1
|
Lee PH, Huang SC, Lee JC, Li SC, Tsai JW, Chang YM, Kao YC, Fan WL, Chang CD, Chen HC, Li CH, Hu CF, Liu TT, Wu PS, Nam MH, Yu SC, Wang JC, Huang HY. In-frame insertions of SOX10 are highly enriched and characterize a distinct transcriptomic profile in gastrointestinal schwannomas. J Pathol 2025. [PMID: 40272443 DOI: 10.1002/path.6426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/24/2025] [Accepted: 03/09/2025] [Indexed: 04/25/2025]
Abstract
Gastrointestinal schwannomas are molecularly and histologically distinct from their non-gastrointestinal counterparts, lacking NF2 alterations, although the primary drivers of these tumors are barely understood. A recent study has identified SOX10 in-frame insertions in schwannomas, particularly in intracranial non-vestibular lesions, whereas their role in gastrointestinal schwannomas remains unexplored. Whole exome sequencing of 15 gastrointestinal and two non-gastrointestinal schwannomas revealed recurrent SOX10 in-frame insertions in 14 gastrointestinal cases (93%) without other nerve sheath tumor-related alterations, such as NF2 mutations or SH3PXD2A::HTRA1 fusions (~14% in non-gastrointestinal cases). The prevalence, mutation spectrum, and specificity of SOX10 insertions were validated using Sanger sequencing in a large cohort comprising 61 gastrointestinal and 98 non-gastrointestinal schwannomas, as well as 110 non-schwannomatous mesenchymal and melanocytic neoplasms. SOX10 insertions, occurring within or near the high mobility group box domain, were significantly enriched in gastrointestinal schwannomas (91.8%) compared with non-gastrointestinal cases (5.1%). The most common insertion, p.Y173_Q174insKY, was present in 86.9% of gastrointestinal schwannomas but absent in non-gastrointestinal cases. Another recurrent insertion, p.P175_R176insKYQP, was rare and exclusively found in non-gastrointestinal schwannomas (3/98), while all non-schwannomatous controls were SOX10-normal. SOX10-inserted schwannomas exhibited histologic features characteristic of gastrointestinal schwannomas, including a microtrabecular arrangement of Schwann cells, peripheral lymphoid cuffs, and a lack of encapsulation. Both SOX10-inserted and SOX10-normal schwannomas demonstrated diffuse SOX10 immunoreactivity. The SOX10-inserted group was significantly associated with gastrointestinal locations (p < 0.001), older patients (p < 0.001), fusion negativity (p < 0.001), and larger tumor size (p = 0.013). Gene expression profiling of 44 cases revealed distinct transcriptomic profiles between primarily SOX10-inserted and SOX10-normal groups, with the latter group being classifiable into fusion-poor and fusion-enriched sub-clusters. This study highlights the genetic heterogeneity of schwannomas and suggests that SOX10 insertions play a pivotal role in the tumorigenesis of gastrointestinal schwannomas, distinctly separating them from non-gastrointestinal counterparts and contributing to their unique molecular profile. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Pei-Hang Lee
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Chiang Huang
- Department of Anatomic Pathology, Linkou Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jen-Chieh Lee
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Chou Li
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Dental Technology, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| | - Jen-Wei Tsai
- Department of Anatomic Pathology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yi-Ming Chang
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yu-Chien Kao
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Lang Fan
- Department of Medical Research, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ching-Di Chang
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hui-Chun Chen
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Hao Li
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Fa Hu
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ting-Ting Liu
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Laboratory Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pao-Shu Wu
- Department of Anatomical Pathology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Mann-Hua Nam
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shih-Chen Yu
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jui-Chu Wang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsuan-Ying Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Sun B, Stamou MI, Stockman SL, Campbell MB, Plummer L, Salnikov KB, Kotan LD, Topaloglu AK, Hisama FM, Davis EE, Seminara SB, Balasubramanian R. Expanding the Spectrum of Endocrine Abnormalities Associated With SOX11-related Disorders. J Clin Endocrinol Metab 2025; 110:1044-1052. [PMID: 39290158 DOI: 10.1210/clinem/dgae620] [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: 07/02/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
CONTEXT SOX11 variants cause Coffin-Siris syndrome, characterized by developmental delay, hypogonadotropic hypogonadism, and skeletal and facial defects. OBJECTIVE To examine the contribution of SOX11 variants to the pathogenesis of idiopathic hypogonadotropic hypogonadism (IHH), a disorder caused by hypothalamic GnRH deficiency. SETTING The Reproductive Endocrine Unit and the Pediatric Endocrinology Division, Massachusetts General Hospital. PATIENTS OR OTHER PARTICIPANTS A cohort of 1810 unrelated IHH probands. INTERVENTIONS Exome sequencing data from the entire cohort were examined for SOX11 rare single nucleotide variants (SNVs) (minor allele frequency in the gnomAD database <0.1%). Rare SOX11 variant association testing was performed between the IHH and gnomAD population. Phenotyping of individuals harboring pathogenic/likely pathogenic SNVs (determined by the American College of Medical Genetics criteria) was performed. MAIN OUTCOMES/RESULTS Four pathogenic SOX11 SNVs were identified in 5 IHH probands. The IHH cohort was enriched for SOX11 protein truncating SNVs (frameshift/nonsense) across the entire protein (2 SNVs in 3 IHH cases [p.S303X (de novo); p.S345Afs*13]; P = .0004981) and for SOX11 missense SNVs within the SOX11 high-mobility group domain (2 SNVs in 2 IHH cases p.G84D [de novo]; p.P114S; P = .00313922). The phenotypic spectrum of SOX11 variant carriers revealed additional endocrine defects including anosmic and normosmic forms of IHH, GH deficiency, pituitary and hypothalamic structural defects, and hypothyroidism. A pathogenic SOX11 SNV was also identified in a patient with functional hypogonadotropic hypogonadism (p.R100Q). Coffin-Siris syndrome-associated features were present in 4/5 probands. CONCLUSION Deleterious SOX11 variants cause IHH and other pituitary hormone deficiencies, suggesting that the human SOX11-associated disorder may stem from both hypothalamic and pituitary level defects.
Collapse
Affiliation(s)
- Bang Sun
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maria I Stamou
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sara L Stockman
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mark B Campbell
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lacey Plummer
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kathryn B Salnikov
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Leman Damla Kotan
- Division of Pediatric Endocrinology, Faculty of Medicine, Cukurova University, Adana, 01250, Turkey
| | - A Kemal Topaloglu
- Division of Pediatric Endocrinology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02113, USA
| | - Fuki M Hisama
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Erica E Davis
- Advanced Center for Translational and Genetic Medicine, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Pediatrics and Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Stephanie B Seminara
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ravikumar Balasubramanian
- Reproductive Endocrine Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
3
|
Taroc EZM, Amato E, Semon A, Dolphin N, Beck B, Belin S, Poitelon Y, Forni PE. Shared Lineage, Distinct Outcomes: Yap and Taz Loss Differentially Impact Schwann and Olfactory Ensheathing Cell Development Without Disrupting GnRH-1 Migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638196. [PMID: 40027653 PMCID: PMC11870449 DOI: 10.1101/2025.02.13.638196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Olfactory Ensheathing Cells (OECs) are glial cells originating from the neural crest, critical for bundling olfactory axons to the brain. Their development is crucial for the migration of Gonadotropin-Releasing Hormone-1 (GnRH-1) neurons, which are essential for puberty and fertility. OECs have garnered interest as potential therapeutic targets for central nervous system lesions, although their development is not fully understood. Our single-cell RNA sequencing of mouse embryonic nasal tissues suggests that OECs and Schwann cells share a common origin from Schwann cell precursors yet exhibit significant genetic differences. The transcription factors Yap and Taz have previously been shown to play a crucial role in Schwann cell development. We used Sox10 -Cre mice to conditionally ablate Yap and Taz in migrating the neural crest and its derivatives. Our analyses showed reduced Sox10+ glial cells along nerves in the nasal region, altered gene expression of SCs, melanocytes, and OECs, and a significant reduction in olfactory sensory neurons and vascularization in the vomeronasal organ. However, despite these changes, GnRH-1 neuronal migration remained unaffected. Our findings highlight the importance of the Hippo pathway in OEC development and how changes in cranial neural crest derivatives indirectly impact the development of olfactory epithelia.
Collapse
|
4
|
Read JE, Vasile‐Tudorache A, Newsome A, Lorente MJ, Agustín‐Pavón C, Howard SR. Disorders of puberty and neurodevelopment: A shared etiology? Ann N Y Acad Sci 2024; 1541:83-99. [PMID: 39431640 PMCID: PMC11580780 DOI: 10.1111/nyas.15246] [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: 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 InstituteQueen Mary University of LondonLondonUK
| | - Alexandru Vasile‐Tudorache
- Department of Cell Biology, Functional Biology and Physical AnthropologyFaculty of Biological Sciences, University of ValenciaValenciaSpain
| | - Angel Newsome
- Centre for Endocrinology, William Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - María José Lorente
- Department of Cell Biology, Functional Biology and Physical AnthropologyFaculty of Biological Sciences, University of ValenciaValenciaSpain
| | - Carmen Agustín‐Pavón
- Department of Cell Biology, Functional Biology and Physical AnthropologyFaculty of Biological Sciences, University of ValenciaValenciaSpain
| | - Sasha R. Howard
- Centre for Endocrinology, William Harvey Research InstituteQueen Mary University of LondonLondonUK
- Department of Paediatric EndocrinologyBarts Health NHS TrustLondonUK
| |
Collapse
|
5
|
Graziani L, Carriero ML, Pozzi F, Minotti C, Andreadi A, Bellia A, Ruta R, Bengala M, Novelli A, Lauro D, Novelli G. Genetic Variability of SOX10-Related Disorders within an Italian Family: Straddling the Line between Kallmann and Waardenburg Syndrome. Mol Syndromol 2024; 15:339-346. [PMID: 39119450 PMCID: PMC11305686 DOI: 10.1159/000536574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/31/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Kallmann syndrome (KS) is a genetically heterogeneous developmental disorder that most often manifests hypogonadotropic hypogonadism (HH) and hypo-/anosmia due to early embryonic impairment in the migration of gonadotropin-releasing hormone neurons. SOX10 (SRY-Box 10; MIM*602229), a key transcriptional activator involved in the development of neural crest cells, has been associated with KS and is identified as one of the causative genes of Waardenburg syndrome (WS). Case Presentation A 28-year-old female patient, who was clinically diagnosed with KS in her childhood, presented with HH and anosmia, mild bilateral sensorineural hearing loss (SNHL), and pigmentation abnormalities. Next-generation sequencing analysis detected a missense heterozygous SOX10 pathogenic variant (NM_006941.4:c.506C>T) in the proposita and in her mother, whose phenotype included exclusively anosmia and hypopigmented skin patches. The same variant has been described by Pingault et al. [Clin Genet. 2015;88(4):352-9] in a patient with apparently isolated bilateral severe SNHL. Conclusion Our finding substantiates the extreme phenotypic variability of SOX10-related disorders, which range from classical KS and/or WS to contracted endophenotypes that could share a common pathway in the development of neural crest cells and highlights the need for careful evaluation and long-term follow-up of SOX10 patients, with special focus on atypical/additional and/or late-onset phenotypic traits.
