1
|
Jayasena CN, Devine K, Barber K, Comninos AN, Conway GS, Crown A, Davies MC, Ewart A, Seal LJ, Smyth A, Turner HE, Webber L, Anderson RA, Quinton R. Society for endocrinology guideline for understanding, diagnosing and treating female hypogonadism. Clin Endocrinol (Oxf) 2024; 101:409-442. [PMID: 39031660 DOI: 10.1111/cen.15097] [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: 10/31/2023] [Revised: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 07/22/2024]
Abstract
Female hypogonadism (FH) is a relatively common endocrine disorder in women of premenopausal age, but there are significant uncertainties and wide variation in its management. Most current guidelines are monospecialty and only address premature ovarian insufficiency (POI); some allude to management in very brief and general terms, and most rely upon the extrapolation of evidence from the studies relating to physiological estrogen deficiency in postmenopausal women. The Society for Endocrinology commissioned new guidance to provide all care providers with a multidisciplinary perspective on managing patients with all forms of FH. It has been compiled using expertise from Endocrinology, Primary Care, Gynaecology and Reproductive Health practices, with contributions from expert patients and a patient support group, to help clinicians best manage FH resulting from both POI and hypothalamo-pituitary disorders, whether organic or functional.
Collapse
Affiliation(s)
- Channa N Jayasena
- Section of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Kerri Devine
- Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
- Translational & Clinical Research Institute, University of Newcastle-upon-Tyne, Newcastle-upon-Tyne, UK
| | - Katie Barber
- Community Gynaecology (NHS), Principal Medical Limited, Bicester, Oxfordshire, UK
- Oxford Menopause Ltd, Ardington, Wantage, UK
| | - Alexander N Comninos
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Gerard S Conway
- Reproductive Medicine Unit, University College London Hospitals, London, UK
| | - Anna Crown
- Department of Endocrinology, Royal Sussex County Hospital, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Melanie C Davies
- Reproductive Medicine Unit, University College London Hospitals, London, UK
| | - Ann Ewart
- Kallman Syndrome and Congenital Hypogonadotropic Hypogonadism Support Group, Dallas, Texas, United States
| | - Leighton J Seal
- Department of Endocrinology, St George's Hospital Medical School, London, UK
| | - Arlene Smyth
- UK Turner Syndrome Support Society, Clydebank, UK
| | - Helen E Turner
- Department of Endocrinology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Lisa Webber
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Singapore
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Richard Quinton
- Section of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
- Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
- Translational & Clinical Research Institute, University of Newcastle-upon-Tyne, Newcastle-upon-Tyne, UK
| |
Collapse
|
2
|
Read JE, Vasile-Tudorache A, Newsome A, Lorente MJ, Pavón CA, Howard SR. Disorders of puberty and neurodevelopment: A shared etiology? Ann N Y Acad Sci 2024. [PMID: 39431640 DOI: 10.1111/nyas.15246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
The neuroendocrine control of puberty and reproduction is fascinatingly complex, with up- and down-regulation of key reproductive hormones during fetal, infantile, and later childhood periods that determine the correct function of the hypothalamic-pituitary-gonadal axis and the timing of puberty. Neuronal development is a vital element of these processes, and multiple conditions of disordered puberty and reproduction have their etiology in abnormal neuronal migration or function. Although there are numerous documented cases across multiple conditions wherein patients have both neurodevelopmental disorders and pubertal abnormalities, this has mostly been described ad hoc and the associations are not clearly documented. In this review, we aim to describe the overlap between these two groups of conditions and to increase awareness to ensure that puberty and reproductive function are carefully monitored in patients with neurodevelopmental conditions, and vice versa. Moreover, this commonality can be explored for clues about the disease mechanisms in these patient groups and provide new avenues for therapeutic interventions for affected individuals.
Collapse
Affiliation(s)
- Jordan E Read
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Alexandru Vasile-Tudorache
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
| | - Angel Newsome
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - María José Lorente
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
| | - Carmen Agustín Pavón
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
| | - Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Paediatric Endocrinology, Barts Health NHS Trust, London, UK
| |
Collapse
|
3
|
Pozzi E, Ila V, Petrella F, Corsini C, Ghomeshi A, Dureja R, Boaretto D, Somasundar T, Salonia A, Ramasamy R. Evaluating Sperm Recovery Time and Efficacy of Monotherapy vs. Combination Therapies in Men with Congenital Hypogonadotropic Hypogonadism: A Systematic Review and Meta-Analysis. World J Mens Health 2024; 42:42.e90. [PMID: 39434392 DOI: 10.5534/wjmh.240095] [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: 04/17/2024] [Revised: 07/13/2024] [Accepted: 07/27/2024] [Indexed: 10/23/2024] Open
Abstract
PURPOSE There is a lack of pooled data exploring the time and rates for human chorionic gonadotropin (hCG) monotherapy vs. combination therapies (hCG+human menopausal gonadotropin or recombinant human follicle-stimulating hormone) to restore spermatogenesis in azoospermic men with congenital hypogonadotropic hypogonadism (CHH). We aimed to investigate the time and rates to recover spermatogenesis among azoospermic CHH men receiving monotherapy vs. combination therapy. MATERIALS AND METHODS We conducted a systematic review and meta-analysis following the PRISMA guidelines. The search was performed on PubMed, EMBASE, Web of Science, and Scopus databases up to November 2023. Forrest plots were generated to visually present the pooled effect sizes for time to recover spermatogenesis, specifically employing the standardized mean difference (SMD). Publication bias was assessed utilizing funnel plots. PROSPERO ID: CRD42023473615. RESULTS The search identified 720 studies meeting inclusion criteria. Our meta-analysis of 1,240 men with CHH revealed significant differences in the time to recover spermatogenesis between combination therapies and monotherapy. The weighted mean recovery time was significantly shorter for combination therapies (10 months) compared to monotherapy (33 months). The SMD under the common effect model was 8.8 for combination therapies and 24.98 for monotherapy, indicating a more rapid recovery with combination therapies, p<0.01. The rates of sperm recovery were 66.76% for combination therapies and 51.9% for monotherapy, p=0.03. Significant heterogeneity was observed in both groups (I²=86% for combination therapies and I²=68% for monotherapy), suggesting considerable variation in individual responses. CONCLUSIONS The present meta-analysis reveals that in men with CHH, combination therapies expedite spermatogenesis recovery more than monotherapy. Additionally, combination therapies yield a higher rate of sperm appearing in the ejaculate as compared to hCG monotherapy. The significant heterogeneity observed in both groups underscores the variability in individual responses, warranting further investigation and caution in interpreting these results.
Collapse
Affiliation(s)
- Edoardo Pozzi
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology, Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Vishal Ila
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Francis Petrella
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Christian Corsini
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology, Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Armin Ghomeshi
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Rohan Dureja
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Daniel Boaretto
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Tharun Somasundar
- Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andrea Salonia
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology, Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ranjith Ramasamy
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
4
|
Pozzi E, Corsini C, Salonia A. Medical therapy for male infertility. Curr Opin Urol 2024:00042307-990000000-00195. [PMID: 39380445 DOI: 10.1097/mou.0000000000001231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
PURPOSE OF REVIEW To provide up-to-date evidence and clinical guidance on the role of medical therapy in the context of hormonal imbalances affecting human spermatogenesis. RECENT FINDINGS Compelling evidence has accumulated over the years regarding the role of gonadotropins, selective estrogen modulators, and aromatase inhibitors to either improve or restore spermatogenesis in men with hormonal abnormalities (e.g. hypogonadotropic/hypergonadotropic hypogonadism, hyperprolactinemia) or supraphysiologic levels (e.g. exogenous testosterone/anabolic steroid use). Despite the increasing number of studies being performed, most of the available evidence relies on small nonrandomized studies, mainly in men with hypergonadotropic hypogonadism or with history of exogenous testosterone/anabolic steroid use. As such, the efficacy of medical therapy is highly variable emphasizing the necessity of randomized clinical trials and individualized approaches. SUMMARY This narrative review provides clinical guidance on medical therapies for male factor infertility based on the most up-to-date evidence, focusing on treatments for hormonal abnormalities (either hypogonadotropic or hypergonadotropic hypogonadism and hyperprolactinemia) and supraphysiologic levels (and exogenous testosterone/anabolic steroid use) to improve spermatogenesis.
Collapse
Affiliation(s)
- Edoardo Pozzi
- Vita-Salute San Raffaele University
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Christian Corsini
- Vita-Salute San Raffaele University
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Salonia
- Vita-Salute San Raffaele University
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
5
|
Kumar Yadav R, Qi B, Wen J, Gang X, Banerjee S. Kallmann syndrome: Diagnostics and management. Clin Chim Acta 2024; 565:119994. [PMID: 39384129 DOI: 10.1016/j.cca.2024.119994] [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: 08/20/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
Kallmann syndrome is a genetic disorder characterized by delayed or absence of puberty and a reduced or absent sense of smell (anosmia). Kallmann syndrome is a form of hypogonadotropic hypogonadism due to lack of the production of sex hormones which is associated with development of secondary sexual characteristics. Kallmann Syndrome is a genetically heterogeneous disorder, characterized by the combination of hypogonadotropic hypogonadism (a deficiency in sex hormone production) and anosmia. Germline mutations in KAL1 gene causes deficiency in GnRH hormone followed by low level of circulating gonadotropin and testosterone which finally leads to the failure of puberty (development of secondary sexual characters). Kallmann Syndrome can be inherited in several manners including X-linked recessive (e.g., mutations within KAL1) and autosomal dominant and recessive forms. Germline mutation in KAL1 gene was identified among 8% of patients with Kallmann Syndrome. A review of the recent literature done reveals numerous clinical manifestations in Kallmann Syndrome patients with the KAL1 mutation, including microgenitalia, impotence, reduced libido, infertility, unilateral renal agenesis, and synkinesia. Genetic molecular diagnostics through prenatal diagnosis and preimplantation genetic testing are most significant way to reduce the risk of Kallmann syndrome in next generation. Complication associated with Kallmann syndrome can be prevented by early diagnosis, diet supplementation and medical therapy. Goal of therapeutic intervention is to the development of secondary sexual characteristics, build and sustain bone density as well as muscle mass and restore fertility. This review aims to explore the genetic diagnosis and management strategies for Kallmann Syndrome, particularly focusing on KAL1 gene mutations.
Collapse
Affiliation(s)
- Rajiv Kumar Yadav
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China; Department of Endocrinology (Internal Medicine), First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Baiyu Qi
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Jianping Wen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Xiaokun Gang
- Department of Endocrinology (Internal Medicine), First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Santasree Banerjee
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
6
|
Singhania N, Devi KB, Kaur J, Bhansali A, Gorsi U, Sachdeva N, Arora S, Rai A, Walia R. Effect of Combined Low Dose Human Gonadotropic Hormone, Follicle Stimulating Hormone, and Testosterone Therapy (LFT Regimen) Versus Conventional High Dose Human Gonadotropic Hormone and Follicle Stimulating Hormone on Spermatogenesis and Biomarkers in Men With Hypogonadotropic Hypogonadism. Endocr Pract 2024; 30:978-986. [PMID: 39025301 DOI: 10.1016/j.eprac.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVE In male congenital hypogonadotropic hypogonadism (CHH), it was observed that lower dose human gonadotropic hormone (hCG) can maintain normal intratesticular testosterone levels. We propose this study to compare the low-dose hCG, follicle stimulating hormone (FSH), and Testosterone (T) [LFT Regimen] to conventional treatment to induce virilization and fertility. DESIGN This open-label randomized pilot study was conducted from June 2020 to December 2021. SUBJECTS AND OUTCOME MEASURES CHH were randomly assigned to either the LFT regimen (Group A)-low-dose hCG (500U thrice per week), FSH (150U thrice per week), and T(100 mg biweekly) or conventional therapy(GroupB) with high hCG dose(2000U thrice per week) and the same FSH dose. The hCG dosage was titrated to reduce anti-mullerian hormone (AMH) by 50% and normalization of plasma T in groups A and B, respectively. The primary objective was to compare the percentage of individuals who achieved spermatogenesis between the two groups. RESULTS Out of 30 patients, 23 (76·7%) subjects achieved spermatogenesis, and the median time was 12 (9-14·9) months. There was no difference in achieving spermatogenesis between the two groups (64·3% vs 7·5%,P = 0·204), and even the median time for spermatogenesis was similar (15months vs 12months,P = 0·248). Both groups had nonsignificant median plasma AMH at spermatogenesis, [6·6 ng/ml (3·3-9·76) vs4·41 ng/ml (2·3-6·47), P = 0·298]. Similarly, the median plasma Inhibin B at spermatogenesis between groups were comparable [152·4 pg/ml (101·7-198·0) vs49·1 pg/ml (128·7-237·3), P = 0·488]. CONCLUSIONS A reasonable approach to induce fertility in male CHH is to initiate combination therapy using FSH, low-dose hCG targeting AMH <6·9 ng/ml, along with T to achieve normal range. Monitoring AMH could serve as a proxy indicator of spermatogenesis.
Collapse
Affiliation(s)
| | | | - Japleen Kaur
- Department of Obstetrics and Gynecology, PGIMER, Chandigarh, India
| | - Anil Bhansali
- Department of Endocrinology, PGIMER, Chandigarh, India
| | - Ujjwal Gorsi
- Department of Radiology, PGIMER, Chandigarh, India
| | | | - Sunil Arora
- Department of Translational and Regenerative Medicine, PGIMER, Chandigarh, India
| | - Ashutosh Rai
- Department of Endocrinology, PGIMER, Chandigarh, India
| | - Rama Walia
- Department of Endocrinology, PGIMER, Chandigarh, India.
| |
Collapse
|
7
|
Donald DM, McDonnell T, O'Reilly MW, Sherlock M. Replacement with sex steroids in hypopituitary men and women: implications for gender differences in morbidities and mortality. Rev Endocr Metab Disord 2024; 25:839-854. [PMID: 39370498 PMCID: PMC11470859 DOI: 10.1007/s11154-024-09897-7] [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] [Accepted: 07/17/2024] [Indexed: 10/08/2024]
Abstract
Hypopituitarism is a heterogenous disorder characterised by a deficiency in one or more anterior pituitary hormones. There are marked sex disparities in the morbidity and mortality experienced by patients with hypopituitarism. In women with hypopituitarism, the prevalence of many cardiovascular risk factors, myocardial infarction, stroke and mortality are significantly elevated compared to the general population, however in men, they approach that of the general population. The hypothalamic-pituitary-gonadal axis (HPG) is the most sexually dimorphic pituitary hormone axis. Gonadotropin deficiency is caused by a deficiency of either hypothalamic gonadotropin-releasing hormone (GnRH) or pituitary gonadotropins, namely follicle-stimulating hormone (FSH) and luteinising hormone (LH). HPG axis dysfunction results in oestrogen and testosterone deficiency in women and men, respectively. Replacement of deficient sex hormones is the mainstay of treatment in individuals not seeking fertility. Oestrogen and testosterone replacement in women and men, respectively, have numerous beneficial health impacts. These benefits include improved body composition, enhanced insulin sensitivity, improved atherogenic lipid profiles and increased bone mineral density. Oestrogen replacement in women also reduces the risk of developing type 2 diabetes mellitus. When women and men are considered together, untreated gonadotropin deficiency is independently associated with an increased mortality risk. However, treatment with sex hormone replacement reduces the mortality risk comparable to those with an intact gonadal axis. The reasons for the sex disparities in mortality remain poorly understood. Potential explanations include the reversal of women's natural survival advantage over men, premature loss of oestrogen's cardioprotective effect, less aggressive cardiovascular risk factor modification and inadequate oestrogen replacement in women with gonadotropin deficiency. Regrettably, historical inertia and unfounded concerns about the safety of oestrogen replacement in women of reproductive age have impeded the treatment of gonadotropin deficiency.
