic regions, such as the hippocampus, amygdala, and habenula. The role of KISS1-KISS1R in humans and Kiss1-Kiss1R in rodents is associated with puberty, gonadal maturation, and the reproductive axis. However, recent gene deletion studies in zebrafish and medaka have provided controversial results, suggesting that the reproductive role of kiss is dispensable. This review highlights the evolutionary history, localisation, and significance of Kiss-KissR in reproduction and reproductive behaviours in mammalian and non-mammalian vertebrates.
Collapse
|
48
|
Moise-Silverman J, Silverman LA. A review of the genetics and epigenetics of central precocious puberty. Front Endocrinol (Lausanne) 2022; 13:1029137. [PMID: 36531492 PMCID: PMC9757059 DOI: 10.3389/fendo.2022.1029137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022] Open
Abstract
Gonadotrophin dependent sexual precocity, commonly referred to as central precocious puberty (CPP), results from a premature reactivation of the hypothalamic-pituitary-gonadal (HPG) axis before the normal age of pubertal onset. CPP is historically described as girls who enter puberty before the age of eight, and boys before the age of nine. Females are more likely to be diagnosed with idiopathic CPP; males diagnosed with CPP have a greater likelihood of a defined etiology. These etiologies may include underlying CNS congenital defects, tumors, trauma, or infections as well as environmental, genetic, and epigenetic factors. Recently, genetic variants and mutations which may cause CPP have been identified at both the level of the hypothalamus and the pituitary. Single nucleotide polymorphisms (SNPs), monogenetic mutations, and modifications of the epigenome have been evaluated in relationship to the onset of puberty; these variants are thought to affect the development, structure and function of GnRH neurons which may lead to either precocious, delayed, or absent pubertal reactivation. This review will describe recent advances in the field of the genetic basis of puberty and provide a clinically relevant approach to better understand these varying etiologies of CPP.
Collapse
Affiliation(s)
| | - Lawrence A. Silverman
- Division of Pediatric Endocrinology Goyreb Children’s Hospital – Atlantic Health System, Morristown, NJ, United States
- *Correspondence: Lawrence A. Silverman,
| |
Collapse
|
49
|
Li Y, Tao N, Chen M, Chu J, Huang X, Kong X. Gene Polymorphisms Associated with Central Precocious Puberty and Hormone Levels in Chinese Girls. Int J Endocrinol 2022; 2022:9450663. [PMID: 36046800 PMCID: PMC9420594 DOI: 10.1155/2022/9450663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/04/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Central precocious puberty (CPP) is associated with adverse health outcomes in females; however, CPP pathogenesis remains unclear. In this study, we investigated the association of 20 single nucleotide polymorphisms (SNPs) in eight genes with CPP risk and hormone levels. A case-control study on 247 and 243 girls with and without CPP, respectively, was conducted at Kunming Children's Hospital, China, from September 2019 to August 2020. The genotype of the SNPs and their haplotypes were identified. Additionally, the effects of the polymorphisms on hormone levels were investigated. Three variants (rs10159082, rs7538038, and rs5780218) in KISS1 and two variants (rs7895833 and rs3758391) in SIRT1 were related to an increased CPP risk (odds ratio (OR) = 1.524, 1.507, 1.409, 1.348, and 1.737; 95% confidence interval (CI) = 1.176-1.974, 1.152-1.970, 1.089-1.824, 1.023-1.777, and 1.242-2.430, respectively). Rs3740051in SIRT1 and rs1544410 in VDR reduced CPP risk (OR = 0.689, 0.464; 95% CI, 0.511-0.928, 0.232-0.925, respectively). Rs1544410, rs7975232, and rs731236 in VDR were negatively correlated with peak follicle-stimulating hormone (FSH; β = -2.181; P=0.045), basal FSH (β = -0.391; P=0.010), and insulin-like growth factor (β = -50.360; P=0.041) levels, respectively. KISS1, SIRT1, and VDR variants were associated with CPP susceptibility, and VDR SNPs influenced hormonal levels in Chinese females with CPP. In particular, VDR polymorphism rs1544410 was associated with both CPP risk and GnRH-stimulated peak FSH levels. Further functional research and large-scale genetic studies of these loci and genes are required to confirm our findings.
Collapse
Affiliation(s)
- Yunwei Li
- Faculty of Life Science and Biotechnology, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
- Department of Pharmacy, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Na Tao
- Department of Endocrinology, Genetics and Metabolism of Children, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Minghui Chen
- Medical School, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Jiang Chu
- Medical School, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Xinwei Huang
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xiangyang Kong
- Medical School, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| |
Collapse
|
50
|
Faienza MF, Urbano F, Moscogiuri LA, Chiarito M, De Santis S, Giordano P. Genetic, epigenetic and enviromental influencing factors on the regulation of precocious and delayed puberty. Front Endocrinol (Lausanne) 2022; 13:1019468. [PMID: 36619551 PMCID: PMC9813382 DOI: 10.3389/fendo.2022.1019468] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
The pubertal development onset is controlled by a network of genes that regulate the gonadotropin releasing hormone (GnRH) pulsatile release and the subsequent increase of the circulating levels of pituitary gonadotropins that activate the gonadal function. Although the transition from pre-pubertal condition to puberty occurs physiologically in a delimited age-range, the inception of pubertal development can be anticipated or delayed due to genetic and epigenetic changes or environmental conditions. Most of the genetic and epigenetic alterations concern genes which encode for kisspeptin, GnRH, LH, FSH and their receptor, which represent crucial factors of the hypothalamic-pituitary-gonadal (HPG) axis. Recent data indicate a central role of the epigenome in the regulation of genes in the hypothalamus and pituitary that could mediate the flexibility of pubertal timing. Identification of epigenetically regulated genes, such as Makorin ring finger 3 (MKRN3) and Delta-like 1 homologue (DLK1), respectively responsible for the repression and the activation of pubertal development, provides additional evidence of how epigenetic variations affect pubertal timing. This review aims to investigate genetic, epigenetic, and environmental factors responsible for the regulation of precocious and delayed puberty.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Bari, Italy
- Giovanni XXIII Pediatric Hospital, Bari, Italy
- *Correspondence: Maria Felicia Faienza,
| | | | | | | | - Stefania De Santis
- Department of Pharmacy-Pharmaceutical Science, University of Bari “Aldo Moro”, Bari, Italy
| | - Paola Giordano
- Giovanni XXIII Pediatric Hospital, Bari, Italy
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|