Collapse
Affiliation(s)
- Ludovico Graziani
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Miriam Lucia Carriero
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Flavio Pozzi
- Endocrinology and Diabetology Unit, Tor Vergata University Hospital, Rome, Italy
| | - Chiara Minotti
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Aikaterini Andreadi
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Alfonso Bellia
- Endocrinology and Diabetology Unit, Tor Vergata University Hospital, Rome, Italy
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Rosario Ruta
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Mario Bengala
- Medical Genetics Unit, Tor Vergata University Hospital, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Davide Lauro
- Endocrinology and Diabetology Unit, Tor Vergata University Hospital, Rome, Italy
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Medical Genetics Unit, Tor Vergata University Hospital, Rome, Italy
| |
Collapse
|
6
|
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
|
7
|
Chen F, Zhang Q, Zhang Q, Wang Q. [Research progress on hereditary endocrine and metabolic diseases associated with sensorineural hearing loss]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2024; 38:63-69. [PMID: 38297851 PMCID: PMC11116158 DOI: 10.13201/j.issn.2096-7993.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Indexed: 02/02/2024]
Abstract
Hereditary endocrine and metabolic diseases , caused by genetic factors, exhibit complex and diverse symptoms, including the possibility of concurrent sensorineural deafness. Currently, there is a limited clinical understanding of hereditary endocrine and metabolic diseases that manifest with deafness, the pathogenesis remains unclear,and there is a lack of effective diagnostic and treatment methods. This article summarizes the research progress of hereditary endocrine and metabolic diseases complicated with deafness from the pathogenesis, clinical phenotype, diagnosis and treatment. Understanding the current research progress and integrating genetic analysis into clinical practice are crucial for accurate diagnosis and treatment, evaluating clinical efficacy, and providing effective genetic counseling for these diseases.
Collapse
Affiliation(s)
- Fang Chen
- Traditional Chinese Medicine Endocrinology Department of the 985th Hospital of the Joint Logistics Support Force of the People's Liberation Army,Taiyuan,030001,China
| | - Qinying Zhang
- Traditional Chinese Medicine Endocrinology Department of the 985th Hospital of the Joint Logistics Support Force of the People's Liberation Army,Taiyuan,030001,China
| | - Qiujing Zhang
- Department of Audiology and Vestibular Medicine,Institute of Otolaryngology,Senior Department of Otolaryngology Head and Neck Surgery,the First Medical Center of Chinese PLA General Hospital,National Clinical Research Center for Otolaryngologic Diseases
| | - Qiuju Wang
- Department of Audiology and Vestibular Medicine,Institute of Otolaryngology,Senior Department of Otolaryngology Head and Neck Surgery,the First Medical Center of Chinese PLA General Hospital,National Clinical Research Center for Otolaryngologic Diseases
| |
Collapse
|
8
|
Murakami S, Ohki-Hamazaki H, Uchiyama Y. Somatostatin affects GnRH neuronal development and migration and stimulates olfactory-related fiber fasciculation. Dev Neurobiol 2024; 84:3-17. [PMID: 38072668 DOI: 10.1002/dneu.22931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 01/19/2024]
Abstract
Transient expression of somatostatin (SST) has been observed in the olfactory epithelium (OE) and nerves of chick embryos. Intense expression of SST in these regions on embryonic days (E) 5-8 coincides with the migration of neurons producing gonadotropin-releasing hormone (GnRH) from the OE to the forebrain (FB), suggesting that SST plays a role in the development of GnRH neurons. Using in ovo electroporation of small interfering RNA, we found that the suppression of SST mRNA in the olfactory placode (OP) of E3.5 chick embryos significantly reduced the number of GnRH and Islet-1-immunoreactive neurons in the nasal region without affecting the entry of GnRH neurons into the FB at E5.5-6. SST knockdown did not lead to changes in the number of apoptotic, proliferating, or HuC/D-positive neuronal cells in the OE; therefore, it is possible that SST is involved in the neurogenesis/differentiation of GnRH neurons and OP-derived GnRH-negative migratory neurons. In whole OP explant cultures, we also found that SST or its analog octreotide treatment significantly increased the number of migratory GnRH neurons and the migratory distance from the explants. The co-application of an SST antagonist blocked the octreotide-induced increase in the number of GnRH neurons. Furthermore, the fasciculation of polysialylated neural cell adhesion molecule-immunoreactive fibers emerging from the explants was dependent on octreotide. Taken together, our results provide evidence that SST exerts facilitatory effects on the development of neurons expressing GnRH or Islet-1 and on GnRH neuronal migration, in addition to olfactory-related fiber fasciculation.
Collapse
Affiliation(s)
- Shizuko Murakami
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroko Ohki-Hamazaki
- College of Liberal Arts and Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Bertani-Torres W, Lezirovitz K, Alencar-Coutinho D, Pardono E, da Costa SS, Antunes LDN, de Oliveira J, Otto PA, Pingault V, Mingroni-Netto RC. Waardenburg Syndrome: The Contribution of Next-Generation Sequencing to the Identification of Novel Causative Variants. Audiol Res 2023; 14:9-25. [PMID: 38391765 PMCID: PMC10886116 DOI: 10.3390/audiolres14010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/24/2024] Open
Abstract
Waardenburg syndrome (WS) is characterized by hearing loss and pigmentary abnormalities of the eyes, hair, and skin. The condition is genetically heterogeneous, and is classified into four clinical types differentiated by the presence of dystopia canthorum in type 1 and its absence in type 2. Additionally, limb musculoskeletal abnormalities and Hirschsprung disease differentiate types 3 and 4, respectively. Genes PAX3, MITF, SOX10, KITLG, EDNRB, and EDN3 are already known to be associated with WS. In WS, a certain degree of molecularly undetected patients remains, especially in type 2. This study aims to pinpoint causative variants using different NGS approaches in a cohort of 26 Brazilian probands with possible/probable diagnosis of WS1 (8) or WS2 (18). DNA from the patients was first analyzed by exome sequencing. Seven of these families were submitted to trio analysis. For inconclusive cases, we applied a targeted NGS panel targeting WS/neurocristopathies genes. Causative variants were detected in 20 of the 26 probands analyzed, these being five in PAX3, eight in MITF, two in SOX10, four in EDNRB, and one in ACTG1 (type 2 Baraitser-Winter syndrome, BWS2). In conclusion, in our cohort of patients, the detection rate of the causative variant was 77%, confirming the superior detection power of NGS in genetically heterogeneous diseases.
Collapse
Affiliation(s)
- William Bertani-Torres
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
- Department of Embryology and Genetics of Malformations, INSERM (Institut National de la Santé et de la Recherche Médicale) UMR (Unité Mixte de Recherche) 1163, Université Paris-Cité and Institut Imagine, 75015 Paris, France
| | - Karina Lezirovitz
- Otorhinolaryngology Lab-LIM 32, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Danillo Alencar-Coutinho
- Otorhinolaryngology Lab-LIM 32, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Eliete Pardono
- Instituto de Ciências da Saúde, Universidade Paulista UNIP, São Paulo 04026-002, Brazil
- Colégio Miguel de Cervantes, São Paulo 05618-001, Brazil
| | - Silvia Souza da Costa
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Larissa do Nascimento Antunes
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Judite de Oliveira
- Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Paulo Alberto Otto
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Véronique Pingault
- Department of Embryology and Genetics of Malformations, INSERM (Institut National de la Santé et de la Recherche Médicale) UMR (Unité Mixte de Recherche) 1163, Université Paris-Cité and Institut Imagine, 75015 Paris, France
- Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Regina Célia Mingroni-Netto
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| |
Collapse
|
10
|
Bahmad HF, Thiravialingam A, Sriganeshan K, Gonzalez J, Alvarez V, Ocejo S, Abreu AR, Avellan R, Arzola AH, Hachem S, Poppiti R. Clinical Significance of SOX10 Expression in Human Pathology. Curr Issues Mol Biol 2023; 45:10131-10158. [PMID: 38132479 PMCID: PMC10742133 DOI: 10.3390/cimb45120633] [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: 11/20/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
The embryonic development of neural crest cells and subsequent tissue differentiation are intricately regulated by specific transcription factors. Among these, SOX10, a member of the SOX gene family, stands out. Located on chromosome 22q13, the SOX10 gene encodes a transcription factor crucial for the differentiation, migration, and maintenance of tissues derived from neural crest cells. It plays a pivotal role in developing various tissues, including the central and peripheral nervous systems, melanocytes, chondrocytes, and odontoblasts. Mutations in SOX10 have been associated with congenital disorders such as Waardenburg-Shah Syndrome, PCWH syndrome, and Kallman syndrome, underscoring its clinical significance. Furthermore, SOX10 is implicated in neural and neuroectodermal tumors, such as melanoma, malignant peripheral nerve sheath tumors (MPNSTs), and schwannomas, influencing processes like proliferation, migration, and differentiation. In mesenchymal tumors, SOX10 expression serves as a valuable marker for distinguishing between different tumor types. Additionally, SOX10 has been identified in various epithelial neoplasms, including breast, ovarian, salivary gland, nasopharyngeal, and bladder cancers, presenting itself as a potential diagnostic and prognostic marker. However, despite these associations, further research is imperative to elucidate its precise role in these malignancies.
Collapse
Affiliation(s)
- Hisham F. Bahmad
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA;
| | - Aran Thiravialingam
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Karthik Sriganeshan
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Jeffrey Gonzalez
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Veronica Alvarez
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Stephanie Ocejo
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Alvaro R. Abreu
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Rima Avellan
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Alejandro H. Arzola
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (A.T.); (K.S.); (J.G.); (S.O.); (A.R.A.); (R.A.); (A.H.A.)
| | - Sana Hachem
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon;
| | - Robert Poppiti
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA;
- Department of Pathology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
11
|
Bertani-Torres W, Serey-Gaut M, de Oliveira J, Bole C, Parisot M, Nistschké P, Maurin ML, Lapierre JM, Loundon N, Belhous K, Bondurand N, Marlin S, Pingault V. A 22q13.1 duplication in mosaicism including SOX10. Am J Med Genet A 2023; 191:2813-2818. [PMID: 37533297 DOI: 10.1002/ajmg.a.63362] [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: 05/24/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023]
Abstract
Waardenburg syndrome (WS) is characterized by the association of sensorineural hearing loss and pigmentation abnormalities. Among the four types, WS Type 2 (WS2) is the only one without a remarkable distinguishing feature. Here, we report a patient initially diagnosed with WS2 who exhibits a 446 kb mosaic duplication in chromosome 22q13.1, encompassing SOX10, and detected using whole genome sequencing in a trio. The patient, a 46,XY boy, presents with profound bilateral sensorineural hearing loss, right heterochromia iridium, left bright blue iris, and skin-depigmented areas in the abdomen and limbs. Vestibular and imaging tests are normal, without inner ear or olfactory bulb malformations. Bilateral cochlear implantation did not prevent language and speech delays. Moderate congenital chronic constipation and neurodevelopmental difficulties were also present. Given the few genes included in this duplicated region (only one OMIM gene with dominant inheritance), this report provides further delineation of the phenotype related to duplications encompassing the entire SOX10 gene.