Collapse
Affiliation(s)
- Darran Mc Donald
- Department of Endocrinology, Beaumont Hospital, Royal College of Surgeons of Ireland, Dublin 9, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tara McDonnell
- Department of Endocrinology, Beaumont Hospital, Royal College of Surgeons of Ireland, Dublin 9, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael W O'Reilly
- Department of Endocrinology, Beaumont Hospital, Royal College of Surgeons of Ireland, Dublin 9, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mark Sherlock
- Department of Endocrinology, Beaumont Hospital, Royal College of Surgeons of Ireland, Dublin 9, Dublin, Ireland.
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
8
|
Chachlaki K, Duc KL, Storme L, Prévot V. Novel insights into minipuberty and GnRH: Implications on neurodevelopment, cognition, and COVID-19 therapeutics. J Neuroendocrinol 2024; 36:e13387. [PMID: 38565500 PMCID: PMC7616535 DOI: 10.1111/jne.13387] [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: 01/03/2024] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
In humans, the first 1000 days of life are pivotal for brain and organism development. Shortly after birth, gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus are activated, a phenomenon known as minipuberty. This phenomenon, observed in all mammals studied, influences the postnatal development of the hypothalamic-pituitary-gonadal (HPG) axis and reproductive function. This review will put into perspective the results of recent studies showing that the impact of minipuberty extends beyond reproductive function, influencing sensory and cognitive maturation. Studies in mice have revealed the role of nitric oxide (NO) in regulating minipuberty amplitude, with NO deficiency linked to cognitive and olfactory deficits. Additionally, findings indicate that cognitive and sensory defects in adulthood in a mouse model of Down syndrome are associated with an age-dependent decline of GnRH production, whose origin can be traced back to minipuberty, and point to the potential therapeutic role of pulsatile GnRH administration in cognitive disorders. Furthermore, this review delves into the repercussions of COVID-19 on GnRH production, emphasizing potential consequences for neurodevelopment and cognitive function in infected individuals. Notably, GnRH neurons appear susceptible to SARS-CoV-2 infection, raising concerns about potential long-term effects on brain development and function. In conclusion, the intricate interplay between GnRH neurons, GnRH release, and the activity of various extrahypothalamic brain circuits reveals an unexpected role for these neuroendocrine neurons in the development and maintenance of sensory and cognitive functions, supplementing their established function in reproduction. Therapeutic interventions targeting the HPG axis, such as inhaled NO therapy in infancy and pulsatile GnRH administration in adults, emerge as promising approaches for addressing neurodevelopmental cognitive disorders and pathological aging.
Collapse
Affiliation(s)
- Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| | - Kevin Le Duc
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Laurent Storme
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| |
Collapse
|
9
|
Dwyer AA, McDonald IR, Quinton R. Current landscape of fertility induction in males with congenital hypogonadotropic hypogonadism. Ann N Y Acad Sci 2024; 1540:133-146. [PMID: 39190467 PMCID: PMC11471374 DOI: 10.1111/nyas.15214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Congenital hypogonadotropic hypogonadism (CHH) is a rare reproductive disorder caused by deficient secretion or action of gonadotropin-releasing hormone (GnRH) and is a hormonally treatable form of male infertility. Both pulsatile GnRH treatment and combined gonadotropin therapy effectively induce spermatogenesis in 75%-80% of males with CHH, albeit the ejaculate does not usually approach normal semen parameters by WHO criteria. This is in some contrast to the cumulative fertility outcomes in females with CHH on gonadotropin treatment that are indistinguishable from those of reproductively normal females. Emerging data provide insights into early life determinants of male fertility (i.e., minipuberty), and research has identified key predictors of outcomes for fertility-inducing treatment in men with CHH. Such developments provide mounting evidence for tailoring approaches to maximize fertility potential in CHH, although there is no clear consensus to date on the optimal approach to fertility-inducing treatment. This review provides an up-to-date review on the current evidence underpinning therapeutic approaches for inducing spermatogenesis in males with CHH. In the absence of evidence-based clinical guidelines, this synthesis of current evidence provides guidance for clinicians working with males with CHH seeking fertility.
Collapse
Affiliation(s)
- Andrew A Dwyer
- P50 Massachusetts General Hospital-Harvard Center for Reproductive Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- William F. Connell School of Nursing, Boston College, Chestnut Hill, Massachusetts, USA
| | - Isabella R McDonald
- William F. Connell School of Nursing, Boston College, Chestnut Hill, Massachusetts, USA
| | - Richard Quinton
- Department of Metabolism, Digestion & Reproduction, Imperial College London, London, UK
- Northern Regional Gender Dysphoria Service, Cumbria, Northumberland, Tyne & Wear NHS Foundation Trust, Newcastle, UK
| |
Collapse
|
10
|
Devi KB, Gorsi U, Lal A, Saini S, Jerath A, Thingujam D, Baruah MM, Walia R. Evaluation of testicular volume in males with congenital hypogonadotropic hypogonadism: a comparative analysis. Endocrine 2024:10.1007/s12020-024-04032-7. [PMID: 39320590 DOI: 10.1007/s12020-024-04032-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Estimating accurate testicular volume (TV) of congenital hypogonadotropic hypogonadism (CHH) individuals is challenging due to the typically small testicular size. Ultrasound (USG) emerges as a vital solution, enabling precise measurements and reproducible results. The purpose of the study was to assess the three-dimensional measurement of the testis using USG and its volume was estimated using Ellipsoid (E) and Lambert (L) formulae and compared these with the TV by Prader orchidometer (OrTV). METHODS This is an exploratory analysis of data taken from a clinical trial conducted from May 2022 to March 2024 which included 94 testes from 47 CHH participants. The OrTVs and USGTVs were assessed at baseline and every three months till the completion of the study making a total of 348 observations. The three-dimensional measurement of the testes was noted and TVs were calculated using the above formulae. RESULTS The mean age of the participants was 25.8 ± 6.14 years with a mean height of 169.9 ± 8.42 cm and body mass index (BMI) of 22.4 ± 4.72 kg/m2. The baseline mean OrTV, USGTV(E) and USGTV(L) were 2.15 ± 0.79 ml, 0.69 ± 0.43 ml and 0.93 ± 0.59 ml respectively. The smallest OrTV observed was 1 ml with its respective mean USGTV of 0.41 ± 0.2 ml(E) and 0.56 ± 0.27 ml(L). An OrTV of 4 ml had a mean USGTV of 1.11 ± 0.42 ml(E) and 1.51 ± 9.57 ml(L). At spermatogenesis, the mean OrTV was 8.84 ± 3.13 ml with the USGTV determined to be 4 ± 1.46 ml(E) and 5.46 ± 1.99 ml(L). CONCLUSION The study revealed that all CHH patients at diagnosis had OrTV < 4 ml. This corresponds to a USG TV cut-off of 1.11 ml using the Ellipsoid formula and 1.51 ml with the Lambert formula, which could serve as a USG diagnostic criterion for CHH.
Collapse
Affiliation(s)
- Konsam Biona Devi
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ujjwal Gorsi
- Department of Radiodiagnosis, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anupam Lal
- Department of Radiodiagnosis, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Shubham Saini
- Department of Radiodiagnosis, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Akhil Jerath
- Department of Radiodiagnosis, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Mintu Mani Baruah
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Rama Walia
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
11
|
Wang X, Liu Q, Zhuang Z, Cheng J, Zhang W, Jiang Q, Guo Y, Li R, Lu X, Cui L, Weng J, Tang Y, Yue J, Gao S, Hong K, Qiao J, Jiang H, Guo J, Zhang Z. Decoding the pathogenesis of spermatogenic failure in cryptorchidism through single-cell transcriptomic profiling. Cell Rep Med 2024; 5:101709. [PMID: 39226895 PMCID: PMC11528238 DOI: 10.1016/j.xcrm.2024.101709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/20/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
Cryptorchidism, commonly known as undescended testis, affects 1%-9% of male newborns, posing infertility and testis tumor risks. Despite its prevalence, the detailed pathophysiology underlying male infertility within cryptorchidism remains unclear. Here, we profile and analyze 46,644 single-cell transcriptomes from individual testicular cells obtained from adult males diagnosed with cryptorchidism and healthy controls. Spermatogenesis compromise in cryptorchidism links primarily to spermatogonium self-renewal and differentiation dysfunctions. We illuminate the involvement of testicular somatic cells, including immune cells, thereby unveiling the activation and degranulation of mast cells in cryptorchidism. Mast cells are identified as contributors to interstitial fibrosis via transforming growth factor β1 (TGF-β1) and cathepsin G secretion. Furthermore, significantly increased levels of secretory proteins indicate mast cell activation and testicular fibrosis in the seminal plasma of individuals with cryptorchidism compared to controls. These insights serve as valuable translational references, enriching our comprehension of testicular pathogenesis and informing more precise diagnosis and targeted therapeutic strategies for cryptorchidism.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Qiang Liu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Ziyan Zhuang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Jianxing Cheng
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Wenxiu Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Qiaoling Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yifei Guo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Ran Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Xiaojian Lu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Lina Cui
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Jiaming Weng
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yanlin Tang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Jingwei Yue
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Songzhan Gao
- Department of Andrology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kai Hong
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing, China; Department of Urology, Institute of Urology, Peking University First Hospital, Beijing, China.
| | - Jingtao Guo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China.
| | - Zhe Zhang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Department of Urology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
12
|
Chen X, Hu M, Du T, Yang L, Li Y, Feng L, Luo J, Yao H, Chen X. Homozygous mutation of KISS1 receptor ( KISS1R) gene identified in a Chinese patient with congenital hypogonadotropic hypogonadism (CHH): case report and literature review. J Pediatr Endocrinol Metab 2024:jpem-2024-0119. [PMID: 39262158 DOI: 10.1515/jpem-2024-0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/24/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVES Congenital hypogonadotropic hypogonadism (CHH) is a rare condition caused by a defect in the production, secretion or action of gonadotropin-releasing hormone. The absence of puberty and varying degrees of gonadotropic deficiency are common symptoms of this disorder. Heterogeneity exists in the clinical presentation of the different clinical subtypes and multiple genes have been implicated in CHH. A number of genetic defects have been identified as causes normosmic CHH, including mutations of GnRHR, GNRH1, KISS1R, KISS1, TACR3 and TAC3. Loss-of-function mutations in KISS1R gene are a rare cause of normosmic CHH. CASE PRESENTATION We described an 11.5 years old Chinese patient who presented at birth with micropenis, microorchidia and bilateral cryptorchidism. Whole-exome sequencing was also performed and identified a homozygous mutation of KISS1R gene, c.1010_1028del (p.V337Afs*82). The variant was predicted as "deleterious" and classified as "likely pathogenic". This variant has never been reported in patients with CHH. Furthermore, we summarized the clinical presentations and analyzed the phenotype-genotype correlation between CHH and KISS1R mutations in previous reports. CONCLUSIONS This study details the clinical phenotypes and hormone levels of the patient and expands the spectrum of mutations in the KISS1R gene associated with CHH.
Collapse
Affiliation(s)
- Xiaoqian Chen
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Man Hu
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Du
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Luhong Yang
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yakun Li
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lifang Feng
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Luo
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yao
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Chen
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Carriço JN, Gonçalves CI, Al-Naama A, Syed N, Aragüés JM, Bastos M, Fonseca F, Borges T, Pereira BD, Pignatelli D, Carvalho D, Cunha F, Saavedra A, Rodrigues E, Saraiva J, Ruas L, Vicente N, Martin Martins J, De Sousa Lages A, Oliveira MJ, Castro-Correia C, Melo M, Martins RG, Couto J, Moreno C, Martins D, Oliveira P, Martins T, Martins SA, Marques O, Meireles C, Garrão A, Nogueira C, Baptista C, Gama-de-Sousa S, Amaral C, Martinho M, Limbert C, Barros L, Vieira IH, Sabino T, Saraiva LR, Lemos MC. Genetic architecture of congenital hypogonadotropic hypogonadism: insights from analysis of a Portuguese cohort. Hum Reprod Open 2024; 2024:hoae053. [PMID: 39308770 PMCID: PMC11415827 DOI: 10.1093/hropen/hoae053] [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: 04/11/2024] [Revised: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
STUDY QUESTION What is the contribution of genetic defects in Portuguese patients with congenital hypogonadotropic hypogonadism (CHH)? SUMMARY ANSWER Approximately one-third of patients with CHH were found to have a genetic cause for their disorder, with causal pathogenic and likely pathogenic germline variants distributed among 10 different genes; cases of oligogenic inheritance were also included. WHAT IS KNOWN ALREADY CHH is a rare and genetically heterogeneous disorder characterized by deficient production, secretion, or action of GnRH, LH, and FSH, resulting in delayed or absent puberty, and infertility. STUDY DESIGN SIZE DURATION Genetic screening was performed on a cohort of 81 Portuguese patients with CHH (36 with Kallmann syndrome and 45 with normosmic hypogonadotropic hypogonadism) and 263 unaffected controls. PARTICIPANTS/MATERIALS SETTING METHODS The genetic analysis was performed by whole-exome sequencing followed by the analysis of a virtual panel of 169 CHH-associated genes. The main outcome measures were non-synonymous rare sequence variants (population allele frequency <0.01) classified as pathogenic, likely pathogenic, and variants of uncertain significance (VUS). MAIN RESULTS AND THE ROLE OF CHANCE A genetic cause was identified in 29.6% of patients. Causal pathogenic and likely pathogenic variants were distributed among 10 of the analysed genes. The most frequently implicated genes were GNRHR, FGFR1, ANOS1, and CHD7. Oligogenicity for pathogenic and likely pathogenic variants was observed in 6.2% of patients. VUS and oligogenicity for VUS variants were observed in 85.2% and 54.3% of patients, respectively, but were not significantly different from that observed in controls. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION The identification of a large number of VUS presents challenges in interpretation and these may require reclassification as more evidence becomes available. Non-coding and copy number variants were not studied. Functional studies of the variants were not undertaken. WIDER IMPLICATIONS OF THE FINDINGS This study highlights the genetic heterogeneity of CHH and identified several novel variants that expand the mutational spectrum of the disorder. A significant proportion of patients remained without a genetic diagnosis, suggesting the involvement of additional genetic, epigenetic, or environmental factors. The high frequency of VUS underscores the importance of cautious variant interpretation. These findings contribute to the understanding of the genetic architecture of CHH and emphasize the need for further studies to elucidate the underlying mechanisms and identify additional causes of CHH. STUDY FUNDING/COMPETING INTERESTS This research was funded by the Portuguese Foundation for Science and Technology (grant numbers PTDC/SAU-GMG/098419/2008, UIDB/00709/2020, CEECINST/00016/2021/CP2828/CT0002, and 2020.04924.BD) and by Sidra Medicine-a member of the Qatar Foundation (grant number SDR400038). The authors declare no competing interests.