Collapse
Affiliation(s)
- William Bertani-Torres
- Université Paris Cité, Paris, France
- Embryologie et génétique des malformations, Institut Imagine, INSERM U1163, Paris, France
| | - Margaux Serey-Gaut
- Service de Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker, Paris, France
- Centre de Référence Surdités Génétiques, AP-HP, Hôpital Necker, Paris, France
| | - Judite de Oliveira
- Service de Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker, Paris, France
| | - Christine Bole
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Université Paris Cité, Paris, France
| | - Mélanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Université Paris Cité, Paris, France
| | - Patrick Nistschké
- Université Paris Cité, Paris, France
- Bioinformatics Platform, Institut Imagine, INSERM U1163, Paris, France
| | - Marie-Laure Maurin
- Service de Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker, Paris, France
| | - Jean-Michel Lapierre
- Service de Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker, Paris, France
| | - Natalie Loundon
- ENT Department, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Kahina Belhous
- Department of Radiology, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Nadège Bondurand
- Université Paris Cité, Paris, France
- Embryologie et génétique des malformations, Institut Imagine, INSERM U1163, Paris, France
| | - Sandrine Marlin
- Embryologie et génétique des malformations, Institut Imagine, INSERM U1163, Paris, France
- Service de Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker, Paris, France
- Centre de Référence Surdités Génétiques, AP-HP, Hôpital Necker, Paris, France
| | - Véronique Pingault
- Université Paris Cité, Paris, France
- Embryologie et génétique des malformations, Institut Imagine, INSERM U1163, Paris, France
- Service de Médecine Génomique des Maladies Rares, AP-HP, Hôpital Necker, Paris, France
| |
Collapse
|
12
|
Abstract
Pattern recognition of specific temporal bone radiological phenotypes, in association with abnormalities in other organ systems, is critical in the diagnosis and management of syndromic causes of hearing loss. Several recent publications have demonstrated the presence of specific radiological appearances, allowing precise genetic and/or syndromic diagnosis, in the right clinical context. This review article aims to provide an extensive but practical guide to the radiologist dealing with syndromic causes of hearing loss.
Collapse
Affiliation(s)
- Martin Lewis
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond St. London, London, WC1N3JH, UK
| | - Caroline D Robson
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Felice D'Arco
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond St. London, London, WC1N3JH, UK. felice.d'
| |
Collapse
|
13
|
He D, Sun H, Zhang M, Li Y, Liu F, Zhang Y, He M, Ban B. Clinical Manifestations, Genetic Variants and Therapeutic Evaluation in Sporadic Chinese Patients with Idiopathic Hypogonadotropic Hypogonadism. Int J Gen Med 2023; 16:4429-4439. [PMID: 37799300 PMCID: PMC10547821 DOI: 10.2147/ijgm.s430904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/18/2023] [Indexed: 10/07/2023] Open
Abstract
Purpose Genetic factors account for a large proportion of idiopathic hypogonadotropic hypogonadism (IHH) etiologies, although not necessarily a complete genetic basis. This study aimed to characterize the clinical presentations, genetic variants, and therapeutic outcomes of patients with sporadic IHH, which may be helpful for genetic counseling and treatment decisions. Patients and Methods Eleven Chinese patients with IHH were retrospectively analyzed. Rare genetic variants were evaluated using whole-exome sequencing and bioinformatics analysis and were further classified according to the ACMG-AMP guidelines. The therapeutic responses of patients were further evaluated. Results Six heterozygous variants of SOX10, WDR11, PROKR2, CHD7 and FGF17 were detected in five Kallmann syndrome (KS) patients, whereas two heterozygous variants of CHD7 and PROKR2 were detected in two normosmic IHH (nIHH) patients. Among these variants, a novel likely pathogenic variant in the SOX10 (c.429-1G>C) was considered to cause the KS phenotype in patient 02, and two potential variants of uncertain significance (VUS) in CHD7 (c.3344G>A and c.7391A>G) possibly contributed to the KS phenotype in patient 05 and the nIHH phenotype in patient 07, which need to be confirmed by further evidence. Additionally, long-term testosterone or estradiol replacement treatment effectively improved the development of sexual characteristics in patients with IHH. Conclusion Next-generation sequencing is a powerful tool for identifying the molecular etiology and early diagnosis of IHH. Efficient therapeutic outcomes strongly indicate a need for timely treatment.
Collapse
Affiliation(s)
- Dongye He
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
| | - Hailing Sun
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
| | - Mei Zhang
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
- Chinese Research Center for Behavior Medicine in Growth and Development, Jining, 272029, People’s Republic of China
| | - Yanying Li
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
- Chinese Research Center for Behavior Medicine in Growth and Development, Jining, 272029, People’s Republic of China
| | - Fupeng Liu
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
| | - Yanhong Zhang
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
- Chinese Research Center for Behavior Medicine in Growth and Development, Jining, 272029, People’s Republic of China
| | - Mingming He
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
| | - Bo Ban
- Department of Endocrinology, Genetics and Metabolism, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, People’s Republic of China
- Chinese Research Center for Behavior Medicine in Growth and Development, Jining, 272029, People’s Republic of China
| |
Collapse
|
14
|
Eskici N, Madhusudan S, Vaaralahti K, Yellapragada V, Gomez-Sanchez C, Kärkinen J, Almusa H, Brandstack N, Miettinen PJ, Wang Y, Raivio T. Congenital hypogonadotropic hypogonadism in a patient with a de novo POGZ mutation. Eur J Endocrinol 2023; 189:271-280. [PMID: 37619992 DOI: 10.1093/ejendo/lvad111] [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: 01/27/2023] [Revised: 06/28/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVE Congenital hypogonadotropic hypogonadism (CHH) is a rare, genetically heterogeneous reproductive disorder caused by gonadotropin-releasing hormone (GnRH) deficiency. Approximately half of CHH patients also have decreased or absent sense of smell, that is, Kallmann syndrome (KS). We describe a patient with White-Sutton syndrome (developmental delay and autism spectrum disorder) and KS due to a heterozygous de novo mutation in POGZ (c.2857C>T, p.(Gln953*)), a gene encoding pogo transposable element derived with zinc finger domain, which acts as a transcriptomic regulator of neuronal networks. DESIGN AND METHODS We modeled the role of POGZ in CHH by generating 2 clonal human pluripotent stem cell lines with CRISPR/Cas9, carrying either the heterozygous patient mutation (H11 line) or a homozygous mutation (c.2803-2906del; p.E935Kfs*7 encoding a truncated POGZ protein; F6del line). RESULTS During the differentiation to GnRH neurons, neural progenitors derived from F6del line displayed severe proliferation defect, delayed wound-healing capacity, downregulation of intermediate progenitor neuron genes TBR1 and TBR2, and immature neuron markers PAX6 and TUBB3 and gave rise to fewer neurons with shorter neurites and less neurite branch points compared to the WT and H11 lines (P < .005). Both lines, however, could be successfully differentiated to GnRH neurons. CONCLUSIONS In conclusion, this is the first report on the overlap between White-Sutton syndrome and CHH. POGZ mutations do not hinder GnRH neuron formation but may cause CHH/KS by affecting the size and motility of the anterior neural progenitor pool and neurite outgrowth.
Collapse
Affiliation(s)
- Nazli Eskici
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Shrinidhi Madhusudan
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Kirsi Vaaralahti
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Venkatram Yellapragada
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Celia Gomez-Sanchez
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Juho Kärkinen
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki 00014, Finland
| | - Nina Brandstack
- Department of Radiology, Helsinki University Hospital and University of Helsinki, Helsinki 00014, Finland
| | - Päivi J Miettinen
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
| | - Yafei Wang
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| | - Taneli Raivio
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
| |
Collapse
|
15
|
Al Sayed Y, Howard SR. Panel testing for the molecular genetic diagnosis of congenital hypogonadotropic hypogonadism – a clinical perspective. Eur J Hum Genet 2022; 31:387-394. [PMID: 36517585 PMCID: PMC10133250 DOI: 10.1038/s41431-022-01261-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/16/2022] Open
Abstract
AbstractCongenital hypogonadotropic hypogonadism (CHH) is a rare endocrine disorder that results in reproductive hormone deficiency and reduced potential for fertility in adult life. Discoveries of the genetic aetiology of CHH have advanced dramatically in the past 30 years, with currently over 40 genes recognised to cause or contribute to the development of this condition. The genetic complexity of CHH is further increased by the observation of di- and oligogenic, as well as classic monogenic, inheritance and incomplete penetrance. Very recently in the UK, a panel of 14 genes has been curated for the genetic diagnosis of CHH within the NHS Genomic Medicine Service programme. The aim of this review is to appraise the advantages and potential pitfalls of the use of a CHH panel in clinical endocrine diagnostics, and to consider the future avenues for developing this panel including the potential of whole exome or whole genome sequencing data analysis in this condition.
Collapse
|
16
|
Association between Synonymous SNPs of SOX10 and Plumage Color and Reproductive Traits of Ducks. Animals (Basel) 2022; 12:ani12233345. [PMID: 36496864 PMCID: PMC9736065 DOI: 10.3390/ani12233345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
Mutations in the SOX10 gene affect the plumage color of chickens and pigeons. The mutation also causes abnormal pigmentation of the skin and hair color, as well as postnatal growth retardation and reproduction problems in humans and mice. In this study, we investigated the association between the SOX10 gene and plumage color and reproductive traits of ducks using SNPs. We found six novel SNPs from 11 identified SNP sites using direct sequencing for PCR products from three different mixed DNA pools. We found two coding SNPs to be associated with the plumage color of ducks (ZJU1.0 Chr1. g.54065419C>T and g.54070844C>T), and found three coding SNPs associated with the reproductive traits of ducks (g.54065419C>T, g.54070844C>T, and g.54070904C>T), which were age at sexual maturity, body weight at sexual maturity, and the Haugh unit for egg quality traits and egg production in different productive periods. These results also indicated that the T alleles of the three SNPs of the coding region of SOX10 contribute to lower reproductive traits.