Collapse
Affiliation(s)
- Josianne Nunes Carriço
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | | | | | - José Maria Aragüés
- Serviço de Endocrinologia, Diabetes e Metabolismo, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Margarida Bastos
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Fernando Fonseca
- Serviço de Endocrinologia, Hospital de Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal
| | - Teresa Borges
- Unidade de Endocrinologia Pediátrica, Serviço de Pediatria, Centro Materno Infantil do Norte, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | | | - Duarte Pignatelli
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Davide Carvalho
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Filipe Cunha
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Ana Saavedra
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Elisabete Rodrigues
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Joana Saraiva
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Luisa Ruas
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Nuno Vicente
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - João Martin Martins
- Serviço de Endocrinologia, Diabetes e Metabolismo, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Adriana De Sousa Lages
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Maria João Oliveira
- Unidade de Endocrinologia Pediátrica, Serviço de Pediatria, Centro Materno Infantil do Norte, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Cíntia Castro-Correia
- Unidade de Endocrinologia e Diabetologia Pediátrica, Departamento de Pediatria, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Miguel Melo
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Raquel Gomes Martins
- Serviço de Endocrinologia, Instituto Português de Oncologia do Porto, Porto, Portugal
| | - Joana Couto
- Serviço de Endocrinologia, Instituto Português de Oncologia do Porto, Porto, Portugal
| | - Carolina Moreno
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Diana Martins
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Patrícia Oliveira
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Teresa Martins
- Serviço de Endocrinologia, Instituto Português de Oncologia de Coimbra, Coimbra, Portugal
| | - Sofia Almeida Martins
- Unidade de Endocrinologia Pediátrica, Serviço de Pediatria, Hospital de Braga, Braga, Portugal
| | - Olinda Marques
- Serviço de Endocrinologia, Hospital de Braga, Braga, Portugal
| | - Carla Meireles
- Serviço de Pediatria, Hospital da Senhora da Oliveira Guimarães, Guimarães, Portugal
| | - António Garrão
- Departamento de Endocrinologia, Hospital da Luz Lisboa, Lisboa, Portugal
| | - Cláudia Nogueira
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Carla Baptista
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Susana Gama-de-Sousa
- Serviço de Pediatria, Centro Hospitalar do Médio Ave, Unidade de V. N. Famalicão, Vila Nova de Famalicão, Portugal
| | - Cláudia Amaral
- Serviço de Endocrinologia, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Mariana Martinho
- Serviço de Endocrinologia, Centro Hospitalar do Tâmega e Sousa, Guilhufe, Portugal
| | - Catarina Limbert
- Unidade de Endocrinologia Pediátrica, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Luisa Barros
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Inês Henriques Vieira
- Serviço de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Teresa Sabino
- Serviço de Endocrinologia, Hospital de Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal
| | - Luís R Saraiva
- Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Manuel Carlos Lemos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
14
|
Everaere H, Simon V, Bachelot A, Leroy M, Decanter C, Dewailly D, Catteau-Jonard S, Robin G. Pulsatile gonadotropin-releasing hormone therapy: comparison of efficacy between functional hypothalamic amenorrhea and congenital hypogonadotropic hypogonadism. Fertil Steril 2024:S0015-0282(24)02007-7. [PMID: 39233038 DOI: 10.1016/j.fertnstert.2024.08.354] [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: 02/29/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE To compare the ongoing pregnancy rate per initiated cycle between patients with functional hypothalamic amenorrhea (FHA) and patients with congenital hypogonadotropic hypogonadism (CHH) treated with pulsatile gonadotropin-releasing hormone (GnRH) administration. DESIGN Retrospective monocentric cohort study conducted at the University Hospital of Lille from 2004 to 2022. SETTING Lille University Hospital, Department of Endocrine Gynecology. PATIENT(S) A total of 141 patients diagnosed with central suprapituitary amenorrhea during infertility evaluation and subsequently treated with pulsatile GnRH therapy. 111 and 30 patients were diagnosed with FHA or CHH, respectively. INTERVENTION(S) Pulsatile GnRH administration. MAIN OUTCOME MEASURE(S) Ongoing pregnancy rate per initiated cycle. RESULT(S) Ongoing pregnancy rates per initiated cycle were comparable between groups: 21.5% in the FHA group vs. 22% in the CHH group. Comparison of baseline characteristics showed a more pronounced follicle-stimulating hormone (FSH) deficiency in patients with CHH than in those with FHA: 2.55 (0.6-4.92) vs. 4.80 (3.90-5.70) UI/L. Within the CHH group, basal FSH level was positively associated with the occurrence of ongoing pregnancies (odds ratio, 1.57; 95% confidence interval, 1.11-2.22). In the CHH group, the duration of treatment was higher than in the FHA group: 23.59 (± 8.02) vs. 18.16 (± 7.66) days. CONCLUSION(S) The baseline FSH level is lower in patients with CHH than in patients with FHA. The lower the FSH, the lower the chance of pregnancy in patients with CHH. These patients also require more days of GnRH administration. However, the rate of ongoing pregnancies is comparable between the two groups.
Collapse
Affiliation(s)
- Hortense Everaere
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France.
| | - Virginie Simon
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Development and Plasticity of the Neuroendocrine Brain, U1172-Lille Neurosciences and Cognition (Jean-Pierre Aubert Research Center)-Lille Neurosciences and Cognition, Université Lille, Lille, France
| | - Anne Bachelot
- Endocrinology and Reproductive Medicine Department, Reference Center for Rare Growth and Development Endocrine Diseases, Assistance Publique-Hôpitaux de Paris, Sorbonne University, Pitié Salpêtrière University Hospital, Paris, France
| | - Maxime Leroy
- Biostatistics Department, Centre Hospitalier Universitaire Lille, Lille, France
| | - Christine Decanter
- Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France
| | | | - Sophie Catteau-Jonard
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Development and Plasticity of the Neuroendocrine Brain, U1172-Lille Neurosciences and Cognition (Jean-Pierre Aubert Research Center)-Lille Neurosciences and Cognition, Université Lille, Lille, France
| | - Geoffroy Robin
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Development and Plasticity of the Neuroendocrine Brain, U1172-Lille Neurosciences and Cognition (Jean-Pierre Aubert Research Center)-Lille Neurosciences and Cognition, Université Lille, Lille, France
| |
Collapse
|
15
|
Stankevice D, Fjaeldstad AW, Ovesen T. Smell and taste disorders in childhood: Diagnostic challenges and significant impacts on a child's well-being. Int J Pediatr Otorhinolaryngol 2024; 184:112081. [PMID: 39208514 DOI: 10.1016/j.ijporl.2024.112081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
AIM Smell and taste are senses that contribute to a child's overall well-being. Disorders affecting these senses can impact a child's daily life from enjoying meals to detecting potential dangers through scent. The aim of this study is to describe patient characteristics and etiological causes of olfactory (OD) and/or gustatory disorders (GD) in children referred to a smell and taste clinic. Secondly, we aim to suggest a clinical work up. METHODS Retrospective study where data were collected from 57 children who were referred consecutively to the University Clinic for Flavour, Balance, and Sleep; Department of Otorhinolaryngology (ORL), Head and Neck Surgery; Goedstrup Hospital, Denmark, for assessment due to OD/GD from January 2017 to May 2023. RESULTS Most of the children had anosmia (60 %), whereas sensation of the basic tastes was intact in all but eight children (16 %). The lowest TDI scores were in children with congenital OD. The underlying etiology was congenital followed by postinfectious mostly related to Covid-19. Picky eating including anorectic traits were seen in 16 % of patients. CONCLUSION The focus on smell loss in pediatric population is low, and probably does not adequately reflect either underlying prevalence in this group or the possible consequences on a child's well-being. Moreover, increased awareness of children's smell and taste loss is needed, as it may be associated with eating disturbances.
Collapse
Affiliation(s)
- D Stankevice
- University Clinic for Flavour, Balance and Sleep, Department of Otorhinolaryngology, Head and Neck Surgery, Goedstrup Hospital, Hospitalsparken 15, DK-7400, Herning, Denmark.
| | - A W Fjaeldstad
- University Clinic for Flavour, Balance and Sleep, Department of Otorhinolaryngology, Head and Neck Surgery, Goedstrup Hospital, Hospitalsparken 15, DK-7400, Herning, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensen's Boulevard 82, DK-8200, Aarhus N, Denmark
| | - T Ovesen
- University Clinic for Flavour, Balance and Sleep, Department of Otorhinolaryngology, Head and Neck Surgery, Goedstrup Hospital, Hospitalsparken 15, DK-7400, Herning, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensen's Boulevard 82, DK-8200, Aarhus N, Denmark
| |
Collapse
|
16
|
Decoster L, Trova S, Zucca S, Bulk J, Gouveia A, Ternier G, Lhomme T, Legrand A, Gallet S, Boehm U, Wyatt A, Wahl V, Wartenberg P, Hrabovszky E, Rácz G, Luzzati F, Nato G, Fogli M, Peretto P, Schriever SC, Bernecker M, Pfluger PT, Steculorum SM, Bovetti S, Rasika S, Prevot V, Silva MSB, Giacobini P. A GnRH neuronal population in the olfactory bulb translates socially relevant odors into reproductive behavior in male mice. Nat Neurosci 2024; 27:1758-1773. [PMID: 39095587 DOI: 10.1038/s41593-024-01724-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons regulate fertility and integrate hormonal status with environmental cues to ensure reproductive success. Here we show that GnRH neurons in the olfactory bulb (GnRHOB) of adult mice can mediate social recognition. Specifically, we show that GnRHOB neurons extend neurites into the vomeronasal organ and olfactory epithelium and project to the median eminence. GnRHOB neurons in males express vomeronasal and olfactory receptors, are activated by female odors and mediate gonadotropin release in response to female urine. Male preference for female odors required the presence and activation of GnRHOB neurons, was impaired after genetic inhibition or ablation of these cells and relied on GnRH signaling in the posterodorsal medial amygdala. GnRH receptor expression in amygdala kisspeptin neurons appear to be required for GnRHOB neurons' actions on male mounting behavior. Taken together, these results establish GnRHOB neurons as regulating fertility, sex recognition and mating in male mice.
Collapse
Affiliation(s)
- Laurine Decoster
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sara Trova
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Centro CMP3VdA, Istituto Italiano di Tecnologia (IIT), Aosta, Italy
| | - Stefano Zucca
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Janice Bulk
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Ayden Gouveia
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Amandine Legrand
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sarah Gallet
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Hun-Ren Institute of Experimental Medicine, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Federico Luzzati
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Giulia Nato
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Marco Fogli
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sonja C Schriever
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Miriam Bernecker
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Paul T Pfluger
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Sophie M Steculorum
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sowmyalakshmi Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Mauro S B Silva
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France.
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France.
| |
Collapse
|
17
|
Friedrich C, Tüttelmann F. Genetics of female and male infertility. MED GENET-BERLIN 2024; 36:161-170. [PMID: 39253719 PMCID: PMC11380935 DOI: 10.1515/medgen-2024-2040] [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] [Indexed: 09/11/2024]
Abstract
Infertility is defined as the inability to conceive within one year of unprotected intercourse, and the causes are equally distributed between both sexes. Genetics play a crucial role in couple infertility and respective diagnostic testing should follow available guidelines. Appropriate tiered genetic analyses require comprehensive physical examination of both partners in an infertile couple. A wide range of chromosomal and monogenic variants can be the underlying genetic cause of infertility in both women and men. Accurate clinical phenotyping, together with identification of the genetic origin, helps to recommend the proper treatment and to counsel couples on the success rates and potential risks for offspring.
Collapse
Affiliation(s)
- Corinna Friedrich
- University and University Hospital Münster Centre of Medical Genetics, Institute of Reproductive Genetics Vesaliusweg 12-14 48149 Münster Germany
| | - Frank Tüttelmann
- University and University Hospital Münster Centre of Medical Genetics, Institute of Reproductive Genetics Vesaliusweg 12-14 48149 Münster Germany
| |
Collapse
|
18
|
Zouaghi Y, Choudhary AM, Irshad S, Adamo M, Rehman KU, Fatima A, Shahid M, Najmi N, De Azevedo Correa F, Habibi I, Boizot A, Niederländer NJ, Ansar M, Santoni F, Acierno J, Pitteloud N. Genome sequencing reveals novel causative structural and single nucleotide variants in Pakistani families with congenital hypogonadotropic hypogonadism. BMC Genomics 2024; 25:787. [PMID: 39143522 PMCID: PMC11325732 DOI: 10.1186/s12864-024-10598-3] [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: 10/23/2023] [Accepted: 07/05/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES This study aims to elucidate the genetic causes of congenital hypogonadotropic hypogonadism (CHH), a rare genetic disorder resulting in GnRH deficiency, in six families from Pakistan. METHODS Eighteen DNA samples from six families underwent genome sequencing followed by standard evaluation for pathogenic single nucleotide variants (SNVs) and small indels. All families were subsequently analyzed for pathogenic copy number variants (CNVs) using CoverageMaster. RESULTS Novel pathogenic homozygous SNVs in known CHH genes were identified in four families: two families with variants in GNRHR, and two others harboring KISS1R variants. Subsequent investigation of CNVs in the remaining two families identified novel unique large deletions in ANOS1. CONCLUSION A combined, systematic analysis of single nucleotide and CNVs helps to improve the diagnostic yield for variants in patients with CHH.
Collapse
Affiliation(s)
- Yassine Zouaghi
- University of Lausanne, Lausanne, Switzerland
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
| | - Anbreen Mazhar Choudhary
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
- FMH College of Medicine & Dentistry, Lahore, Pakistan
| | - Saba Irshad
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Michela Adamo
- University of Lausanne, Lausanne, Switzerland
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
| | | | - Ambrin Fatima
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Mariam Shahid
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Nida Najmi
- Department of Obstetrics and Gynaecology, The Aga Khan University Hospital, Karachi, Pakistan
| | - Fernanda De Azevedo Correa
- University of Lausanne, Lausanne, Switzerland
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
| | - Imen Habibi
- University of Lausanne, Lausanne, Switzerland
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
| | - Alexia Boizot
- University of Lausanne, Lausanne, Switzerland
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
| | - Nicolas J Niederländer
- University of Lausanne, Lausanne, Switzerland
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
| | - Muhammad Ansar
- Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile Des Aveugles, Lausanne, Switzerland
- Advanced Molecular Genetics and Genomics Disease Research and Treatment Centre, Dow University of Health Sciences, Karachi, Pakistan
| | - Federico Santoni
- University of Lausanne, Lausanne, Switzerland
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
- , Medigenome, Geneva, Switzerland
| | - James Acierno
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland
| | - Nelly Pitteloud
- University of Lausanne, Lausanne, Switzerland.