Collapse
|
17
|
Chachlaki K, Messina A, Delli V, Leysen V, Maurnyi C, Huber C, Ternier G, Skrapits K, Papadakis G, Shruti S, Kapanidou M, Cheng X, Acierno J, Rademaker J, Rasika S, Quinton R, Niedziela M, L'Allemand D, Pignatelli D, Dirlewander M, Lang-Muritano M, Kempf P, Catteau-Jonard S, Niederländer NJ, Ciofi P, Tena-Sempere M, Garthwaite J, Storme L, Avan P, Hrabovszky E, Carleton A, Santoni F, Giacobini P, Pitteloud N, Prevot V. NOS1 mutations cause hypogonadotropic hypogonadism with sensory and cognitive deficits that can be reversed in infantile mice. Sci Transl Med 2022; 14:eabh2369. [PMID: 36197968 PMCID: PMC7613826 DOI: 10.1126/scitranslmed.abh2369] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The nitric oxide (NO) signaling pathway in hypothalamic neurons plays a key role in the regulation of the secretion of gonadotropin-releasing hormone (GnRH), which is crucial for reproduction. We hypothesized that a disruption of neuronal NO synthase (NOS1) activity underlies some forms of hypogonadotropic hypogonadism. Whole-exome sequencing was performed on a cohort of 341 probands with congenital hypogonadotropic hypogonadism to identify ultrarare variants in NOS1. The activity of the identified NOS1 mutant proteins was assessed by their ability to promote nitrite and cGMP production in vitro. In addition, physiological and pharmacological characterization was carried out in a Nos1-deficient mouse model. We identified five heterozygous NOS1 loss-of-function mutations in six probands with congenital hypogonadotropic hypogonadism (2%), who displayed additional phenotypes including anosmia, hearing loss, and intellectual disability. NOS1 was found to be transiently expressed by GnRH neurons in the nose of both humans and mice, and Nos1 deficiency in mice resulted in dose-dependent defects in sexual maturation as well as in olfaction, hearing, and cognition. The pharmacological inhibition of NO production in postnatal mice revealed a critical time window during which Nos1 activity shaped minipuberty and sexual maturation. Inhaled NO treatment at minipuberty rescued both reproductive and behavioral phenotypes in Nos1-deficient mice. In summary, lack of NOS1 activity led to GnRH deficiency associated with sensory and intellectual comorbidities in humans and mice. NO treatment during minipuberty reversed deficits in sexual maturation, olfaction, and cognition in Nos1 mutant mice, suggesting a potential therapy for humans with NO deficiency.
Collapse
Affiliation(s)
- Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France.,Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland.,University Research Institute of Child Health and Precision Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens 115 27, Greece
| | - Andrea Messina
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Virginia Delli
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Valerie Leysen
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Csilla Maurnyi
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, 43 Szigony St., Budapest 1083, Hungary
| | - Chieko Huber
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, Geneva 1211, Switzerland
| | - Gaëtan Ternier
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, 43 Szigony St., Budapest 1083, Hungary
| | - Georgios Papadakis
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Sonal Shruti
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Maria Kapanidou
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
| | - Xu Cheng
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - James Acierno
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Jesse Rademaker
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Sowmyalakshmi Rasika
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Richard Quinton
- Translational and Clinical Research Institute and the Royal Victoria Infirmary, University of Newcastle , Tyne NE1 3BZ, UK
| | - Marek Niedziela
- Department of Paediatric Endocrinology and Rheumatology, Poznan University of Medical Sciences, Poznan 61-701, Poland
| | - Dagmar L'Allemand
- Department of Endocrinology, Children's Hospital of Eastern Switzerland, St. Gallen 9000, Switzerland
| | - Duarte Pignatelli
- Department of Endocrinology, Hospital S João; Department of Biomedicine, Faculty of Medicine of the University of Porto; IPATIMUP Research Institute, Porto 4200-319, Portugal
| | - Mirjam Dirlewander
- Pediatric Endocrine and Diabetes Unit, Children's Hospital, University Hospitals and Faculty of Medicine, Geneva CH1205, Switzerland
| | - Mariarosaria Lang-Muritano
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, Zürich 8032, Switzerland
| | - Patrick Kempf
- Department of Diabetes, Endocrinology, Clinical Nutrition and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Sophie Catteau-Jonard
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France.,Department of Gynaecology and Obstretic, Jeanne de Flandres Hospital, Centre Hospitalier Universitaire de Lille, Lille F-59000, France
| | - Nicolas J Niederländer
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Philippe Ciofi
- Inserm, U1215, Neurocentre Magendie, Université de Bordeaux, Bordeaux F-33077, France
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba 14004, Spain.,Instituto Maimonides de Investigación Biomédica de Cordoba (IMIBIC/HURS), Cordoba 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba 14004, Spain
| | - John Garthwaite
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6DH, UK
| | - Laurent Storme
- FHU 1000 Days for Health, School of Medicine, Lille F-59000, France.,Department of Neonatology, Hôpital Jeanne de Flandre, CHU of Lille, Lille F-59000, France
| | - Paul Avan
- Université de Clerremont-Ferrand, Clermont-Ferrand F-63000, France
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, 43 Szigony St., Budapest 1083, Hungary
| | - Alan Carleton
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, Geneva 1211, Switzerland
| | - Federico Santoni
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, Lille F-59000, France.,FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
| |
Collapse
|
18
|
Saengkaew T, Howard SR. Genetics of pubertal delay. Clin Endocrinol (Oxf) 2022; 97:473-482. [PMID: 34617615 PMCID: PMC9543006 DOI: 10.1111/cen.14606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
The timing of pubertal development is strongly influenced by the genetic background, and clinical presentations of delayed puberty are often found within families with clear patterns of inheritance. The discovery of the underlying genetic regulators of such conditions, in recent years through next generation sequencing, has advanced the understanding of the pathogenesis of disorders of pubertal timing and the potential for genetic testing to assist diagnosis for patients with these conditions. This review covers the significant advances in the understanding of the biological mechanisms of delayed puberty that have occurred in the last two decades.
Collapse
Affiliation(s)
- Tansit Saengkaew
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Endocrinology Unit, Department of Paediatrics, Faculty of MedicinePrince of Songkla UniversitySongkhlaThailand
| | - Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
19
|
Winters SJ. Hypogonadism in Males With Genetic Neurodevelopmental Syndromes. J Clin Endocrinol Metab 2022; 107:e3974-e3989. [PMID: 35913018 DOI: 10.1210/clinem/dgac421] [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: 04/07/2022] [Indexed: 11/19/2022]
Abstract
Genetic syndromes that affect the nervous system may also disrupt testicular function, and the mechanisms for these effects may be interrelated. Most often neurological signs and symptoms predominate and hypogonadism remains undetected and untreated, while in other cases, a thorough evaluation of a hypogonadal male reveals previously unrecognized ataxia, movement disorder, muscle weakness, tremor, or seizures, leading to a syndromic diagnosis. Androgen deficiency in patients with neurological diseases may aggravate muscle weakness and fatigue and predispose patients to osteoporosis and obesity. The purpose of this mini review is to provide a current understanding of the clinical, biochemical, histologic, and genetic features of syndromes in which male hypogonadism and neurological dysfunction may coexist and may be encountered by the clinical endocrinologist.
Collapse
Affiliation(s)
- Stephen J Winters
- Division of Endocrinology, Metabolism & Diabetes, University of Louisville, Louisville, KY, USA
| |
Collapse
|
20
|
Duittoz AH, Tillet Y, Geller S. The great migration: how glial cells could regulate GnRH neuron development and shape adult reproductive life. J Chem Neuroanat 2022; 125:102149. [PMID: 36058434 DOI: 10.1016/j.jchemneu.2022.102149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 10/31/2022]
Abstract
In mammals, reproductive function is under the control of hypothalamic neurons named Gonadotropin-Releasing Hormone (GnRH) neurons. These neurons migrate from the olfactory placode to the brain, during embryonic development. For the past 40 years, these neurons have been considered an example of tangential migration, i.e., dependent on the olfactory/vomeronasal/terminal nerves. Numerous studies have highlighted the factors involved in the migration of these neurons but thus far overlooked the cellular microenvironment that produces them. Many of these factors are dysregulated in hypogonadotropic hypogonadism, resulting in subfertility/infertility. Nevertheless, over the past ten years, several papers have reported the influence of glial cells (named olfactory ensheathing cells [OECs]) in the migration and differentiation of GnRH neurons. This review will describe the atypical origins, migration, and differentiation of these neurons, focusing on the latest discoveries. There will be a more specific discussion on the involvement of OECs in the development of GnRH neurons, during embryonic and perinatal life; as well as on their potential implication in the development of congenital or idiopathic hypogonadotropic hypogonadism (such as Kallmann syndrome).
Collapse
Affiliation(s)
- Anne H Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRA Val de Loire, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Yves Tillet
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRA Val de Loire, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Sarah Geller
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
21
|
Moon S, Zhao YT. Convergent biological pathways underlying the Kallmann syndrome-linked genes Hs6st1 and Fgfr1. Hum Mol Genet 2022; 31:4207-4216. [PMID: 35899427 PMCID: PMC9759331 DOI: 10.1093/hmg/ddac172] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/05/2022] [Accepted: 07/24/2022] [Indexed: 01/21/2023] Open
Abstract
Kallmann syndrome (KS) is a congenital disorder characterized by idiopathic hypogonadotropic hypogonadism and olfactory dysfunction. KS is linked to variants in >34 genes, which are scattered across the human genome and show disparate biological functions. Although the genetic basis of KS is well studied, the mechanisms by which disruptions of these diverse genes cause the same outcome of KS are not fully understood. Here we show that disruptions of KS-linked genes affect the same biological processes, indicating convergent molecular mechanisms underlying KS. We carried out machine learning-based predictions and found that KS-linked mutations in heparan sulfate 6-O-sulfotransferase 1 (HS6ST1) are likely loss-of-function mutations. We next disrupted Hs6st1 and another KS-linked gene, fibroblast growth factor receptor 1 (Fgfr1), in mouse neuronal cells and measured transcriptome changes using RNA sequencing. We found that disruptions of Hs6st1 and Fgfr1 altered genes in the same biological processes, including the upregulation of genes in extracellular pathways and the downregulation of genes in chromatin pathways. Moreover, we performed genomics and bioinformatics analyses and found that Hs6st1 and Fgfr1 regulate gene transcription likely via the transcription factor Sox9/Sox10 and the chromatin regulator Chd7, which are also associated with KS. Together, our results demonstrate how different KS-linked genes work coordinately in a convergent signaling pathway to regulate the same biological processes, thus providing new insights into KS.