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Avenue de La Sallaz 8, Lausanne, CH-1011, Switzerland.
| |
Collapse
|
19
|
Bonomi M, Cangiano B, Cianfarani S, Garolla A, Gianfrilli D, Lanfranco F, Rastrelli G, Sbardella E, Corona G, Isidori AM, Rochira V. "Management of andrological disorders from childhood and adolescence to transition age: guidelines from the Italian Society of Andrology and Sexual Medicine (SIAMS) in collaboration with the Italian Society for Pediatric Endocrinology and Diabetology (SIEDP)-Part-1". J Endocrinol Invest 2024:10.1007/s40618-024-02435-x. [PMID: 39126560 DOI: 10.1007/s40618-024-02435-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
PURPOSE Andrological pathologies in the adulthood are often the results of conditions that originate during childhood and adolescence and sometimes even during gestation and neonatal period. Unfortunately, the reports in the literature concerning pediatric andrological diseases are scares and mainly concerning single issues. Furthermore, no shared position statement are so far available. METHODS The Italian Society of Andrology and Sexual Medicine (SIAMS) commissioned an expert task force involving the Italian Society of Pediatric Endocrinology and Diabetology (SIEDP) to provide an updated guideline on the diagnosis and management of andrological disorders from childhood and adolescence to transition age. Derived recommendations were based on the grading of recommendations, assessment, development, and evaluation (GRADE) system. RESULTS A literature search of articles in English for the term "varicoceles", "gynecomastia", "fertility preservation", "macroorchidism", "precocious puberty" and "pubertal delay" has been performed. Three major aspects for each considered disorder were assessed including diagnosis, clinical management, and treatment. Recommendations and suggestions have been provided for each of the mentioned andrological disorders. CONCLUSIONS These are the first guidelines based on a multidisciplinary approach that involves important societies related to the field of andrological medicine from pediatric to transition and adult ages. This fruitful discussion allowed for a general agreement on several recommendations and suggestions to be reached, which can support all stakeholders in improving andrological and general health of the transitional age.
Collapse
Affiliation(s)
- M Bonomi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149, Milan, Italy.
| | - B Cangiano
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149, Milan, Italy
| | - S Cianfarani
- Endocrinology and Diabetes Unit, Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - A Garolla
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy
| | - D Gianfrilli
- Section of Medical Pathophysiology and Endocrinology, Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
- Centre for Rare Diseases (Endo-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - F Lanfranco
- Division of Endocrinology, Andrology and Metabolism, Department of Medical Sciences, Humanitas Gradenigo, University of Turin, Turin, Italy
| | - G Rastrelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Careggi Hospital, University of Florence, Florence, Italy
| | - E Sbardella
- Section of Medical Pathophysiology and Endocrinology, Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
- Centre for Rare Diseases (Endo-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - G Corona
- Endocrinology Unit, Medical Department, Maggiore-Bellaria Hospital, Azienda Usl, Bologna, Italy
| | - A M Isidori
- Section of Medical Pathophysiology and Endocrinology, Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
- Centre for Rare Diseases (Endo-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - V Rochira
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria Di Modena Policlinico Di Modena, Ospedale Civile Di Baggiovara, Via Giardini 1355, 41126, Modena, Italy.
| |
Collapse
|
20
|
Poch A, Dougherty MP, Roman RA, Chorich L, Hawkins Z, Kim SH, Kim HG, Layman LC. Prevalence of pathogenic variants and digenic disease in patients diagnosed with normosmic hypogonadotropic hypogonadism/Kallmann Syndrome. Mol Cell Endocrinol 2024; 589:112224. [PMID: 38593951 DOI: 10.1016/j.mce.2024.112224] [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: 02/02/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Hypogonadotropic hypogonadism (HH) is due to impaired gonadotropin releasing hormone (GnRH) action resulting in absent puberty and infertility. At least 44 genes have been identified to possess genetic variants in 40-50% of nHH/KS, and 2-20% have presumed digenic disease, but not all variants have been characterized in vitro. HYPOTHESIS The prevalence of pathogenic (P)/likely pathogenic (LP) variants in monogenic and digenic nHH/KS is lower than reported. DESIGN Cross-sectional study. SETTING University Research Laboratory. SUBJECTS 158 patients with nHH/KS. METHODS Exome sequencing (ES) was performed and variants were filtered for 44 known genes using Varsome and confirmed by Sanger Sequencing. MAIN OUTCOME MEASURES P/LP variants in nHH/KS genes. RESULTS ES resulted in >370,000 variants, from which variants in 44 genes were filtered. Thirty-one confirmed P/LP variants in 10 genes (ANOS1, CHD7, DUSP6, FGFR1, HS6ST1, KISS1, PROKR2, SEMA3A, SEMA3E, TACR3), sufficient to cause disease, were identified in 30/158 (19%) patients. Only 2/158 (1.2%) patients had digenic variant combinations: a male with hemizygous ANOS1 and heterozygous TACR3 variants and a male with heterozygous SEMA3A and SEMA3E variants. Two patients (1.2%) had compound heterozygous GNRHR (autosomal recessive) variants-one P and one variant of uncertain significance (VUS). Five patients (3.2%) had heterozygous P/LP variants in either GNRHR or TACR3 (both autosomal recessive), but no second variant. CONCLUSION Our prevalence of P/LP variants in nHH/KS was 19%, and digenicity was observed in 1.2%. These findings are less than those previously reported, and probably represent a more accurate estimation since VUS are not included.
Collapse
Affiliation(s)
- Alexandra Poch
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Michael P Dougherty
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Robert A Roman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Lynn Chorich
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Zoe Hawkins
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Soo-Hyun Kim
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, United Kingdom
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Center, Hamad Bin Khalifa University, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Lawrence C Layman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Medical College of Georgia at Augusta University, Augusta, GA, USA; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
21
|
Ali G, Shin KC, Ahmed N, Habbab W, Alkhadairi G, Razzaq A, Bejaoui Y, El Hajj N, Mifsud B, Park Y, Stanton LW. Deletion in RMST lncRNA impairs hypothalamic neuronal development in a human stem cell-based model of Kallmann Syndrome. Cell Death Discov 2024; 10:330. [PMID: 39030180 PMCID: PMC11271498 DOI: 10.1038/s41420-024-02074-4] [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: 02/29/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 07/21/2024] Open
Abstract
Rhabdomyosarcoma 2-associated transcript (RMST) long non-coding RNA has previously been shown to cause Kallmann syndrome (KS), a rare genetic disorder characterized by congenital hypogonadotropic hypogonadism (CHH) and olfactory dysfunction. In the present study, we generated large deletions of approximately 41.55 kb in the RMST gene in human pluripotent stem cells using CRISPR/Cas9 gene editing. To evaluate the impact of RMST deletion, these cells were differentiated into hypothalamic neurons that include 10-15% neurons that express gonadotrophin-releasing hormone (GnRH). We found that deletion in RMST did not impair the neurogenesis of GnRH neurons, however, the hypothalamic neurons were electro-physiologically hyperactive and had increased calcium influx activity compared to control. Transcriptomic and epigenetic analyses showed that RMST deletion caused altered expression of key genes involved in neuronal development, ion channels, synaptic signaling and cell adhesion. The in vitro generation of these RMST-deleted GnRH neurons provides an excellent cell-based model to dissect the molecular mechanism of RMST function in Kallmann syndrome and its role in hypothalamic neuronal development.
Collapse
Affiliation(s)
- Gowher Ali
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad, Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Kyung Chul Shin
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad, Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Nisar Ahmed
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Wesal Habbab
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad, Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Ghaneya Alkhadairi
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad, Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Aleem Razzaq
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Yosra Bejaoui
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Nady El Hajj
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
- College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Borbala Mifsud
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
- William Harvey Research Institute, Queen Mary University London, London, UK
| | - Yongsoo Park
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad, Bin Khalifa University, Qatar Foundation, Doha, Qatar
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Lawrence W Stanton
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad, Bin Khalifa University, Qatar Foundation, Doha, Qatar.
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.
| |
Collapse
|
22
|
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
|
23
|
Kałużna M, Budny B, Rabijewski M, Dubiel A, Trofimiuk-Müldner M, Szutkowski K, Piotrowski A, Wrotkowska E, Hubalewska-Dydejczyk A, Ruchała M, Ziemnicka K. Variety of genetic defects in GnRH and hypothalamic-pituitary signaling and development in normosmic patients with IHH. Front Endocrinol (Lausanne) 2024; 15:1396805. [PMID: 39010903 PMCID: PMC11246878 DOI: 10.3389/fendo.2024.1396805] [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: 03/06/2024] [Accepted: 05/27/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction Normosmic isolated hypogonadotropic hypogonadism (nIHH) is a clinically and genetically heterogeneous disorder. Deleterious variants in over 50 genes have been implicated in the etiology of IHH, which also indicates a possible role of digenicity and oligogenicity. Both classes of genes controlling GnRH neuron migration/development and hypothalamic/pituitary signaling and development are strongly implicated in nIHH pathogenesis. The study aimed to investigate the genetic background of nIHH and further expand the genotype-phenotype correlation. Methods A total of 67 patients with nIHH were enrolled in the study. NGS technology and a 38-gene panel were applied. Results Causative defects regarded as at least one pathogenic/likely pathogenic (P/LP) variant were found in 23 patients (34%). For another 30 individuals, variants of unknown significance (VUS) or benign (B) were evidenced (45%). The most frequently mutated genes presenting P/LP alterations were GNRHR (n = 5), TACR3 (n = 3), and CHD7, FGFR1, NSMF, BMP4, and NROB1 (n = 2 each). Monogenic variants with solid clinical significance (P/LP) were observed in 15% of subjects, whereas oligogenic defects were detected in 19% of patients. Regarding recurrence, 17 novel pathogenic variants affecting 10 genes were identified for 17 patients. The most recurrent pathogenic change was GNRHR:p.Arg139His, detected in four unrelated subjects. Another interesting observation is that P/LP defects were found more often in genes related to hypothalamic-pituitary pathways than those related to GnRH. Conclusions The growing importance of the neuroendocrine pathway and related genes is drawing increasing attention to nIHH. However, the underestimated potential of VUS variants in IHH etiology, particularly those presenting recurrence, should be further elucidated.
Collapse
Affiliation(s)
- Małgorzata Kałużna
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Bartłomiej Budny
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Rabijewski
- Department of Reproductive Health, Centre for Postgraduate Medical Education, Warsaw, Poland
| | - Agnieszka Dubiel
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | | | - Kosma Szutkowski
- NanoBioMedical Centre at Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Adam Piotrowski
- Department of Biomedical Physics at Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Elżbieta Wrotkowska
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Ziemnicka
- Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
24
|
Handelsman DJ, Idan A, Desai R, Grainger J, Goebel C, Sleiman S, Savkovic S, Kouzios D, Jayadev V, Conway AJ. Single and multi-dose pharmacology of recombinant and urinary human chorionic gonadotrophin in men. Clin Endocrinol (Oxf) 2024; 101:42-50. [PMID: 38446525 DOI: 10.1111/cen.15040] [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: 01/17/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVE Human choriogonadotrophin (hCG) treatment of gonadotrophin-deficient infertile men uses hCG of urinary (uhCG) or recombinant (rhCG) origin, but these treatments have not been compared nor are there studies defining rhCG dosing in men. DESIGN hCG products were studied in randomized cross-over single-dose studies of standard (Study 1, 1500 IU and 62.5 µg, respectively) or high (Study 2, 5000 IU and 250 µg) dose and a multi-dose population pharmacology study of hCG use. PARTICIPANTS Eight (Study 1) and seven (Study 2) volunteers in cross-over and 52 gonadotrophin-deficient men in the multi-dose study MEASUREMENTS: In cross-over studies, serum testosterone (T), dihydrotestosterone (DHT) and estradiol by liquid chromatography-mass spectrometry (LCMS) and serum hCG, LH, FSH, SHBG and T (observational study) by immunoassays. RESULTS After standard and high-dose injection, serum hCG and testosterone responses had similar timing and peak concentrations except for a mildly lower early (<48 h) serum testosterone with uhCG. In the multi-dosing study, both hCGs had similar pharmacokinetics (pooled half-life 5.8 days, p < .001), while serum testosterone concentrations were stable after injection and did not differ between hCG products. Bench testing verified that 20% of pens from 4/10 individuals were used inappropriately. CONCLUSIONS Although hCG pharmacokinetics are not formally bioequivalent, the similar pharmacodynamic effects on serum testosterone indicate that at the doses tested both hCGs provide comparable clinical effects. The starting dose of rhCG for treating gonadotrophin-deficient men should be 62.5 µg (6 clicks) of the rhCG pen.
Collapse
Affiliation(s)
- David J Handelsman
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
- Andrology Department, Concord Hospital, Sydney, New South Wales, Australia
| | - Amanda Idan
- Andrology Department, Concord Hospital, Sydney, New South Wales, Australia
| | - Reena Desai
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Janelle Grainger
- Australian Sports Drug Testing Laboratory, National Measurement Institute, North Ryde, New South Wales, Australia
| | - Catrin Goebel
- Australian Sports Drug Testing Laboratory, National Measurement Institute, North Ryde, New South Wales, Australia
| | - Sue Sleiman
- Andrology Department, Concord Hospital, Sydney, New South Wales, Australia
| | - Sasha Savkovic
- Andrology Department, Concord Hospital, Sydney, New South Wales, Australia
| | - Dorothy Kouzios
- Diagnostic Pathology Unit, NSW Health Pathology, Concord Hospital, New South Wales, Australia
| | - Venna Jayadev
- Andrology Department, Concord Hospital, Sydney, New South Wales, Australia
| | - Ann J Conway
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
- Andrology Department, Concord Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Castets S, Albarel F, Bachelot A, Brun G, Bouligand J, Briet C, Bui Quoc E, Cazabat L, Chabbert-Buffet N, Christin-Maitre S, Courtillot C, Cuny T, De Filippo G, Donadille B, Illouz F, Pellegrini I, Reznik Y, Saveanu A, Teissier N, Touraine P, Vantyghem MC, Vergier J, Léger J, Brue T, Reynaud R. Position statement on the diagnosis and management of congenital pituitary deficiency in adults: The French National Diagnosis and Treatment Protocol (NDTP). ANNALES D'ENDOCRINOLOGIE 2024; 85:327-339. [PMID: 38452869 DOI: 10.1016/j.ando.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Pituitary deficiency, or hypopituitarism, is a rare chronic disease. It is defined by insufficient synthesis of one or more pituitary hormones (growth hormone, TSH, ACTH, LH-FSH, prolactin), whether or not associated with arginine vasopressin deficiency (formerly known as diabetes insipidus). In adult patients, it is usually acquired (notably during childhood), but can also be congenital, due to abnormal pituitary development. The present study focuses on congenital pituitary deficiency in adults, from diagnosis to follow-up, including special situations such as pregnancy or the elderly. The clinical presentation is highly variable, ranging from isolated deficit to multiple deficits, which may be part of a syndromic form or not. Diagnosis is based on a combination of clinical, biological (assessment of all hormonal axes), radiological (brain and hypothalamic-pituitary MRI) and genetic factors. Treatment consists in hormonal replacement therapy, adapted according to the period of life and the deficits, which may be progressive. Comorbidities, risk of complications and acute decompensation, and the impact on fertility and quality of life all require adaptative multidisciplinary care and long-term monitoring.