Collapse
Affiliation(s)
- Sohyun Moon
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, USA
| | - Ying-Tao Zhao
- To whom correspondence should be addressed: Tel: 516-686-3764; Fax: 516-686-3832;
| |
Collapse
|
22
|
Prevot V, Sharif A. The polygamous GnRH neuron: Astrocytic and tanycytic communication with a neuroendocrine neuronal population. J Neuroendocrinol 2022; 34:e13104. [PMID: 35233849 DOI: 10.1111/jne.13104] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/12/2022] [Accepted: 01/30/2022] [Indexed: 11/28/2022]
Abstract
To ensure the survival of the species, hypothalamic neuroendocrine circuits controlling fertility, which converge onto neurons producing gonadotropin-releasing hormone (GnRH), must respond to fluctuating physiological conditions by undergoing rapid and reversible structural and functional changes. However, GnRH neurons do not act alone, but through reciprocal interactions with multiple hypothalamic cell populations, including several glial and endothelial cell types. For instance, it has long been known that in the hypothalamic median eminence, where GnRH axons terminate and release their neurohormone into the pituitary portal blood circulation, morphological plasticity displayed by distal processes of tanycytes modifies their relationship with adjacent neurons as well as the spatial properties of the neurohemal junction. These alterations not only regulate the capacity of GnRH neurons to release their neurohormone, but also the activation of discrete non-neuronal pathways that mediate feedback by peripheral hormones onto the hypothalamus. Additionally, a recent breakthrough has demonstrated that GnRH neurons themselves orchestrate the establishment of their neuroendocrine circuitry during postnatal development by recruiting an entourage of newborn astrocytes that escort them into adulthood and, via signalling through gliotransmitters such as prostaglandin E2, modulate their activity and GnRH release. Intriguingly, several environmental and behavioural toxins perturb these neuron-glia interactions and consequently, reproductive maturation and fertility. Deciphering the communication between GnRH neurons and other neural cell types constituting hypothalamic neuroendocrine circuits is thus critical both to understanding physiological processes such as puberty, oestrous cyclicity and aging, and to developing novel therapeutic strategies for dysfunctions of these processes, including the effects of endocrine disruptors.
Collapse
Affiliation(s)
- Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| |
Collapse
|
23
|
Duittoz AH, Forni PE, Giacobini P, Golan M, Mollard P, Negrón AL, Radovick S, Wray S. Development of the gonadotropin-releasing hormone system. J Neuroendocrinol 2022; 34:e13087. [PMID: 35067985 PMCID: PMC9286803 DOI: 10.1111/jne.13087] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/02/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022]
Abstract
This review summarizes the current understanding of the development of the neuroendocrine gonadotropin-releasing hormone (GnRH) system, including discussion on open questions regarding (1) transcriptional regulation of the Gnrh1 gene; (2) prenatal development of the GnRH1 system in rodents and humans; and (3) paracrine and synaptic communication during migration of the GnRH cells.
Collapse
Affiliation(s)
| | - Paolo E. Forni
- Department of Biological SciencesUniversity at AlbanyAlbanyNYUSA
- The RNA InstituteUniversity at AlbanyAlbanyNYUSA
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Postnatal BrainLille Neuroscience & Cognition, UMR‐S1172, Inserm, CHU LilleUniversity of LilleLilleFrance
| | - Matan Golan
- Institute of Animal SciencesAgricultural Research Organization – Volcani CenterRishon LetziyonIsrael
| | - Patrice Mollard
- Institute of Functional GenomicsCNRS, InsermMontpellier UniversityMontpellierFrance
| | - Ariel L. Negrón
- Clinical and Translational ResearchRutgers Robert Wood Johnson Medical SchoolNew BrunswickNJUSA
| | - Sally Radovick
- Clinical and Translational ResearchRutgers Robert Wood Johnson Medical SchoolNew BrunswickNJUSA
| | - Susan Wray
- Cellular and Developmental Neurobiology SectionNational Institute of Neurological Disorders and Stroke/National Institutes of HealthBethesdaMDUSA
| |
Collapse
|
24
|
Deller M, Gellrich J, Lohrer EC, Schriever VA. Genetics of congenital olfactory dysfunction: a systematic review of the literature. Chem Senses 2022; 47:6847567. [PMID: 36433800 DOI: 10.1093/chemse/bjac028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Olfaction, as one of our 5 senses, plays an important role in our daily lives. It is connected to proper nutrition, social interaction, and protection mechanisms. Disorders affecting this sense consequently also affect the patients' general quality of life. Because the underlying genetics of congenital olfactory disorders (COD) have not been thoroughly investigated yet, this systematic review aimed at providing information on genes that have previously been reported to be mutated in patients suffering from COD. This was achieved by systematically reviewing existing literature on 3 databases, namely PubMed, Ovid Medline, and ISI Web of Science. Genes and the type of disorder, that is, isolated and/or syndromic COD were included in this study, as were the patients' associated abnormal features, which were categorized according to the affected organ(-system). Our research yielded 82 candidate genes/chromosome loci for isolated and/or syndromic COD. Our results revealed that the majority of these are implicated in syndromic COD, a few accounted for syndromic and isolated COD, and the least underly isolated COD. Most commonly, structures of the central nervous system displayed abnormalities. This study is meant to assist clinicians in determining the type of COD and detecting potentially abnormal features in patients with confirmed genetic variations. Future research will hopefully expand this list and thereby further improve our understanding of COD.
Collapse
Affiliation(s)
- Matthias Deller
- Charité-Universitätsmedizin Berlin, Department of Pediatric Neurology, Berlin, Germany
| | - Janine Gellrich
- Abteilung Neuropädiatrie Medizinische Fakultät Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Elisabeth C Lohrer
- Abteilung Neuropädiatrie Medizinische Fakultät Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Valentin A Schriever
- Charité-Universitätsmedizin Berlin, Department of Pediatric Neurology, Berlin, Germany.,Abteilung Neuropädiatrie Medizinische Fakultät Carl Gustav Carus, Technische Universität, Dresden, Germany.,Charité-Universitätsmedizin Berlin, Center for Chronically Sick Children (Sozialpädiatrisches Zentrum, SPZ), Berlin, Germany
| |
Collapse
|
25
|
Abstract
Idiopathic hypogonadotropic hypogonadism (IHH) is a group of rare developmental disorders characterized by low gonadotropin levels in the face of low sex steroid hormone concentrations. IHH is practically divided into two major groups according to the olfactory function: normal sense of smell (normosmia) nIHH, and reduced sense of smell (hyposmia/anosmia) Kallmann syndrome (KS). Although mutations in more than 50 genes have been associated with IHH so far, only half of those cases were explained by gene mutations. Various combinations of deleterious variants in different genes as causes of IHH have been increasingly recognized (Oligogenic etiology). In addition to the complexity of inheritance patterns, the spontaneous or sex steroid-induced clinical recovery from IHH, which is seen in approximately 10–20% of cases, blurs further the phenotype/genotype relationship in IHH, and poses challenging steps in new IHH gene discovery. Beyond helping for clinical diagnostics, identification of the genetic mutations in the pathophysiology of IHH is hoped to shed light on the central governance of the hypothalamo-pituitary-gonadal axis through life stages. This review aims to summarize the genetic etiology of IHH and discuss the clinical and physiological ramifications of the gene mutations.
Collapse
|
26
|
Howard SR. Interpretation of reproductive hormones before, during and after the pubertal transition-Identifying health and disordered puberty. Clin Endocrinol (Oxf) 2021; 95:702-715. [PMID: 34368982 PMCID: PMC9291332 DOI: 10.1111/cen.14578] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 11/28/2022]
Abstract
Puberty is a process of transition from childhood to adult reproductive capacity, governed by the reactivation of the hypothalamic-pituitary-gonadal axis after a long period of dormancy in mid-childhood. As such, the reproductive hormones are in a state of flux during the adolescent years, and interpretation of both the onset of healthy, concordant puberty and the differentiation of precocious, delayed or disordered puberty, can be challenging. This review is focused on the description of the endocrine axes in healthy puberty and the markers of disorders of puberty that can aid diagnosis and management for patients with these conditions. It will cover the hypothalamic, pituitary and gonadal hormone systems, the dynamic changes that occur during puberty, conditions leading to precocious, delayed or absent puberty and other syndromes with disordered puberty, and the biochemical diagnosis of these different disorders of puberty.
Collapse
Affiliation(s)
- Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
27
|
Pingault V, Zerad L, Bertani-Torres W, Bondurand N. SOX10: 20 years of phenotypic plurality and current understanding of its developmental function. J Med Genet 2021; 59:105-114. [PMID: 34667088 PMCID: PMC8788258 DOI: 10.1136/jmedgenet-2021-108105] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/19/2021] [Indexed: 12/25/2022]
Abstract
SOX10 belongs to a family of 20 SRY (sex-determining region Y)-related high mobility group box-containing (SOX) proteins, most of which contribute to cell type specification and differentiation of various lineages. The first clue that SOX10 is essential for development, especially in the neural crest, came with the discovery that heterozygous mutations occurring within and around SOX10 cause Waardenburg syndrome type 4. Since then, heterozygous mutations have been reported in Waardenburg syndrome type 2 (Waardenburg syndrome type without Hirschsprung disease), PCWH or PCW (peripheral demyelinating neuropathy, central dysmyelination, Waardenburg syndrome, with or without Hirschsprung disease), intestinal manifestations beyond Hirschsprung (ie, chronic intestinal pseudo-obstruction), Kallmann syndrome and cancer. All of these diseases are consistent with the regulatory role of SOX10 in various neural crest derivatives (melanocytes, the enteric nervous system, Schwann cells and olfactory ensheathing cells) and extraneural crest tissues (inner ear, oligodendrocytes). The recent evolution of medical practice in constitutional genetics has led to the identification of SOX10 variants in atypical contexts, such as isolated hearing loss or neurodevelopmental disorders, making them more difficult to classify in the absence of both a typical phenotype and specific expertise. Here, we report novel mutations and review those that have already been published and their functional consequences, along with current understanding of SOX10 function in the affected cell types identified through in vivo and in vitro models. We also discuss research options to increase our understanding of the origin of the observed phenotypic variability and improve the diagnosis and medical care of affected patients.
Collapse
Affiliation(s)
- Veronique Pingault
- Department of Embryology and Genetics of Malformations, INSERM UMR 1163, Université de Paris and Institut Imagine, Paris, France .,Service de Génétique des Maladies Rares, AP-HP, Hopital Necker-Enfants Malades, Paris, France
| | - Lisa Zerad
- Department of Embryology and Genetics of Malformations, INSERM UMR 1163, Université de Paris and Institut Imagine, Paris, France
| | - William Bertani-Torres
- Department of Embryology and Genetics of Malformations, INSERM UMR 1163, Université de Paris and Institut Imagine, Paris, France
| | - Nadege Bondurand
- Department of Embryology and Genetics of Malformations, INSERM UMR 1163, Université de Paris and Institut Imagine, Paris, France
| |
Collapse
|
28
|
Фролова ЕБ, Петров ВМ, Васильев ЕВ, Макрецкая НA, Пилипенко ОВ, Тюльпаков АН. [Kalmann syndrome in monozygous twins as an isolated manifestation of the SOX10 gene defect]. PROBLEMY ENDOKRINOLOGII 2021; 67:43-47. [PMID: 34766489 PMCID: PMC9112924 DOI: 10.14341/probl12789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/16/2021] [Accepted: 09/07/2021] [Indexed: 12/16/2022]
Abstract
More than 30 genes are known to take part in hypothalamic-pituitary-gonadal axis development at the date and role of more than 10 other genes is studied. Despite it about 50% of isolated hypogonadotropic hypogonadism cases still have no molecular genetic explanation.A number of specific associations between iHH and different not-reproductive manifestations called syndromic forms are distinguished in general group of iHH. For example, the combination of Kalmann syndrome with sensorineural hearing loss is known as manifestation for defects of some genes encoding factors of neuronal migration; in patients with this phenotype CHD7, SOX10 genes defects are most frequent. However, defects in the genes of neuronal migration factors are characterized by a wide variability of phenotype, which is explained by the epigenetic mechanisms influence. Carriers of the mutation within the same family may lack some non-reproductive manifestations as well as hypogonadism.Here we present a case of Kalmann syndrome in monozygous twins, caused by a previously not described heterozygous mutation c.462C> G: p.I154M in the SOX10 gene in the absence of sensorineural hearing loss. The mutation was inherited from a father who has only isolated anosmia in the phenotype. This mutation was identified during full exome sequencing. This unique observation for Russia shows on the one hand expediency to check SOX10 sequence in addition to the other factors of neuronal migration and differentiation and, on the other hand, the prospect of full exome sequencing in a group of patients with undifferentiated iHH.
Collapse
Affiliation(s)
- Е. Б. Фролова
- Национальный медицинский исследовательский центр здоровья детей
| | - В. М. Петров
- Национальный медицинский исследовательский центр эндокринологии
| | - Е. В. Васильев
- Национальный медицинский исследовательский центр эндокринологии
| | | | | | - А. Н. Тюльпаков
- Медико-генетический научный центр им. Н.П. Бочкова; Республиканская детская клиническая больница
| |
Collapse
|
29
|
Oleari R, Massa V, Cariboni A, Lettieri A. The Differential Roles for Neurodevelopmental and Neuroendocrine Genes in Shaping GnRH Neuron Physiology and Deficiency. Int J Mol Sci 2021; 22:9425. [PMID: 34502334 PMCID: PMC8431607 DOI: 10.3390/ijms22179425] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 01/19/2023] Open
Abstract
Gonadotropin releasing hormone (GnRH) neurons are hypothalamic neuroendocrine cells that control sexual reproduction. During embryonic development, GnRH neurons migrate from the nose to the hypothalamus, where they receive inputs from several afferent neurons, following the axonal scaffold patterned by nasal nerves. Each step of GnRH neuron development depends on the orchestrated action of several molecules exerting specific biological functions. Mutations in genes encoding for these essential molecules may cause Congenital Hypogonadotropic Hypogonadism (CHH), a rare disorder characterized by GnRH deficiency, delayed puberty and infertility. Depending on their action in the GnRH neuronal system, CHH causative genes can be divided into neurodevelopmental and neuroendocrine genes. The CHH genetic complexity, combined with multiple inheritance patterns, results in an extreme phenotypic variability of CHH patients. In this review, we aim at providing a comprehensive and updated description of the genes thus far associated with CHH, by dissecting their biological relevance in the GnRH system and their functional relevance underlying CHH pathogenesis.
Collapse
Affiliation(s)
- Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy;
| | - Valentina Massa
- Department of Health Sciences, University of Milan, 20142 Milano, Italy;
- CRC Aldo Ravelli for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, 20142 Milano, Italy
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy;
| | - Antonella Lettieri
- Department of Health Sciences, University of Milan, 20142 Milano, Italy;
- CRC Aldo Ravelli for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, 20142 Milano, Italy
| |
Collapse
|
30
|
Wen J, Song J, Bai Y, Liu Y, Cai X, Mei L, Ma L, He C, Feng Y. A Model of Waardenburg Syndrome Using Patient-Derived iPSCs With a SOX10 Mutation Displays Compromised Maturation and Function of the Neural Crest That Involves Inner Ear Development. Front Cell Dev Biol 2021; 9:720858. [PMID: 34426786 PMCID: PMC8379019 DOI: 10.3389/fcell.2021.720858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/22/2021] [Indexed: 12/20/2022] Open
Abstract
Waardenburg syndrome (WS) is an autosomal dominant inherited disorder that is characterized by sensorineural hearing loss and abnormal pigmentation. SOX10 is one of its main pathogenicity genes. The generation of patient-specific induced pluripotent stem cells (iPSCs) is an efficient means to investigate the mechanisms of inherited human disease. In our work, we set up an iPSC line derived from a WS patient with SOX10 mutation and differentiated into neural crest cells (NCCs), a key cell type involved in inner ear development. Compared with control-derived iPSCs, the SOX10 mutant iPSCs showed significantly decreased efficiency of development and differentiation potential at the stage of NCCs. After that, we carried out high-throughput RNA-seq and evaluated the transcriptional misregulation at every stage. Transcriptome analysis of differentiated NCCs showed widespread gene expression alterations, and the differentially expressed genes (DEGs) were enriched in gene ontology terms of neuron migration, skeletal system development, and multicellular organism development, indicating that SOX10 has a pivotal part in the differentiation of NCCs. It's worth noting that, a significant enrichment among the nominal DEGs for genes implicated in inner ear development was found, as well as several genes connected to the inner ear morphogenesis. Based on the protein-protein interaction network, we chose four candidate genes that could be regulated by SOX10 in inner ear development, namely, BMP2, LGR5, GBX2, and GATA3. In conclusion, SOX10 deficiency in this WS subject had a significant impact on the gene expression patterns throughout NCC development in the iPSC model. The DEGs most significantly enriched in inner ear development and morphogenesis may assist in identifying the underlying basis for the inner ear malformation in subjects with WS.
Collapse
Affiliation(s)
- Jie Wen
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Song
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yijiang Bai
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yalan Liu
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinzhang Cai
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lingyun Mei
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Ma
- Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Chufeng He
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, China.,Department of Geriatrics, National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Feng
- Department of Otorhinolaryngology, Xiangya Hospital Central South University, Changsha, China.,Department of Otorhinolaryngology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
31
|
Louden ED, Poch A, Kim HG, Ben-Mahmoud A, Kim SH, Layman LC. Genetics of hypogonadotropic Hypogonadism-Human and mouse genes, inheritance, oligogenicity, and genetic counseling. Mol Cell Endocrinol 2021; 534:111334. [PMID: 34062169 DOI: 10.1016/j.mce.2021.111334] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022]
Abstract
Hypogonadotropic hypogonadism, which may be normosmic (nHH) or anosmic/hyposmic, known as Kallmann syndrome (KS), is due to gonadotropin-releasing hormone deficiency, which results in absent puberty and infertility. Investigation of the genetic basis of nHH/KS over the past 35 years has yielded a substantial increase in our understanding, as variants in 44 genes in OMIM account for ~50% of cases. The first genes for KS (ANOS1) and nHH (GNRHR) were followed by the discovery that FGFR1 variants may cause either nHH or KS. Associated anomalies include midline facial defects, neurologic deficits, cardiac anomalies, and renal agenesis, among others. Mouse models for all but one gene (ANOS1) generally support findings in humans. About half of the known genes implicated in nHH/KS are inherited as autosomal dominant and half are autosomal recessive, whereas only 7% are X-linked recessive. Digenic and oligogenic inheritance has been reported in 2-20% of patients, most commonly with variants in genes that may result in either nHH or KS inherited in an autosomal dominant fashion. In vitro analyses have only been conducted for both gene variants in eight cases and for one gene variant in 20 cases. Rigorous confirmation that two gene variants in the same individual cause the nHH/KS phenotype is lacking for most. Clinical diagnosis is probably best accomplished by targeted next generation sequencing of the known candidate genes with confirmation by Sanger sequencing. Elucidation of the genetic basis of nHH/KS has resulted in an enhanced understanding of this disorder, as well as normal puberty, which makes genetic diagnosis clinically relevant.
Collapse
Affiliation(s)
- Erica D Louden
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Department of Neuroscience & Regenerative Medicine, Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Alexandra Poch
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Department of Neuroscience & Regenerative Medicine, Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Afif Ben-Mahmoud
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Soo-Hyun Kim
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, United Kingdom
| | - Lawrence C Layman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Department of Neuroscience & Regenerative Medicine, Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
32
|
Davis EE, Balasubramanian R, Kupchinsky ZA, Keefe DL, Plummer L, Khan K, Meczekalski B, Heath KE, Lopez-Gonzalez V, Ballesta-Martinez MJ, Margabanthu G, Price S, Greening J, Brauner R, Valenzuela I, Cusco I, Fernandez-Alvarez P, Wierman ME, Li T, Lage K, Barroso PS, Chan YM, Crowley WF, Katsanis N. TCF12 haploinsufficiency causes autosomal dominant Kallmann syndrome and reveals network-level interactions between causal loci. Hum Mol Genet 2021; 29:2435-2450. [PMID: 32620954 DOI: 10.1093/hmg/ddaa120] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/27/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Dysfunction of the gonadotropin-releasing hormone (GnRH) axis causes a range of reproductive phenotypes resulting from defects in the specification, migration and/or function of GnRH neurons. To identify additional molecular components of this system, we initiated a systematic genetic interrogation of families with isolated GnRH deficiency (IGD). Here, we report 13 families (12 autosomal dominant and one autosomal recessive) with an anosmic form of IGD (Kallmann syndrome) with loss-of-function mutations in TCF12, a locus also known to cause syndromic and non-syndromic craniosynostosis. We show that loss of tcf12 in zebrafish larvae perturbs GnRH neuronal patterning with concomitant attenuation of the orthologous expression of tcf3a/b, encoding a binding partner of TCF12, and stub1, a gene that is both mutated in other syndromic forms of IGD and maps to a TCF12 affinity network. Finally, we report that restored STUB1 mRNA rescues loss of tcf12 in vivo. Our data extend the mutational landscape of IGD, highlight the genetic links between craniofacial patterning and GnRH dysfunction and begin to assemble the functional network that regulates the development of the GnRH axis.