Collapse
Affiliation(s)
- Sarah Castets
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France.
| | - Frédérique Albarel
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Conception, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Anne Bachelot
- IE3M, ICAN, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Centre de Référence des Pathologies Gynécologiques Rares, hôpital Pitié-Salpêtrière, AP-HP, Paris, France; Sorbonne université, Paris, France
| | - Gilles Brun
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Hôpital Européen, Pôle imagerie médicale, 13003, Marseille, France
| | - Jérôme Bouligand
- Molecular Genetic, Pharmacogenetic and Hormonology, Kremlin-Bicêtre Hospital, Paris-Saclay University, AP-HP, Le Kremlin-Bicêtre, France
| | - Claire Briet
- Département d'endocrinologie-diabétologie nutrition, Centre de référence des maladies rares de la Thyroïde et des Récepteurs Hormonaux, Endo-ERN centre for rare endocrine diseases, CHU d'Angers, 4, rue larrey, 49100 Angers, France; Laboratoire MITOVASC, UMR CNRS 6015, Inserm 1083, Université d'Angers, rue Roger Amsler, 49100 Angers, France
| | - Emmanuelle Bui Quoc
- Ophthalmology Department, Robert-Debré University Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Laure Cazabat
- Department of Endocrinology, Diabetology and Nutrition, Ambroise Paré Hospital, AP-HP, UVSQ, Boulogne-Billancourt, France
| | - Nathalie Chabbert-Buffet
- Department of Gynecology and Obstetrics, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, 75020 Paris, France
| | - Sophie Christin-Maitre
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Carine Courtillot
- IE3M, ICAN, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Centre de Référence des Pathologies Gynécologiques Rares, hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Thomas Cuny
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Gianpaolo De Filippo
- Service d'endocrinologie et diabétologie pédiatrique, centre de référence des maladies endocriniennes de la croissance et du développement, hôpital universitaire Robert-Debré, université Paris Cité, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Bruno Donadille
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Frédéric Illouz
- Département d'endocrinologie-diabétologie nutrition, Centre de référence des maladies rares de la Thyroïde et des Récepteurs Hormonaux, Endo-ERN centre for rare endocrine diseases, CHU d'Angers, 4, rue larrey, 49100 Angers, France; Laboratoire MITOVASC, UMR CNRS 6015, Inserm 1083, Université d'Angers, rue Roger Amsler, 49100 Angers, France
| | - Isabelle Pellegrini
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Conception, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Yves Reznik
- Endocrinology and Diabetes Department, CHU Côte de Nacre and Unicaen, Caen Cedex, France
| | - Alexandru Saveanu
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France
| | - Natacha Teissier
- Department of Pediatric Otolaryngology, Robert Debré Hospital, AP-HP Nord, Paris, France
| | - Philippe Touraine
- Service d'endocrinologie et médecine de la reproduction, centre de maladies endocrinennes rares de la croissance et du développement, médecine-hôpital Pitié-Salpêtrière, Sorbonne université, Paris, France
| | - Marie-Christine Vantyghem
- Service d'endocrinologie, diabétologie et maladies métaboliques, CHRU de Lille, rue Polonowski, Lille cedex, France
| | - Julia Vergier
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Julianne Léger
- Service d'endocrinologie et diabétologie pédiatrique, centre de référence des maladies endocriniennes de la croissance et du développement, hôpital universitaire Robert-Debré, université Paris Cité, Assistance publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1141, Paris, France
| | - Thierry Brue
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France; Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Inserm, MMG, Laboratory of Molecular Biology, Hospital La Conception, Aix-Marseille University, AP-HM, Marseille, France
| | - Rachel Reynaud
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France; Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Inserm, MMG, Laboratory of Molecular Biology, Hospital La Conception, Aix-Marseille University, AP-HM, Marseille, France
| |
Collapse
|
26
|
Rhys-Evans S, Howard SR. Combined gonadotropin therapy to replace mini-puberty in male infants with congenital hypogonadotropic hypogonadism. Ann N Y Acad Sci 2024; 1537:32-40. [PMID: 38924109 DOI: 10.1111/nyas.15177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Infants born with severe central disorders of the hypothalamic-pituitary-gonadal axis leading to gonadotropin deficiency not only lack pubertal development in adolescence, but also lack infantile mini-puberty. This period of mini-puberty, where infants have gonadotropin and sex steroid concentrations up into the adult range, is vital for future reproductive capacity, particularly in boys. At present, there is no consensus on the diagnosis or management of infants with gonadotropin deficiency due to congenital hypogonadotropic hypogonadism or multiple pituitary hormone deficiency. Case series suggest that gonadotropin treatment in male infants with absent mini-puberty is effective in promoting both testicular descent in those with undescended testes and also facilitating increased penile size. Moreover, replacement with follicle-stimulating hormone increases the testicular Sertoli cell population, measurable as an increase in testicular volume and inhibin B, thus hypothetically increasing the capacity for spermatogenesis in adult life for these patients. However, long-term follow-up data is limited for both outcomes pertaining to fertility and nonreproductive sequelae, including neurodevelopment and psychological well-being. The use of international registries for patients with gonadotropin deficiency is a key element in the collection of high-quality, geographically widespread data to inform best-practice management from birth to adulthood.
Collapse
Affiliation(s)
- Sophie Rhys-Evans
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Paediatric Endocrinology, Barts Health NHS Trust, London, UK
| |
Collapse
|
27
|
Yang YF, Ma HL, Wang X, Nie M, Mao JF, Wu XY. Clinical manifestations and spermatogenesis outcomes in Chinese patients with congenital hypogonadotropic hypogonadism caused by inherited or de novo FGFR1 mutations. Asian J Androl 2024; 26:426-432. [PMID: 38227553 PMCID: PMC11280213 DOI: 10.4103/aja202366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 11/02/2023] [Indexed: 01/18/2024] Open
Abstract
Fibroblast growth factor receptor 1 ( FGFR1 ) mutations are associated with congenital hypogonadotropic hypogonadism (CHH) through inheritance or spontaneous occurrence. We detected FGFR1 mutations in a Chinese cohort of 210 CHH patients at Peking Union Medical College Hospital (Beijing, China) using next-generation and Sanger sequencing. We assessed missense variant pathogenicity using six bioinformatics tools and compared clinical features and treatment outcomes between inherited and de novo mutation groups. Among 19 patients with FGFR1 mutations, three were recurrent, and 16 were novel variants. Sixteen of the novel mutations were likely pathogenic according to the American College of Medical Genetics and Genomics (ACMG) guidelines, with the prevalent P366L variant. The majority of FGFR1 mutations was inherited (57.9%), with frameshift mutations exclusive to the de novo mutation group. The inherited mutation group had a lower incidence of cryptorchidism, short stature, and skeletal deformities. In the inherited mutation group, luteinizing hormone (LH) levels were 0.5 IU l -1 , follicle-stimulating hormone (FSH) levels were 1.0 IU l -1 , and testosterone levels were 1.3 nmol l -1 . In contrast, the de novo group had LH levels of 0.2 IU l -1 , FSH levels of 0.5 IU l -1 , and testosterone levels of 0.9 nmol l -1 , indicating milder hypothalamus-pituitary-gonadal axis (HPGA) functional deficiency in the inherited group. The inherited mutation group showed a tendency toward higher spermatogenesis rates. In conclusion, this study underscores the predominance of inherited FGFR1 mutations and their association with milder HPGA dysfunction compared to de novo mutations, contributing to our understanding of the genetic and clinical aspects of FGFR1 mutations.
Collapse
Affiliation(s)
- Yu-Fan Yang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Hai-Lu Ma
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Xi Wang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Min Nie
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Jiang-Feng Mao
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Xue-Yan Wu
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
28
|
Rey RA, Bergadá I, Ballerini MG, Braslavsky D, Chiesa A, Freire A, Grinspon RP, Keselman A, Arcari A. Diagnosing and treating anterior pituitary hormone deficiency in pediatric patients. Rev Endocr Metab Disord 2024; 25:555-573. [PMID: 38112850 DOI: 10.1007/s11154-023-09868-4] [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] [Accepted: 12/12/2023] [Indexed: 12/21/2023]
Abstract
Hypopituitarism, or the failure to secrete hormones produced by the anterior pituitary (adenohypophysis) and/or to release hormones from the posterior pituitary (neurohypophysis), can be congenital or acquired. When more than one pituitary hormone axis is impaired, the condition is known as combined pituitary hormone deficiency (CPHD). The deficiency may be primarily due to a hypothalamic or to a pituitary disorder, or concomitantly both, and has a negative impact on target organ function. This review focuses on the pathophysiology, diagnosis and management of anterior pituitary hormone deficiency in the pediatric age. Congenital hypopituitarism is generally due to genetic disorders and requires early medical attention. Exposure to toxicants or intrauterine infections should also be considered as potential etiologies. The molecular mechanisms underlying the fetal development of the hypothalamus and the pituitary are well characterized, and variants in the genes involved therein may explain the pathophysiology of congenital hypopituitarism: mutations in the genes expressed in the earliest stages are usually associated with syndromic forms whereas variants in genes involved in later stages of pituitary development result in non-syndromic forms with more specific hormone deficiencies. Tumors or lesions of the (peri)sellar region, cranial radiation therapy, traumatic brain injury and, more rarely, other inflammatory or infectious lesions represent the etiologies of acquired hypopituitarism. Hormone replacement is the general strategy, with critical periods of postnatal life requiring specific attention.
Collapse
Affiliation(s)
- Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina.
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| | - María Gabriela Ballerini
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| | - Débora Braslavsky
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| | - Ana Chiesa
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| | - Analía Freire
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| | - Romina P Grinspon
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| | - Ana Keselman
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| | - Andrea Arcari
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, C1425EFD, Argentina
| |
Collapse
|
29
|
Zhan T, Zhang J, Zhang Y, Zhao Q, Chemerinski A, Douglas NC, Zhang Q, Xiao S. A Dose-Response Study on Functional and Transcriptomic Effects of FSH on Ex Vivo Mouse Folliculogenesis. Endocrinology 2024; 165:bqae054. [PMID: 38735763 PMCID: PMC11129714 DOI: 10.1210/endocr/bqae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Follicle-stimulating hormone (FSH) binds to its membrane receptor (FSHR) in granulosa cells to activate various signal transduction pathways and drive the gonadotropin-dependent phase of folliculogenesis. Both FSH insufficiency (due to genetic or nongenetic factors) and FSH excess (as encountered with ovarian stimulation in assisted reproductive technology [ART]) can cause poor female reproductive outcomes, but the underlying molecular mechanisms remain elusive. Herein, we conducted single-follicle and single-oocyte RNA sequencing analysis along with other approaches in an ex vivo mouse folliculogenesis and oogenesis system to investigate the effects of different concentrations of FSH on key follicular events. Our study revealed that a minimum FSH threshold is required for follicle maturation into the high estradiol-secreting preovulatory stage, and such threshold is moderately variable among individual follicles between 5 and 10 mIU/mL. FSH at 5, 10, 20, and 30 mIU/mL induced distinct expression patterns of follicle maturation-related genes, follicular transcriptomics, and follicular cAMP levels. RNA sequencing analysis identified FSH-stimulated activation of G proteins and downstream canonical and novel signaling pathways that may critically regulate follicle maturation, including the cAMP/PKA/CREB, PI3K/AKT/FOXO1, and glycolysis pathways. High FSH at 20 and 30 mIU/mL resulted in noncanonical FSH responses, including premature luteinization, high production of androgen and proinflammatory factors, and reduced expression of energy metabolism-related genes in oocytes. Together, this study improves our understanding of gonadotropin-dependent folliculogenesis and provides crucial insights into how high doses of FSH used in ART may impact follicular health, oocyte quality, pregnancy outcome, and systemic health.
Collapse
Affiliation(s)
- Tingjie Zhan
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| | - Jiyang Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| | - Ying Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| | - Qingshi Zhao
- Department of Obstetrics, Gynecology and Reproductive Health, New Jersey Medical School (NJMS), Rutgers University, Newark, NJ 07103, USA
| | - Anat Chemerinski
- Department of Obstetrics, Gynecology and Reproductive Health, New Jersey Medical School (NJMS), Rutgers University, Newark, NJ 07103, USA
| | - Nataki C Douglas
- Department of Obstetrics, Gynecology and Reproductive Health, New Jersey Medical School (NJMS), Rutgers University, Newark, NJ 07103, USA
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ 07103, USA
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
30
|
Huang Z, Wang X, Yu B, Ma W, Zhang P, Wu X, Nie M, Mao J. Pulsatile gonadotropin releasing hormone therapy for spermatogenesis in congenital hypogonadotropic hypogonadism patients who had poor response to combined gonadotropin therapy. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230101. [PMID: 38739523 PMCID: PMC11156179 DOI: 10.20945/2359-4292-2023-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 09/12/2023] [Indexed: 05/16/2024]
Abstract
Objective Both pulsatile gonadotropin-releasing hormone (GnRH) and combined gonadotropin therapy are effective to induce spermatogenesis in men with congenital hypogonadotropic hypogonadism (CHH). This study aimed to evaluate the effect of pulsatile GnRH therapy on spermatogenesis in male patients with CHH who had poor response to combined gonadotropin therapy. Materials and methods Patients who had poor response to combined gonadotropin therapy ≥ 6 months were recruited and shifted to pulsatile GnRH therapy. The rate of successful spermatogenesis, the median time to achieve spermatogenesis, serum gonadotropins, testosterone, and testicular volume were used for data analysis. Results A total of 28 CHH patients who had poor response to combined gonadotropin (HCG/HMG) therapy for 12.5 (6.0, 17.75) months were recruited and switched to pulsatile GnRH therapy for 10.0 (7.25, 16.0) months. Sperm was detected in 17/28 patients (60.7%). The mean time for the appearance of sperm in semen was 12.0 (7.5, 17.5) months. Compared to those who could not achieve spermatogenesis during pulsatile GnRH therapy, the successful group had a higher level of LH60min (4.32 vs. 1.10 IU/L, P = 0.043) and FSH60min (4.28 vs. 1.90 IU/L, P = 0.021). Testicular size increased during pulsatile GnRH therapy, compared to previous HCG/ HMG therapy (P < 0.05). Conclusion For CHH patients with prior poor response to one year of HCG/ HMG therapy, switching to pulsatile GnRH therapy may induce spermatogenesis.
Collapse
Affiliation(s)
- Zhenxing Huang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning China
- The first two authors contributed equally to this work
| | - Xi Wang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- The first two authors contributed equally to this work
| | - Bingqing Yu
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wanlu Ma
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Pengyu Zhang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueyan Wu
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Min Nie
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiangfeng Mao
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China,
| |
Collapse
|
31
|
Chang E, Shi YF, Liu JF, Wei W. Post-marketing safety concerns with elagolix: a disproportionality analysis of the FDA adverse event reporting system. Expert Opin Drug Saf 2024:1-8. [PMID: 38700323 DOI: 10.1080/14740338.2024.2351451] [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: 11/17/2023] [Accepted: 03/01/2024] [Indexed: 05/05/2024]
Abstract
OBJECTIVE Elagolix is approved for the treatment of moderate-to-severe pain associated with endometriosis. However, the long-term safety of elagolix in a large sample of real-world patients is unknown. METHODS The U.S. Food and Drug Administration Adverse Event Reporting System (FAERS) reports were collected and analyzed from January 2019 to June 2023. Disproportionality analyses, including the reporting odds ratio (ROR), the proportional reporting ratio (PRR), the Bayesian confidence propagation neural network (BCPNN), and the multi-item gamma Poisson shrinker (MGPS) algorithms, were employed in data mining to quantify the signals of elagolix-related adverse events (AEs). RESULTS After removing the non-drug-related AE signals, we detected several AE signals such as hot flushes, bone pain, suicidal ideation, depression, and increased liver enzymes, which were known during the clinical trial phase. In addition to this, we detected several unexpected important AEs that were not mentioned in the drug insert, including cystitis interstitial, parosmia, and epiploic appendagitis. The median onset time of elagolix-associated AEs was 28.5 days. CONCLUSION Our study provides a comprehensive picture of the safety of elagolix in the post-marketing setting, while also identifying potential new AE signals. These findings emphasize the importance of continued monitoring of the potential risks of elagolix.