Collapse
Affiliation(s)
- Erica E Davis
- Center for Human Disease Modeling, Duke University, Durham, NC 27701, USA.,Advanced Center for Translational and Genetic Medicine (ACT-GeM), Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ravikumar Balasubramanian
- Harvard Reproductive Endocrine Science Center, Massachusetts General Hospital (MGH), Boston, MA 02114, USA.,Harvard Medical School, Boston, MA 02115, USA
| | | | - David L Keefe
- Harvard Reproductive Endocrine Science Center, Massachusetts General Hospital (MGH), Boston, MA 02114, USA
| | - Lacey Plummer
- Harvard Reproductive Endocrine Science Center, Massachusetts General Hospital (MGH), Boston, MA 02114, USA
| | - Kamal Khan
- Advanced Center for Translational and Genetic Medicine (ACT-GeM), Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-512 Poznan, Poland
| | - Karen E Heath
- Institute of Medical and Molecular Genetics (INGEMM) Hospital Universitario La Paz, Universidad Autonoma de Madrid, IdiPAZ, Madrid, Spain and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28046 Madrid, Spain
| | - Vanesa Lopez-Gonzalez
- Medical Genetics Unit, Department of Pediatrics, Hospital Clinico, Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain and CIBERER, ISCIII, 28046 Madrid, Spain
| | - Mary J Ballesta-Martinez
- Medical Genetics Unit, Department of Pediatrics, Hospital Clinico, Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain and CIBERER, ISCIII, 28046 Madrid, Spain
| | | | - Susan Price
- Northampton General Hospital, Northampton NN1 5BD, UK
| | - James Greening
- University Hospitals of Leicester, Leicester LE3 9QP, UK
| | - Raja Brauner
- Pediatric Endocrinology Unit, Fondation Ophtalmologique Adolphe de Rothschild and Université Paris Descartes, 75019 Paris, France
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Medicine Genetics Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ivon Cusco
- Department of Clinical and Molecular Genetics, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Medicine Genetics Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Paula Fernandez-Alvarez
- Department of Clinical and Molecular Genetics, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Medicine Genetics Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Margaret E Wierman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Taibo Li
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kasper Lage
- Harvard Medical School, Boston, MA 02115, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Priscila Sales Barroso
- Divisao de Endocrinologia e Metabologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, 05403-900 Brazil
| | - Yee-Ming Chan
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - William F Crowley
- Harvard Medical School, Boston, MA 02115, USA.,MGH Center for Human Genetics & The Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston MA 02114, USA
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University, Durham, NC 27701, USA.,Advanced Center for Translational and Genetic Medicine (ACT-GeM), Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
33
|
Zhang L, Gao Y, Du Q, Liu L, Li Y, Dey SK, Banerjee S, Liao Z. Genetic Profiles and Three-year Follow-up Study of Chinese Males With Congenital Hypogonadotropic Hypogonadism. J Sex Med 2021; 18:1500-1510. [PMID: 37057435 DOI: 10.1016/j.jsxm.2021.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 06/06/2021] [Accepted: 07/06/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND The correlation between long-term treatment outcomes with genotypes in congenital hypogonadotropic hypogonadism (CHH) males is rarely reported. AIM To investigate the correlations among genotypes, phenotypes, and treatment outcomes for CHH male patients. METHODS Whole exome sequencing was performed for 73 Chinese CHH males from one academic center. Patients self-selected one of the 4 treatments: pulsatile Gonadorelin pump (PGP), cyclical gonadotropins therapy (CGT), human menopausal gonadotropin monotherapy, or testosterone replacement treatment. Clinical assessments were performed every 3 months for 3 years. OUTCOMES The pathogenicity of variants was determined. Baseline clinical features, spermatogenesis outcomes were analysed. RESULTS 62 variants were identified in 51 patients (69.9%), 17 of which were novel. Among these mutations, variants on FGFR1, PROKR2, CHD7, ANOS1 and NSMF gene were 16.1%, 16.1%, 11.3%, 8.1% and 8.1% respectively. 11 patients followed the oligogenic pattern (21.6%). All CHD7 patients had hearing impairment, or structural deformities of external/ inner ear and were diagnosed as CHARGE syndrome. 24.7% of CHH patients manifested with ear/hearing anomalies. KS patients had higher rates of cryptorchidism history and ear/hearing anomalies than normosmic CHH subjects. Male patients with PROKR2 mutations showed relatively better testicular development, less dental deformity when compared with FGFR1 mutations. About 30% normosmic patients defined by simple olfactory assessment showed olfactory nerve center (ONC) dysplasia under nasal sinus MRI examination. Among the CHH males treated with CGT or PGP, 70.2% reached spermatogenesis within 3 years of treatment. CLINICAL IMPLICATIONS No direct correlation was observed between certain responsible genes and spermatogenic outcomes. When CHH patients were identified with CHD7 variants, ear/hearing evaluation should be carefully performed. The precise assessment of ONC development was advised for normosmic CHH subjects. STRENGTHS & LIMITATIONS This study provided informative long-term treatment data of CHH male patients screened with whole exome sequencing. The limitations included small number of subgroups with multifaceted gene variants, clinical heterogeneity, uncontrolled sperm-inducing treatment method. The seventeen novel mutations worth experimental validation in the future. CONCLUSION The clinical severity is partially related with specific gene variants, and detailed individualized data and outcomes were provided. Ear/hearing anomalies were closely connected with CHD7 variants, and were common problems for CHH patients. Simple olfactory assessment underestimated the true olfactory deficit. L. Zhang, Y. Gao, Q. Du, et al. Genetic Profiles and Three-year Follow-up Study of Chinese Males With Congenital Hypogonadotropic Hypogonadism. J Sex Med 2021;XX:XXX-XXX.
Collapse
Affiliation(s)
- Luyao Zhang
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuting Gao
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qin Du
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liyi Liu
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanbing Li
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Subrata Kumar Dey
- Department of Biotechnology, Centre for Genetic Studies, School of Biotechnology and Biological Sciences, Maulana Abul Kalam Azad University of Technology (Formerly West Bengal University of Technology), Salt Lake City Kolkata, West Bengal, India
| | - Santasree Banerjee
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zhihong Liao
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Mechanisms of Central Hypogonadism. Int J Mol Sci 2021; 22:ijms22158217. [PMID: 34360982 PMCID: PMC8348115 DOI: 10.3390/ijms22158217] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 02/01/2023] Open
Abstract
Reproductive function depends upon an operational hypothalamo–pituitary–gonadal (HPG) axis. Due to its role in determining survival versus reproductive strategies, the HPG axis is vulnerable to a diverse plethora of signals that ultimately manifest with Central Hypogonadism (CH) in all its many guises. Acquired CH can result from any pituitary or hypothalamic lesion, including its treatment (such as surgical resection and/or radiotherapy). The HPG axis is particularly sensitive to the suppressive effects of hyperprolactinaemia that can occur for many reasons, including prolactinomas, and as a side effect of certain drug therapies. Physiologically, prolactin (combined with the suppressive effects of autonomic neural signals from suckling) plays a key role in suppressing the gonadal axis and establishing temporary CH during lactation. Leptin is a further key endocrine regulator of the HPG axis. During starvation, hypoleptinaemia (from diminished fat stores) results in activation of hypothalamic agouti-related peptide neurons that have a dual purpose to enhance appetite (important for survival) and concomitantly suppresses GnRH neurons via effects on neural kisspeptin release. Obesity is associated with hyperleptinaemia and leptin resistance that may also suppress the HPG axis. The suppressibility of the HPG axis also leaves it vulnerable to the effects of external signals that include morphine, anabolic-androgenic steroids, physical trauma and stress, all of which are relatively common causes of CH. Finally, the HPG axis is susceptible to congenital malformations, with reports of mutations within >50 genes that manifest with congenital CH, including Kallmann Syndrome associated with hyposmia or anosmia (reduction or loss of the sense of smell due to the closely associated migration of GnRH with olfactory neurons during embryogenesis). Analogous to the HPG axis itself, patients with CH are often vulnerable, and their clinical management requires both sensitivity and empathy.
Collapse
|
35
|
Wang G, Li X, Gao X, Su Y, Han M, Gao B, Guo C, Kang D, Huang S, Yuan Y, Dai P. Analysis of genotype-phenotype relationships in 90 Chinese probands with Waardenburg syndrome. Hum Genet 2021; 141:839-852. [PMID: 34142234 DOI: 10.1007/s00439-021-02301-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/08/2021] [Indexed: 12/31/2022]
Abstract
Waardenburg syndrome (WS) is a phenotypically and genetically heterogeneous disorder characterised by hearing loss and pigmentary abnormalities. We clarified the clinical and genetic features in 90 Chinese WS probands. Disease-causing variants were detected in 55 probands, for a molecular diagnosis rate of 61%, including cases related to PAX3 (14.4%), MITF (24.4%), and SOX10 (22.2%). Altogether, 48 variants were identified, including 44 single-nucleotide variants and 4 copy number variants. By parental genotyping, de novo variants were observed in 60% of probands and 15.4% of the de novo variation was associated with mosaicism. Statistical analyses revealed that brown freckles on the skin were more frequently seen in probands with MITF variants; patchy depigmented skin, asymmetric hearing loss, and white forelocks occurred more often in cases with PAX3 variants; and congenital inner ear malformations were more common and cochlear hypoplasia III was exclusively observed in those with SOX10 variants. In addition, we found that ranges of W-index values overlapped between WS probands with different genetic variants, and the use of the W-index as a tool for assessing dystopia canthorum may be problematic in Chinese. Herein, we report the spectrum of a cohort of WS probands and elucidate the relationship between genotype and phenotype.
Collapse
Affiliation(s)
- Guojian Wang
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China
| | - Xiaohong Li
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China.,Department of Otorhinolaryngology Head and Neck Surgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory for Pediatric, Diseases of Otolaryngology-Head and Neck Surgery, Beijing, China
| | - Xue Gao
- Department of Otolaryngology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yu Su
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China
| | - Mingyu Han
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China
| | - Bo Gao
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China
| | - Chang Guo
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China
| | - Dongyang Kang
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China
| | - Shasha Huang
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China.
| | - Yongyi Yuan
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China.
| | - Pu Dai
- Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing, National Clinical Research Center for Otolaryngologic Diseases, College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, #28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
36
|
Defects in GnRH Neuron Migration/Development and Hypothalamic-Pituitary Signaling Impact Clinical Variability of Kallmann Syndrome. Genes (Basel) 2021; 12:genes12060868. [PMID: 34198905 PMCID: PMC8229512 DOI: 10.3390/genes12060868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/30/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
Kallmann syndrome (KS) is a combination of isolated hypogonadotropic hypogonadism (IHH) with olfactory dysfunction, representing a heterogeneous disorder with a broad phenotypic spectrum. The genetic background of KS has not yet been fully established. This study was conducted on 46 Polish KS subjects (41 males, 5 females; average age: 29 years old). The studied KS patients were screened for defects in a 38-gene panel with next-generation sequencing (NGS) technology. The analysis revealed 27 pathogenic and likely pathogenic (P/LP) variants, and 21 variants of uncertain significance (VUS). The P/LP variants were detected in 20 patients (43.5%). The prevalence of oligogenic P/LP defects in selected genes among KS patients was 26% (12/46), whereas the co-occurrence of other variants was detected in 43% (20 probands). The examined KS patients showed substantial genotypic and phenotypic variability. A marked difference in non-reproductive phenotypes, involving defects in genes responsible for GnRH neuron development/migration and genes contributing to pituitary development and signaling, was observed. A comprehensive gene panel for IHH testing enabled the detection of clinically relevant variants in the majority of KS patients, which makes targeted NGS an effective molecular tool. The significance of oligogenicity and the high incidence of alterations in selected genes should be further elucidated.