Collapse
Affiliation(s)
- En Chang
- Department of Pharmacy, People's Hospital of Zhongjiang County, Deyang, Sichuan, China
| | - Yong-Fang Shi
- College of Pharmacy, Heze University, Heze, Shandong, China
| | - Jin-Feng Liu
- Department of Pharmacy, People's Hospital of Zhongjiang County, Deyang, Sichuan, China
| | - Wei Wei
- Department of Pharmacy, People's Hospital of Zhongjiang County, Deyang, Sichuan, China
| |
Collapse
|
32
|
Balen AH, Tamblyn J, Skorupskaite K, Munro MG. A comprehensive review of the new FIGO classification of ovulatory disorders. Hum Reprod Update 2024; 30:355-382. [PMID: 38412452 DOI: 10.1093/humupd/dmae003] [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: 06/12/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND The World Health Organization (WHO) system for the classification of disorders of ovulation was produced 50 years ago and, by international consensus, has been updated by the International Federation of Gynecology and Obstetrics (FIGO). OBJECTIVE AND RATIONALE This review outlines in detail each component of the FIGO HyPO-P (hypothalamic, pituitary, ovarian, PCOS) classification with a concise description of each cause, and thereby provides a systematic method for diagnosis and management. SEARCH METHODS We searched the published articles in the PubMed database in the English-language literature until October 2022, containing the keywords ovulatory disorders; ovulatory dysfunction; anovulation, and each subheading in the FIGO HyPO-P classification. We did not include abstracts or conference proceedings because the data are usually difficult to assess. OUTCOMES We present the most comprehensive review of all disorders of ovulation, published systematically according to the logical FIGO classification. WIDER IMPLICATIONS Improving the diagnosis of an individual's ovulatory dysfunction will significantly impact clinical practice by enabling healthcare practitioners to make a precise diagnosis and plan appropriate management.
Collapse
Affiliation(s)
- Adam H Balen
- Leeds Centre for Reproductive Medicine, The University of Leeds, Leeds, UK
| | - Jennifer Tamblyn
- Leeds Centre for Reproductive Medicine, The University of Leeds, Leeds, UK
| | | | - Malcolm G Munro
- Department of Obstetrics and Gynecology, The University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
33
|
Santi D, Spaggiari G, Marinelli L, Cacciani M, Scipio S, Bichiri A, Profeta A, Granata ARM, Simoni M, Lanfranco F, Manieri C, Ghigo E, Motta G. Gender-affirming hormone treatment: friend or foe? Long-term follow-up of 755 transgender people. J Endocrinol Invest 2024; 47:1091-1100. [PMID: 37889433 DOI: 10.1007/s40618-023-02220-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/07/2023] [Indexed: 10/28/2023]
Abstract
PURPOSE Gender-affirming hormone treatment (GAHT) is one of the main demands of transgender and gender diverse (TGD) people, who are usually categorised as transgender assigned-male-at birth (AMAB) and assigned-female-at birth (AFAB). The aim of the study is to investigate the long-term therapeutic management of GAHT, considering hormonal targets, treatment adjustments and GAHT safety. METHODS A retrospective, longitudinal, observational, multicentre clinical study was carried out. Transgender people, both AMAB and AFAB, were recruited from two Endocrinology Units in Italy (Turin and Modena) between 2005 and 2022. Each subject was managed with specific and personalized follow-up depending on the clinical practice of the Centre. All clinical data routinely collected were extracted, including anthropometric and biochemical parameters, lifestyle habits, GAHT regime, and cardiovascular events. RESULTS Three-hundred and two transgender AFAB and 453 transgender AMAB were included. Similar follow-up duration (p = 0.974) and visits' number (p = 0.384) were detected between groups. The transgender AFAB group reached therapeutic goals in less time (p = 0.002), fewer visits (p = 0.006) and fewer adjustments of GAHT scheme (p = 0.024). Accordingly, transgender AFAB showed a higher adherence to medical prescriptions compared to transgender AMAB people (p < 0.001). No significantly increased rate of cardiovascular events was detected in both groups. CONCLUSION Our real-world clinical study shows that transgender AFAB achieve hormone target earlier and more frequently in comparison to transgender AMAB individuals. Therefore, transgender AMAB people may require more frequent check-ups in order to tailor feminizing GAHT and increase therapeutic adherence.
Collapse
Affiliation(s)
- D Santi
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy.
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy.
| | - G Spaggiari
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - L Marinelli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - M Cacciani
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - S Scipio
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - A Bichiri
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - A Profeta
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - A R M Granata
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - M Simoni
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - F Lanfranco
- Division of Endocrinology, Andrology and Metabolism, Department of Medical Sciences, Humanitas Gradenigo, University of Turin, Turin, Italy
| | - C Manieri
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - E Ghigo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - G Motta
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| |
Collapse
|
34
|
Lockie AWC, Grice P, Mathur R, Pearce I, Modgil V. Diagnosis and treatment of hypogonadism in men seeking to preserve fertility - what are the options? Int J Impot Res 2024:10.1038/s41443-024-00897-4. [PMID: 38693209 DOI: 10.1038/s41443-024-00897-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Male hypogonadism is a clinical syndrome that results in low testosterone levels and frequently leads to infertility. The syndrome occurs due to disruption at one or more levels of the hypothalamic-pituitary-gonadal axis. Testosterone replacement therapy (TRT) is the most common treatment utilised for male hypogonadism. However, long-acting forms of TRT leads to infertility and so is inappropriate for patients wishing to conceive. For patients who wish to remain fertile, nasal TRT, clomiphene citrate, exogenous gonadotropins, gonadotropin releasing hormone and aromatase inhibitors have been used as alternative treatment options with different degrees of success. A review of the literature was performed to identify the safety and efficacy of alternative treatment options. Gonadotropin releasing hormone can successfully induce spermatogenesis but is impractical to administer. Likewise, aromatase inhibitors have limited use due to inducing osteopenia. Nasal TRT may be a good treatment option for these patients, but its efficacy has so far only been demonstrated in small sample sizes. However, clomiphene citrate and exogenous gonadotropins are safe, offer good symptom control and can successfully induce fertility in hypogonadism patients.
Collapse
Affiliation(s)
| | - Peter Grice
- Northampton General Hospital, Northampton, UK
| | - Raj Mathur
- Manchester Royal Infirmary, Manchester, UK
| | - Ian Pearce
- Manchester Royal Infirmary, Manchester, UK
| | | |
Collapse
|
35
|
de Silva NL, Dissanayake H, Kalra S, Meeran K, Somasundaram NP, Jayasena CN. Global Barriers to Accessing Off-Patent Endocrine Therapies: A Renaissance of the Orphan Disease? J Clin Endocrinol Metab 2024; 109:e1379-e1388. [PMID: 37846800 PMCID: PMC11031238 DOI: 10.1210/clinem/dgad610] [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: 01/12/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
CONTEXT Clinical endocrinology encompasses many diseases requiring long-term drug therapy. Prohibitive pricing of some endocrine drugs classified as essential by the World Health Organization has created suboptimal care of patients with endocrine disorders. EVIDENCE ACQUISITION This review is based on evidence obtained from several databases and search engines including PubMed, Google, and Google Scholar; reference searches; manual searching for web pages of international regulatory bodies; and the authors' experience from different healthcare settings. EVIDENCE SYNTHESIS After the expiry of a patent, generic versions with the opportunity for increased availability and a price reduction are expected. There are access barriers worldwide for many off-patent endocrine drugs. The high price is the main issue for several medicines including insulin, hydrocortisone, testosterone, and gonadotropins. This is caused by several factors including the market monopoly due to the lack of registered generics or suppliers limiting the benefit of competition and a complex supply chain. Additionally, the lack of some medicines has been concerning due to market factors such as the relatively small number of patients, making it less attractive for the manufacturers. Commissioning of nonprofit manufacturers and state manufacturing as well as strict price control measures could alleviate this situation. CONCLUSION Lack of availability and disproportionate price inflation affecting essential off-patent endocrine therapies is common due to several interrelated factors. Global collaboration among healthcare organizations with the support of policymaking bodies might be needed to mitigate this.
Collapse
Affiliation(s)
- Nipun Lakshitha de Silva
- Department of Clinical Sciences, Faculty of Medicine, General Sir John Kotelawala Defence University, Ratmalana 10390, Sri Lanka
| | - Harsha Dissanayake
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo 00800, Sri Lanka
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, Haryana 132001, India
- University Centre for Research and Development, Chandigarh University, Mohali 140413, India
| | - Karim Meeran
- Department of Metabolism, Digestion and Reproduction, Imperial College, W12 0NN, London, UK
| | | | - Channa N Jayasena
- Department of Metabolism, Digestion and Reproduction, Imperial College, W12 0NN, London, UK
| |
Collapse
|
36
|
Guo Q, Wang Y, Yu L, Guan L, Ji X, Li X, Pang G, Ren Z, Ye L, Cheng H. Nicotine restores olfactory function by activation of prok2R/Akt/FoxO3a axis in Parkinson's disease. J Transl Med 2024; 22:350. [PMID: 38609979 PMCID: PMC11015622 DOI: 10.1186/s12967-024-05171-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Olfactory dysfunction occurs frequently in Parkinson's disease (PD). In this study, we aimed to explore the potential biomarkers and underlying molecular pathways of nicotine for the treatment of olfactory dysfunction in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced PD mice. METHODS MPTP was introduced into C57BL/6 male mice to generate a PD model. Regarding in vivo experiments, we performed behavioral tests to estimate the protective effects of nicotine in MPTP-induced PD mice. RNA sequencing and traditional molecular methods were used to identify molecules, pathways, and biological processes in the olfactory bulb of PD mouse models. Then, in vitro experiments were conducted to evaluate whether nicotine can activate the prok2R/Akt/FoxO3a signaling pathway in both HEK293T cell lines and primary olfactory neurons treated with 1-methyl-4-phenylpyridinium (MPP+). Next, prok2R overexpression (prok2R+) and knockdown (prok2R-) were introduced with lentivirus, and the Akt/FoxO3a signaling pathway was further explored. Finally, the damaging effects of MPP+ were evaluated in prok2R overexpression (prok2R+) HEK293T cell lines. RESULTS Nicotine intervention significantly alleviated olfactory and motor dysfunctions in mice with PD. The prok2R/Akt/FoxO3a signaling pathway was activated after nicotine treatment. Consequently, apoptosis of olfactory sensory neurons was significantly reduced. Furthermore, prok2R+ and prok2R- HEK293T cell lines exhibited upregulation and downregulation of the Akt/FoxO3a signaling pathway, respectively. Additionally, prok2R+ HEK293T cells were resistant to MPP+-induced apoptosis. CONCLUSIONS This study showed the effectiveness and underlying mechanisms of nicotine in improving hyposmia in PD mice. These improvements were correlated with reduced apoptosis of olfactory sensory neurons via activated prok2R/Akt/FoxO3a axis. These results explained the potential protective functions of nicotine in PD patients.
Collapse
Affiliation(s)
- Qinglong Guo
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, Anhui, China
| | - Yi Wang
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, Anhui, China
| | - Liangchen Yu
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, Anhui, China
| | - Liao Guan
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, Anhui, China
| | - Xuefei Ji
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, Anhui, China
| | - Xiaohui Li
- Department of Anatomy, Anhui Medical University, Meishan Road 81, Hefei, 230032, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Meishan Road 81, Hefei, 230032, China
| | - Gang Pang
- Department of Anatomy, Anhui Medical University, Meishan Road 81, Hefei, 230032, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Meishan Road 81, Hefei, 230032, China
| | - Zhenhua Ren
- Department of Anatomy, Anhui Medical University, Meishan Road 81, Hefei, 230032, China.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Meishan Road 81, Hefei, 230032, China.
| | - Lei Ye
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, Anhui, China.
| | - Hongwei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, 230022, Anhui, China.
| |
Collapse
|
37
|
Zhang J, Yang S, Zhang Y, Liu F, Hao L, Han L. Clinical phenotype of a Kallmann syndrome patient with IL17RD and CPEB4 variants. Front Endocrinol (Lausanne) 2024; 15:1343977. [PMID: 38628584 PMCID: PMC11019388 DOI: 10.3389/fendo.2024.1343977] [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: 11/24/2023] [Accepted: 02/21/2024] [Indexed: 04/19/2024] Open
Abstract
Background This study aimed to characterize the clinical phenotype and genetic variations in patients with Kallmann syndrome (KS). Methods This study involved the collection and analysis of clinical data from an individual with sporadic KS. Following this, peripheral blood samples were obtained from the patient and his parents. Genomic deoxyribonucleic acid was extracted and subjected to whole-exome sequencing and genomic copy number variation (CNV) detection. Finally, Sanger sequencing was performed to validate the suspected pathogenic variants. Results Whole-exome sequencing confirmed that the child carried both the IL17RD variant (c.2101G>A, p.Gly701Ser) inherited from the mother and the new CPEB4 variant (c.1414C>T, p.Arg472*). No pathogenic CNVs were identified in CNV testing. Conclusion Bioinformatics analysis shows that the IL17RD protein undergoing Gly701Ser mutation and is speculated to be phosphorylated and modified, thereby disrupting fibroblast growth factor signaling. This study also suggested that the CPEB4 might play a crucial role in the key signaling process affecting olfactory bulb morphogenesis. Overall, the findings of this study broaden the gene expression profile of KS-related pathogenic genes. This offers a new avenue for exploring the pathogenic mechanism of KS and provides valuable insights for precise clinical diagnosis and treatment strategies for this condition.
Collapse
Affiliation(s)
- Jianmei Zhang
- Department of Pediatric Endocrinology and Genetics, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, China
| | - Suhong Yang
- Department of Pediatric Endocrinology and Genetics, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, China
| | - Yan Zhang
- Department of Pediatric Endocrinology and Genetics, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, China
| | - Fei Liu
- Department of Pediatric Endocrinology and Genetics, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, China
| | - Lili Hao
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lianshu Han
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Dwyer AA, McDonald IR, Cangiano B, Giovanelli L, Maione L, Silveira LFG, Raivio T, Latronico AC, Young J, Quinton R, Bonomi M, Persani L, Seminara SB, Lee CS. Classes and predictors of reversal in male patients with congenital hypogonadotropic hypogonadism: a cross-sectional study of six international referral centres. Lancet Diabetes Endocrinol 2024; 12:257-266. [PMID: 38437850 PMCID: PMC10996025 DOI: 10.1016/s2213-8587(24)00028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Although some male patients with congenital hypogonadotropic hypogonadism (CHH) undergo spontaneous reversal following treatment, predictors of reversal remain elusive. We aimed to assemble the largest cohort of male patients with CHH reversal to date and identify distinct classes of reversal. METHODS This multicentre cross-sectional study was conducted in six international CHH referral centres in Brazil, Finland, France, Italy, the UK, and the USA. Adult men with CHH (ie, absent or incomplete spontaneous puberty by age 18 years, low serum testosterone concentrations, and no identifiable cause of hypothalamic-pituitary-gonadal [HPG] axis dysfunction) were eligible for inclusion. CHH reversal was defined as spontaneous recovery of HPG axis function off treatment. Centres provided common data elements on patient phenotype, clinical assessment, and genetics using a structured, harmonised data collection form developed by COST Action BM1105. Latent class mixture modelling (LCMM) was applied to establish whether at least two distinct classes of reversal could be identified and differentially predicted, and results were compared with a cohort of patients without CHH reversal to identify potential predictors of reversal. The primary outcome was the presence of at least two distinct classes of reversal. FINDINGS A total of 87 male patients with CHH reversal and 108 without CHH reversal were included in the analyses. LCMM identified two distinct reversal classes (75 [86%] in class 1 and 12 [14%] in class 2) on the basis of mean testicular volume, micropenis, and serum follicle-stimulating hormone (FSH) concentration. Classification probabilities were robust (0·998 for class 1 and 0·838 for class 2) and modelling uncertainty was low (entropy 0·90). Compared with class 1, patients in class 2 had significantly larger testicular volume (p<0·0001), no micropenis, and higher serum FSH concentrations (p=0·041), consistent with the Pasqualini syndrome (fertile eunuch) subtype of CHH. Patients without CHH reversal were more likely to have anosmia (p=0·016), cryptorchidism (p=0·0012), complete absence of puberty (testicular volume <4 cm³; p=0·0016), and two or more rare genetic variants (ie, oligogenicity; p=0·0001). Among patients who underwent genetic testing, no patients (of 75) with CHH reversal had a rare pathogenic ANOS1 variant compared with ten (11%) of 95 patients without CHH reversal. Individuals with CHH reversal had a significantly higher rate of rare variants in GNRHR than did those without reversal (nine [12%] of 75 vs three [3%] of 95; p=0·025). INTERPRETATION Applying LCMM to a large cohort of male patients with CHH reversal uncovered two distinct classes of reversal. Genetic investigation combined with careful clinical phenotyping could help surveillance of reversal after withdrawing treatment, representing the first tailored management approach for male patients with this rare endocrine disorder. FUNDING National Institutes of Health National Center for Advancing Translational Sciences; Ministry of Health, Rome, Italy; Ministry of University, Rome, Italy; National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development; and the Josiah Macy Jr Foundation. TRANSLATION For the Italian translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Andrew A Dwyer
- National Institute of Child Health and Human Development, P50 Massachusetts General Hospital Harvard Center for Reproductive Medicine, Boston, MA, USA; William F Connell School of Nursing, Boston College, Chestnut Hill, MA, USA.
| | | | - Biagio Cangiano
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Luca Giovanelli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy; Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals, Newcastle-upon-Tyne, UK
| | - Luigi Maione
- Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Inserm U 1185, Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre, France
| | - Leticia F G Silveira
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil; Serviço de Endocrinologia, Departamento de Clínica Médica da Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Taneli Raivio
- Children's Hospital, Pediatric Research Center, University of Helsinki-Helsinki University Hospital, Helsinki, Finland; Translational Stem Cell Biology and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ana Claudia Latronico
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Departamento de Clínica Médica, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Jacques Young
- Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Inserm U 1185, Physiologie et Physiopathologie Endocriniennes, Le Kremlin-Bicêtre, France
| | - Richard Quinton
- Department of Endocrinology, Diabetes & Metabolism, Newcastle-upon-Tyne Hospitals, Newcastle-upon-Tyne, UK; Translational & Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK; Department of Metabolism, Digestion & Reproduction, Imperial College London, London, UK
| | - Marco Bonomi
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Luca Persani
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Stephanie B Seminara
- National Institute of Child Health and Human Development, P50 Massachusetts General Hospital Harvard Center for Reproductive Medicine, Boston, MA, USA; Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher S Lee
- William F Connell School of Nursing, Boston College, Chestnut Hill, MA, USA
| |
Collapse
|
39
|
Esteves SC, Humaidan P, Ubaldi FM, Alviggi C, Antonio L, Barratt CLR, Behre HM, Jørgensen N, Pacey AA, Simoni M, Santi D. APHRODITE criteria: addressing male patients with hypogonadism and/or infertility owing to altered idiopathic testicular function. Reprod Biomed Online 2024; 48:103647. [PMID: 38367592 DOI: 10.1016/j.rbmo.2023.103647] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 02/19/2024]
Abstract
RESEARCH QUESTION Can a novel classification system of the infertile male - 'APHRODITE' (Addressing male Patients with Hypogonadism and/or infeRtility Owing to altereD, Idiopathic TEsticular function) - stratify different subgroups of male infertility to help scientists to design clinical trials on the hormonal treatment of male infertility, and clinicians to counsel and treat the endocrinological imbalances in men and, ultimately, increase the chances of natural and assisted conception? DESIGN A collaboration between andrologists, reproductive urologists and gynaecologists, with specialization in reproductive medicine and expertise in male infertility, led to the development of the APHRODITE criteria through an iterative consensus process based on clinical patient descriptions and the results of routine laboratory tests, including semen analysis and hormonal testing. RESULTS Five patient groups were delineated according to the APHRODITE criteria; (1) Hypogonadotrophic hypogonadism (acquired and congenital); (2) Idiopathic male infertility with lowered semen analysis parameters, normal serum FSH and normal serum total testosterone concentrations; (3) A hypogonadal state with lowered semen analysis parameters, normal FSH and reduced total testosterone concentrations; (4) Lowered semen analysis parameters, elevated FSH concentrations and reduced or normal total testosterone concentrations; and (5) Unexplained male infertility in the context of unexplained couple infertility. CONCLUSION The APHRODITE criteria offer a novel and standardized patient stratification system for male infertility independent of aetiology and/or altered spermatogenesis, facilitating communication among clinicians, researchers and patients to improve reproductive outcomes following hormonal therapy. APHRODITE is proposed as a basis for future trials of the hormonal treatment of male infertility.
Collapse
Affiliation(s)
- Sandro C Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Campinas, Brazil.; Department of Surgery (Division of Urology), University of Campinas (UNICAMP), Campinas, Brazil.; Faculty of Health, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark..
| | - Peter Humaidan
- Fertility Clinic at Skive Regional Hospital, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Filippo M Ubaldi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Carlo Alviggi
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Leen Antonio
- Department of Endocrinology, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | | | - Hermann M Behre
- Center for Reproductive Medicine and Andrology, University Medicine Halle, Halle, Germany
| | - Niels Jørgensen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Allan A Pacey
- Faculty of Biology, Medicine and Health, Core Technology Facility, University of Manchester, Manchester, UK
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.; Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.; Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.; Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.; Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| |
Collapse
|
40
|
Anawalt BD. Reversibility of congenital hypogonadotropic hypogonadism: lessons from a rare disease. Lancet Diabetes Endocrinol 2024; 12:219-221. [PMID: 38437851 DOI: 10.1016/s2213-8587(24)00065-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/06/2024]
Affiliation(s)
- Bradley D Anawalt
- University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Kung KTF, Louie K, Spencer D, Hines M. Prenatal androgen exposure and sex-typical play behaviour: A meta-analysis of classic congenital adrenal hyperplasia studies. Neurosci Biobehav Rev 2024; 159:105616. [PMID: 38447820 DOI: 10.1016/j.neubiorev.2024.105616] [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: 10/17/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
Thousands of non-human mammal experiments have demonstrated that early androgen exposure exerts long-lasting effects on neurobehavioural sexual differentiation. In humans, females with classic congenital adrenal hyperplasia (CAH) are exposed to unusually high concentrations of androgens prenatally, whereas prenatal concentrations of androgens in males with CAH are largely normal. The current meta-analysis included 20 independent samples and employed multi-level meta-analytic models. Consistently across all 7 male-typical and female-typical play outcomes, in the expected directions, the present study found significant and large average differences between control males and control females (gs = 0.83-2.78) as well as between females with CAH and control females (gs = 0.95-1.08), but differences between males with CAH and control males were mostly negligible and were non-significant for 6 of the 7 outcomes (gs = 0.04-0.27). These meta-analytic findings suggest that prenatal androgen exposure masculinises and defeminises play behaviour in humans. Broader implications in relation to sex chromosomes, brain development, oestrogens, socio-cognitive influences, other aspects of sex-related behavioural development, and gender nonconformity are discussed.
Collapse
Affiliation(s)
- Karson T F Kung
- Department of Psychology, Jockey Club Tower, Centennial Campus, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region.
| | - Krisya Louie
- Department of Psychology, Jockey Club Tower, Centennial Campus, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Debra Spencer
- Department of Psychology, University of Cambridge, Free School Lane, Cambridge CB2 3RQ, United Kingdom
| | - Melissa Hines
- Department of Psychology, University of Cambridge, Free School Lane, Cambridge CB2 3RQ, United Kingdom
| |
Collapse
|
42
|
Castets S, Thomas-Teinturier C, Villanueva C, Amsellem J, Barat P, Brun G, Quoc EB, Carel JC, De Filippo GP, Kipnis C, Martinerie L, Vergier J, Saveanu A, Teissier N, Coutant R, Léger J, Reynaud R. Diagnosis and management of congenital hypopituitarism in children. Arch Pediatr 2024; 31:165-171. [PMID: 38538470 DOI: 10.1016/j.arcped.2024.01.003] [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: 10/18/2023] [Revised: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 04/07/2024]
Abstract
Hypopituitarism (or pituitary deficiency) is a rare disease with an estimated prevalence of between 1/16,000 and 1/26,000 individuals, defined by insufficient production of one or several anterior pituitary hormones (growth hormone [GH], thyroid-stimulating hormone [TSH], adrenocorticotropic hormone [ACTH], luteinizing hormone [LH], follicle-stimulating hormone [FSH], prolactin), in association or not with diabetes insipidus (antidiuretic hormone [ADH] deficiency). While in adults hypopituitarism is mostly an acquired disease (tumors, irradiation), in children it is most often a congenital condition, due to abnormal pituitary development. Clinical symptoms vary considerably from isolated to combined deficiencies and between syndromic and non-syndromic forms. Early signs are non-specific but should not be overlooked. Diagnosis is based on a combination of clinical, laboratory (testing of all hormonal axes), imaging (brain magnetic resonance imaging [MRI] with thin slices centered on the hypothalamic-pituitary region), and genetic (next-generation sequencing of genes involved in pituitary development, array-based comparative genomic hybridization, and/or genomic analysis) findings. Early brain MRI is crucial in neonates or in cases of severe hormone deficiency for differential diagnosis and to inform syndrome workup. This article presents recommendations for hormone replacement therapy for each of the respective deficient axes. Lifelong follow-up with an endocrinologist is required, including in adulthood, with multidisciplinary management for patients with syndromic forms or comorbidities. Treatment objectives include alleviating symptoms, preventing comorbidities and acute complications, and optimal social and educational integration.
Collapse
Affiliation(s)
- Sarah Castets
- Assistance Publique Hôpitaux de Marseille, Hôpital la Timone, service de pédiatrie multidisciplinaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France.
| | - Cécile Thomas-Teinturier
- Assistance Publique-Hôpitaux de Paris, Université Paris-Saclay, Hôpital Bicêtre, service d'Endocrinologie et diabète de l'enfant, Le Kremlin Bicêtre, France; INSERM UMR 1018, Equipe d'épidémiologie des radiations, CESP, 94800 Villejuif, France
| | - Carine Villanueva
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, Bron, France
| | - Jessica Amsellem
- CHU Angers, Service d'endocrinologie diabétologie pédiatrique, Angers, France
| | - Pascal Barat
- Centre hospitalier universitaire de Bordeaux, unite d'endocrinologie pédiatrique, Bordeaux, France
| | - Gilles Brun
- Hôpital Européen, neuroradiologie, Marseille, France
| | - Emmanuel Bui Quoc
- Assistance Publique-Hôpitaux de Paris, Hôpital universitaire Robert Debré, service d'ophtalmologie, Paris, France
| | - Jean-Claude Carel
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1141, Paris, France
| | - Gian Paolo De Filippo
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France
| | - Clara Kipnis
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France
| | - Laetitia Martinerie
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France
| | - Julia Vergier
- Assistance Publique Hôpitaux de Marseille, Hôpital la Timone, service de pédiatrie multidisciplinaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France
| | - Alexandru Saveanu
- Assistance Publique Hôpitaux de Marseille, Hôpital Conception, laboratoire de bioloie moléculaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France; Aix Marseille Université, INSERM, MMG, U 1251, Marseille, France
| | - Natacha Teissier
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service de Chirurgie ORL et cervico-faciale pédiatrique, Paris, France
| | - Régis Coutant
- CHU Angers, Service d'endocrinologie diabétologie pédiatrique, Angers, France
| | - Juliane Léger
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1141, Paris, France
| | - Rachel Reynaud
- Assistance Publique Hôpitaux de Marseille, Hôpital la Timone, service de pédiatrie multidisciplinaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France; Aix Marseille Université, INSERM, MMG, U 1251, Marseille, France
| |
Collapse
|
43
|
Bhattacharya I, Sharma SS, Majumdar SS. Etiology of Male Infertility: an Update. Reprod Sci 2024; 31:942-965. [PMID: 38036863 DOI: 10.1007/s43032-023-01401-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023]
Abstract
Spermatogenesis is a complex process of germ cell division and differentiation that involves extensive cross-talk between the developing germ cells and the somatic testicular cells. Defective endocrine signaling and/or intrinsic defects within the testes can adversely affect spermatogenic progression, leading to subfertility/infertility. In recent years, male infertility has been recognized as a global public health concern, and research over the last few decades has elucidated the complex etiology of male infertility. Congenital reproductive abnormalities, genetic mutations, and endocrine/metabolic dysfunction have been demonstrated to be involved in infertility/subfertility in males. Furthermore, acquired factors like exposure to environmental toxicants and lifestyle-related disorders such as illicit use of psychoactive drugs have been shown to adversely affect spermatogenesis. Despite the large body of available scientific literature on the etiology of male infertility, a substantial proportion of infertility cases are idiopathic in nature, with no known cause. The inability to treat such idiopathic cases stems from poor knowledge about the complex regulation of spermatogenesis. Emerging scientific evidence indicates that defective functioning of testicular Sertoli cells (Sc) may be an underlying cause of infertility/subfertility in males. Sc plays an indispensable role in regulating spermatogenesis, and impaired functional maturation of Sc has been shown to affect fertility in animal models as well as humans, suggesting abnormal Sc as a potential underlying cause of reproductive insufficiency/failure in such cases of unexplained infertility. This review summarizes the major causes of infertility/subfertility in males, with an emphasis on infertility due to dysregulated Sc function.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, Central University of Kerala, Periye Campus, Kasaragod, 671320, Kerala, India.
| | - Souvik Sen Sharma
- National Institute of Animal Biotechnology, Hyderabad, 500 032, Telangana, India
| | - Subeer S Majumdar
- National Institute of Animal Biotechnology, Hyderabad, 500 032, Telangana, India.
- Gujarat Biotechnology University, Gandhinagar, GIFT City, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
44
|
Cotellessa L, Giacobini P. Role of Anti-Müllerian Hormone in the Central Regulation of Fertility. Semin Reprod Med 2024; 42:34-40. [PMID: 38608673 DOI: 10.1055/s-0044-1786050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
In recent years, the expanding roles of anti-Müllerian hormone (AMH) in various aspects of reproductive health have attracted significant attention. Initially recognized for its classical role in male sexual differentiation, AMH is produced postnatally by the Sertoli cells in the male testes and by the granulosa cells in the female ovaries. Traditionally, it was believed to primarily influence gonadal development and function. However, research over the last decade has unveiled novel actions of AMH beyond the gonads, specifically all along the hypothalamic-pituitary-gonadal axis. This review will focus on the emerging roles of AMH within the hypothalamus and discusses its potential implications in reproductive physiology. Additionally, recent preclinical and clinical studies have suggested that elevated levels of AMH may disrupt the hypothalamic network regulating reproduction, which could contribute to the central pathophysiology of polycystic ovary syndrome. These findings underscore the intricate interplay between AMH and the neuroendocrine system, offering new avenues for understanding the mechanisms underlying fertility and reproductive disorders.
Collapse
Affiliation(s)
- Ludovica Cotellessa
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| | - Paolo Giacobini
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| |
Collapse
|
45
|
Wojtas MN, Diaz-González M, Stavtseva N, Shoam Y, Verma P, Buberman A, Izhak I, Geva A, Basch R, Ouro A, Perez-Benitez L, Levy U, Borcel E, Nuñez Á, Venero C, Rotem-Dai N, Veksler-Lublinsky I, Knafo S. Interplay between hippocampal TACR3 and systemic testosterone in regulating anxiety-associated synaptic plasticity. Mol Psychiatry 2024; 29:686-703. [PMID: 38135756 PMCID: PMC11153148 DOI: 10.1038/s41380-023-02361-z] [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: 05/27/2023] [Revised: 11/17/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023]
Abstract
Tachykinin receptor 3 (TACR3) is a member of the tachykinin receptor family and falls within the rhodopsin subfamily. As a G protein-coupled receptor, it responds to neurokinin B (NKB), its high-affinity ligand. Dysfunctional TACR3 has been associated with pubertal failure and anxiety, yet the mechanisms underlying this remain unclear. Hence, we have investigated the relationship between TACR3 expression, anxiety, sex hormones, and synaptic plasticity in a rat model, which indicated that severe anxiety is linked to dampened TACR3 expression in the ventral hippocampus. TACR3 expression in female rats fluctuates during the estrous cycle, reflecting sensitivity to sex hormones. Indeed, in males, sexual development is associated with a substantial increase in hippocampal TACR3 expression, coinciding with elevated serum testosterone and a significant reduction in anxiety. TACR3 is predominantly expressed in the cell membrane, including the presynaptic compartment, and its modulation significantly influences synaptic activity. Inhibition of TACR3 activity provokes hyperactivation of CaMKII and enhanced AMPA receptor phosphorylation, associated with an increase in spine density. Using a multielectrode array, stronger cross-correlation of firing was evident among neurons following TACR3 inhibition, indicating enhanced connectivity. Deficient TACR3 activity in rats led to lower serum testosterone levels, as well as increased spine density and impaired long-term potentiation (LTP) in the dentate gyrus. Remarkably, aberrant expression of functional TACR3 in spines results in spine shrinkage and pruning, while expression of defective TACR3 increases spine density, size, and the magnitude of cross-correlation. The firing pattern in response to LTP induction was inadequate in neurons expressing defective TACR3, which could be rectified by treatment with testosterone. In conclusion, our study provides valuable insights into the intricate interplay between TACR3, sex hormones, anxiety, and synaptic plasticity. These findings highlight potential targets for therapeutic interventions to alleviate anxiety in individuals with TACR3 dysfunction and the implications of TACR3 in anxiety-related neural changes provide an avenue for future research in the field.
Collapse
Affiliation(s)
- Magdalena Natalia Wojtas
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Instituto Biofisika (UPV/EHU, CSIC), Departamento Biología Celular e Histología Facultad de Medicina y Enfermería, University of the Basque Country, Leioa, Spain
| | - Marta Diaz-González
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nadezhda Stavtseva
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yuval Shoam
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Poonam Verma
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Assaf Buberman
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Inbar Izhak
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Aria Geva
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Roi Basch
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alberto Ouro
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- NeuroAging Group Laboratory (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Centro de investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucia Perez-Benitez
- Instituto Biofisika (UPV/EHU, CSIC), Departamento Biología Celular e Histología Facultad de Medicina y Enfermería, University of the Basque Country, Leioa, Spain
| | - Uri Levy
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Erika Borcel
- Department of Psychobiology, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
- Department of Clinical Neuroscience, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Ángel Nuñez
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain
| | - Cesar Venero
- Department of Psychobiology, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Noa Rotem-Dai
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Isana Veksler-Lublinsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Shira Knafo
- Department of Physiology and Cell Biology, The National Institute for Biotechnology in the Negev, and the School of Brain Sciences and Cognition, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Instituto Biofisika (UPV/EHU, CSIC), Departamento Biología Celular e Histología Facultad de Medicina y Enfermería, University of the Basque Country, Leioa, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain.
| |
Collapse
|
46
|
He Z, Ouyang Q, Chen Q, Song Y, Hu J, Hu S, He H, Li L, Liu H, Wang J. Molecular mechanisms of hypothalamic-pituitary-ovarian/thyroid axis regulating age at first egg in geese. Poult Sci 2024; 103:103478. [PMID: 38295497 PMCID: PMC10844868 DOI: 10.1016/j.psj.2024.103478] [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: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Age at first egg (AFE) has consistently garnered interest as a crucial reproductive indicator within poultry production. Previous studies have elucidated the involvement of the hypothalamic-pituitary-ovarian (HPO) and hypothalamic-pituitary-thyroid (HPT) axes in regulating poultry sexual maturity. Concurrently, there was evidence suggesting a potential co-regulatory relationship between these 2 axes. However, as of now, no comprehensive exploration of the key pathways and genes responsible for the crosstalk between the HPO and HPT axes in the regulation of AFE has been reported. In this study, we conducted a comparative analysis of morphological differences and performed transcriptomic analysis on the hypothalamus, pituitary, thyroid, and ovarian stroma between normal laying group (NG) and abnormal laying group (AG). Morphological results showed that the thyroid index difference (D-) value (thyroid index D-value=right thyroid index-left thyroid index) was significantly (P < 0.05) lower in the NG than in the AG, while the ovarian index was significantly (P < 0.01) higher in the NG than in the AG. Furthermore, between NG and AG, we identified 99, 415, 167, and 1182 differentially expressed genes (DEGs) in the hypothalamus, pituitary, thyroid, and ovarian stroma, respectively. Gene ontology (GO) analysis highlighted that DEGs from 4 tissues were predominantly enriched in the "biological processes" category. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that 16, 14, 3, and 26 KEGG pathways were significantly enriched (P < 0.05) in the hypothalamus, pituitary, thyroid, and ovarian stroma. The MAPK signaling pathway emerged as the sole enriched pathway across all 4 tissues. Employing an integrated analysis of the protein-protein interaction (PPI) network and correlation analysis, we found GREB1 emerged as a pivotal component within the HPO axis to regulate estrogen-related signaling in the HPT axis, meanwhile, the HPT axis influenced ovarian development by regulating thyroid hormone-related signaling mainly through OPN5. Then, 10 potential candidate genes were identified, namely IGF1, JUN, ERBB4, KDR, PGF, FGFR1, GREB1, OPN5, DIO3, and THRB. These findings establish a foundation for elucidating the physiological and genetic mechanisms by which the HPO and HPT axes co-regulate goose AFE.
Collapse
Affiliation(s)
- Zhiyu He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Qingliang Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Yang Song
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China.
| |
Collapse
|
47
|
Zhang J, Zhu Y, Zhang R, Liu H, Sun B, Zhang W, Wang X, Nie M, Mao J, Wu X. Pulsatile Gonadotropin-Releasing Hormone Therapy Is Associated With Better Spermatogenic Outcomes than Gonadotropin Therapy in Patients With Pituitary Stalk Interruption Syndrome. Endocr Pract 2024; 30:146-154. [PMID: 38029930 DOI: 10.1016/j.eprac.2023.11.010] [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: 09/05/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVE To compare the effects of combined gonadotropin and pulsatile gonadotropin-releasing hormone (GnRH) therapy on spermatogenesis in patients with pituitary stalk interruption syndrome (PSIS). METHODS Male patients with PSIS (N = 119) were retrospectively studied. Patients received pulsatile GnRH therapy (N = 59) were divided into response and poor-response groups based on luteinizing hormone (LH) levels after 1-month treatment with a cutoff value of 1 or 2 IU/L. Participants with gonadotropin therapy were divided into human menopausal gonadotropin (hMG)/human chorionic gonadotropin (hCG) group (N = 60), and patients with pulsatile GnRH therapy were classified into GnRH group (N = 28) with treatment duration ≥6 months. RESULTS The overall success rates of spermatogenesis for hMG/hCG and GnRH therapy were 51.67% (31/60) vs 33.90% (20/59), respectively. GnRH group required a shorter period to induce spermatogenesis (8 vs 15 months, P = .019). hMG/hCG group had higher median total testosterone than GnRH group [2.16, interquartile range(IQR) 1.06-4.89 vs 1.31, IQR 0.21-2.26 ng/mL, P = .004]. GnRH therapy had a beneficial effect on spermatogenesis compared to hMG/hCG therapy (hazard ratio 1.97, 95% confidence interval 1.08-3.57, P = .026). In patients with pulsatile GnRH therapy, compared with the poor-response group, the response group had a higher successful spermatogenesis rate (5.00% vs 48.72%, P = .002) and higher median basal total testosterone (0.00, IQR 0.00-0.03 vs 0.04, IQR 0.00-0.16 ng/mL, P = .026) with LH = 1 IU/L as the cutoff value after 1-month pulsatile GnRH therapy. CONCLUSIONS Pulsatile GnRH therapy was superior to hMG/hCG therapy for spermatogenesis in patients with PSIS. Earlier spermatogenesis and higher concentrations of sperm could be obtained in the GnRH group if patients received therapy over 6 months.
Collapse
Affiliation(s)
- Junyi Zhang
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiyi Zhu
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Zhang
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongying Liu
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Bang Sun
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Zhang
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xi Wang
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Nie
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiangfeng Mao
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xueyan Wu
- National Health Commission (NHC) Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
48
|
Wyrwoll MJ, van der Heijden GW, Krausz C, Aston KI, Kliesch S, McLachlan R, Ramos L, Conrad DF, O'Bryan MK, Veltman JA, Tüttelmann F. Improved phenotypic classification of male infertility to promote discovery of genetic causes. Nat Rev Urol 2024; 21:91-101. [PMID: 37723288 DOI: 10.1038/s41585-023-00816-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/20/2023]
Abstract
An increasing number of genes are being described in the context of non-syndromic male infertility. Linking the underlying genetic causes of non-syndromic male infertility with clinical data from patients is important to establish new genotype-phenotype correlations. This process can be facilitated by using universal nomenclature, but no standardized vocabulary is available in the field of non-syndromic male infertility. The International Male Infertility Genomics Consortium aimed at filling this gap, providing a standardized vocabulary containing nomenclature based on the Human Phenotype Ontology (HPO). The "HPO tree" was substantially revised compared with the previous version and is based on the clinical work-up of infertile men, including physical examination and hormonal assessment. Some causes of male infertility can already be suspected based on the patient's clinical history, whereas in other instances, a testicular biopsy is needed for diagnosis. We assembled 49 HPO terms that are linked in a logical hierarchy and showed examples of morphological features of spermatozoa and testicular histology of infertile men with identified genetic diagnoses to describe the phenotypes. This work will help to record patients' phenotypes systematically and facilitate communication between geneticists and andrologists. Collaboration across institutions will improve the identification of patients with the same phenotypes, which will promote the discovery of novel genetic causes for non-syndromic male infertility.
Collapse
Affiliation(s)
- Margot J Wyrwoll
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | | | - Csilla Krausz
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, University Hospital of Careggi (AOUC), Florence, Italy
| | - Kenneth I Aston
- Andrology and IVF Laboratory, Department of Surgery (Urology), University of Utah, Salt Lake City, UT, USA
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, Department of Clinical and Surgical Andrology, University of Münster, Münster, Germany
| | - Robert McLachlan
- Department of Clinical Research, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Liliana Ramos
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Donald F Conrad
- Department of Genetics, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Joris A Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany.
| |
Collapse
|
49
|
Howard SR, Quinton R. Outcomes and experiences of adults with congenital hypogonadism can inform improvements in the management of delayed puberty. J Pediatr Endocrinol Metab 2024; 37:1-7. [PMID: 37997801 PMCID: PMC10775020 DOI: 10.1515/jpem-2023-0407] [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: 09/12/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023]
Abstract
Patients with congenital hypogonadism will encounter many health care professionals during their lives managing their health needs; from antenatal and infantile periods, through childhood and adolescence, into adult life and then old age. The pubertal transition from childhood to adult life raises particular challenges for diagnosis, therapy and psychological support, and patients encounter many pitfalls. Many patients with congenital hypogonadism and delayed or absent puberty are only diagnosed and treated after long diagnostic journeys, and their management across different centres and countries is not well standardised. Here we reconsider the management of pubertal delay, whilst addressing problematic diagnostic issues and highlighting the limitations of historic pubertal induction protocols - from the perspective of both an adult and a paediatric endocrinologist, dealing in our everyday work with the long-term adverse consequences to our hypogonadal patients of an incorrect and/or late diagnosis and treatment in childhood.
Collapse
Affiliation(s)
- Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Paediatric Endocrinology, Royal London Children’s Hospital, Barts Health NHS Trust, London, UK
| | - Richard Quinton
- Translational & Clinical Research Institute, University of Newcastle, Newcastle-upon-Tyne, UK
- Newcastle Hospitals NHS Trust, Newcastle-upon-Tyne, UK
| |
Collapse
|
50
|
Ichioka K, Yoshikawa T, Kimura H, Saito R. Additional mutation in PROKR2 and phenotypic differences in a Kallmann syndrome/normosmic congenital hypogonadotropic hypogonadism family carrying FGFR1 missense mutation. BMJ Case Rep 2024; 17:e258042. [PMID: 38272512 PMCID: PMC10826480 DOI: 10.1136/bcr-2023-258042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2024] [Indexed: 01/27/2024] Open
Abstract
Congenital hypogonadotropic hypogonadism (CHH) is a genetically and clinically diverse disorder encompassing Kallmann syndrome (KS) and normosmic CHH (nCHH). Although mutations in numerous genes account for nearly 50% of CHH cases, a significant portion remains genetically uncharacterized. While most mutations follow the traditional Mendelian inheritance patterns, evidence suggests oligogenic interactions between CHH genes, acting as modifier genes to explain variable expressivity and incomplete penetrance associated with certain mutations.In this study, the proband presented with nCHH, while his son exhibited KS. We employed whole-exome sequencing (WES) to investigate the genetic differences between the two, and Sanger sequencing was used to validate the results obtained from WES.Genetic analysis revealed that both the proband and his son harboured a mutation in FGFR1 gene. Notably, an additional rare mutation in PROKR2 gene was exclusively identified in the son, which suggests the cause of the phenotypic difference between KS and nCHH.
Collapse
Affiliation(s)
- Kentaro Ichioka
- Karasumaoike Branch, Ichioka Urological Clinic, Kyoto, Japan
| | | | - Hiroko Kimura
- Mens Fertility Clinic Tokyo, Ichioka Urological Clinic Tokyo Branch, Tokyo, Japan
| | - Ryoichi Saito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto-shi, Japan
| |
Collapse
|