Collapse
|
37
|
Katreddi RR, Forni PE. Mechanisms underlying pre- and postnatal development of the vomeronasal organ. Cell Mol Life Sci 2021; 78:5069-5082. [PMID: 33871676 PMCID: PMC8254721 DOI: 10.1007/s00018-021-03829-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/17/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
The vomeronasal organ (VNO) is sensory organ located in the ventral region of the nasal cavity in rodents. The VNO develops from the olfactory placode during the secondary invagination of olfactory pit. The embryonic vomeronasal structure appears as a neurogenic area where migratory neuronal populations like endocrine gonadotropin-releasing hormone-1 (GnRH-1) neurons form. Even though embryonic vomeronasal structures are conserved across most vertebrate species, many species including humans do not have a functional VNO after birth. The vomeronasal epithelium (VNE) of rodents is composed of two major types of vomeronasal sensory neurons (VSNs): (1) VSNs distributed in the apical VNE regions that express vomeronasal type-1 receptors (V1Rs) and the G protein subunit Gαi2, and (2) VSNs in the basal territories of the VNE that express vomeronasal type-2 receptors (V2Rs) and the G subunit Gαo. Recent studies identified a third subclass of Gαi2 and Gαo VSNs that express the formyl peptide receptor family. VSNs expressing V1Rs or V2Rs send their axons to distinct regions of the accessory olfactory bulb (AOB). Together, VNO and AOB form the accessory olfactory system (AOS), an olfactory subsystem that coordinates the social and sexual behaviors of many vertebrate species. In this review, we summarize our current understanding of cellular and molecular mechanisms that underlie VNO development. We also discuss open questions for study, which we suggest will further enhance our understanding of VNO morphogenesis at embryonic and postnatal stages.
Collapse
Affiliation(s)
- Raghu Ram Katreddi
- Department of Biological Sciences, Center for Neuroscience Research, The RNA Institute, University At Albany, State University of New York, Albany, NY, USA
| | - Paolo E Forni
- Department of Biological Sciences, Center for Neuroscience Research, The RNA Institute, University At Albany, State University of New York, Albany, NY, USA.
| |
Collapse
|
38
|
Wakabayashi T, Takei A, Okada N, Shinohara M, Takahashi M, Nagashima S, Okada K, Ebihara K, Ishibashi S. A novel SOX10 nonsense mutation in a patient with Kallmann syndrome and Waardenburg syndrome. Endocrinol Diabetes Metab Case Rep 2021; 2021:EDM200145. [PMID: 33913437 PMCID: PMC8115407 DOI: 10.1530/edm-20-0145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 11/23/2022] Open
Abstract
SUMMARY The underlying genetic drivers of Kallmann syndrome, a rare genetic disorder characterized by anosmia and hypogonadotropic hypogonadism due to impairment in the development of olfactory axons and in the migration of gonadotropin-releasing hormone (GNRH)-producing neurons during embryonic development, remain largely unknown. SOX10, a key transcription factor involved in the development of neural crest cells and established as one of the causative genes of Waardenburg syndrome, has been shown to be a causative gene of Kallmann syndrome. A 17-year-old male patient, who was diagnosed with Waardenburg syndrome on the basis of a hearing impairment and hypopigmented iris at childhood, was referred to our department because of anosmia and delayed puberty. As clinical examination revealed an aplastic olfactory bulb and hypogonadotropic hypogonadism, we diagnosed him as having Kallmann syndrome. Incidentally, we elucidated that he also presented with subclinical hypothyroidism without evidence of autoimmune thyroiditis. Direct sequence analysis detected a nonsense SOX10 mutation (c.373C>T, p.Glu125X) in this patient. Since this nonsense mutation has never been published as a germline variant, the SOX10 substitution is a novel mutation that results in Kallmann syndrome and Waardenburg syndrome. This case substantiates the significance of SOX10 as a genetic cause of Kallmann syndrome and Waardenburg syndrome, which possibly share a common pathway in the development of neural crest cells. LEARNING POINTS Kallmann syndrome and Waardenburg syndrome possibly share a common pathway during neural crest cell development. SOX10, a key transcription factor involved in the development of neural crest cells, is a common causative gene of Kallmann syndrome and Waardenburg syndrome. Careful evaluation about various phenotypic features may reveal the unknown genetic drivers of Kallmann syndrome.
Collapse
Affiliation(s)
- Tetsuji Wakabayashi
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Akihito Takei
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Nobukazu Okada
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Miki Shinohara
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Manabu Takahashi
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Shuichi Nagashima
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kenta Okada
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Ken Ebihara
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Shun Ishibashi
- Division of Endocrinology and MetabolismDepartment of Internal Medicine, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
39
|
Rojas RA, Kutateladze AA, Plummer L, Stamou M, Keefe DL, Salnikov KB, Delaney A, Hall JE, Sadreyev R, Ji F, Fliers E, Gambosova K, Quinton R, Merino PM, Mericq V, Seminara SB, Crowley WF, Balasubramanian R. Phenotypic continuum between Waardenburg syndrome and idiopathic hypogonadotropic hypogonadism in humans with SOX10 variants. Genet Med 2021; 23:629-636. [PMID: 33442024 PMCID: PMC8335791 DOI: 10.1038/s41436-020-01051-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
PURPOSE SOX10 variants previously implicated in Waardenburg syndrome (WS) have now been linked to Kallmann syndrome (KS), the anosmic form of idiopathic hypogonadotropic hypogonadism (IHH). We investigated whether SOX10-associated WS and IHH represent elements of a phenotypic continuum within a unifying disorder or if they represent phenotypically distinct allelic disorders. METHODS Exome sequencing from 1,309 IHH subjects (KS: 632; normosmic idiopathic hypogonadotropic hypogonadism [nIIHH]: 677) were reviewed for SOX10 rare sequence variants (RSVs). The genotypic and phenotypic spectrum of SOX10-related IHH (this study and literature) and SOX10-related WS cases (literature) were reviewed and compared with SOX10-RSV spectrum in gnomAD population. RESULTS Thirty-seven SOX10-associated IHH cases were identified as follows: current study: 16 KS; 4 nIHH; literature: 16 KS; 1 nIHH. Twenty-three IHH cases (62%; all KS), had ≥1 known WS-associated feature(s). Moreover, five previously reported SOX10-associated WS cases showed IHH-related features. Four SOX10 missense RSVs showed allelic overlap between IHH-ascertained and WS-ascertained cases. The SOX10-HMG domain showed an enrichment of RSVs in disease states versus gnomAD. CONCLUSION SOX10 variants contribute to both anosmic (KS) and normosmic (nIHH) forms of IHH. IHH and WS represent SOX10-associated developmental defects that lie along a unifying phenotypic continuum. The SOX10-HMG domain is critical for the pathogenesis of SOX10-related human disorders.
Collapse
Affiliation(s)
- Rebecca A Rojas
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anna A Kutateladze
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lacey Plummer
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Maria Stamou
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - David L Keefe
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kathyrn B Salnikov
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Angela Delaney
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Janet E Hall
- National Institute of Environmental Health Sciences, Research Triangle, NC, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Eric Fliers
- Amsterdam University Medical Center, location AMC, Department of Endocrinology and Metabolism, Amsterdam, The Netherlands
| | - Katarina Gambosova
- Stormont-Vail Health, Cotton O'Neil Diabetes and Endocrinology, Topeka, KS, USA
| | - Richard Quinton
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-tyne, UK
| | - Paulina M Merino
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| | - Veronica Mericq
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile
| | - Stephanie B Seminara
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - William F Crowley
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ravikumar Balasubramanian
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
40
|
Shima H, Tokuhiro E, Okamoto S, Nagamori M, Ogata T, Narumi S, Nakamura A, Izumi Y, Jinno T, Suzuki E, Fukami M. SOX10 Mutation Screening for 117 Patients with Kallmann Syndrome. J Endocr Soc 2021; 5:bvab056. [PMID: 34095692 PMCID: PMC8170842 DOI: 10.1210/jendso/bvab056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 11/19/2022] Open
Abstract
Introduction Kallmann syndrome (KS) is a genetically heterogeneous condition characterized by hypogonadotropic hypogonadism (HH) and olfactory dysfunction. Although SOX10, a causative gene for Waardenburg syndrome (WS) and peripheral demyelinating neuropathy, central demyelination, WS, and Hirschsprung disease (PCWH) has previously been implicated in KS, the clinical significance of SOX10 variants as the cause of KS remains uncertain. Patients and Methods A total of 117 patients with KS underwent mutation screening of SOX10 and 14 other causative genes for KS/HH. Rare SOX10 variants were subjected to in silico and in vitro analyses. We also examined clinical data of the patients and their parents with SOX10 variants. Results Sequence analysis identified 2 heterozygous variants of SOX10 (c.1225G > T, p.Gly409* and c.475C > T, p.Arg159Trp) in patients 1–3, as well as in the parents of patients 1 and 3. The variants were assessed as pathogenic/likely pathogenic, according to the American College of Medical Genomics guidelines. Both variants lacked in vitro transactivating activity for the MITF promoter and exerted no dominant-negative effects. Patients 1–3 carried no pathogenic variants in other genes examined. The patients presented with typical KS, while such features were absent in the parents of patients 1 and 3. None of the 5 variant-positive individuals exhibited hypopigmentation, while 1 and 2 individuals exhibited complete and partial hearing loss, respectively. Conclusion These results provide evidence that SOX10 haploinsufficiency accounts for a small percentage of KS cases. SOX10 haploinsufficiency is likely to be associated with a broad phenotypic spectrum, which includes KS without other clinical features of WS/PCWH.
Collapse
Affiliation(s)
- Hirohito Shima
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 157-8535 Tokyo, Japan
| | - Etsuro Tokuhiro
- Department of Pediatrics, Fujisawa City Hospital, 251-8550, Fujisawa, Japan
| | - Shingo Okamoto
- Okamoto Internal Medicine and Pediatrics Clinic, 633-0064, Sakurai, Japan
| | - Mariko Nagamori
- Department of Pediatrics, University of Toyama, 930-0194, Toyama, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, 431-3192, Hamamatsu, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 157-8535 Tokyo, Japan
| | - Akie Nakamura
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 157-8535 Tokyo, Japan
| | - Yoko Izumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 157-8535 Tokyo, Japan
| | - Tomoko Jinno
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 157-8535 Tokyo, Japan
| | - Erina Suzuki
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 157-8535 Tokyo, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 157-8535 Tokyo, Japan
| |
Collapse
|
41
|
Millar AC, Faghfoury H, Bieniek JM. Genetics of hypogonadotropic hypogonadism. Transl Androl Urol 2021; 10:1401-1409. [PMID: 33850776 PMCID: PMC8039576 DOI: 10.21037/tau.2020.03.33] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 03/11/2020] [Indexed: 01/01/2023] Open
Abstract
Male congenital hypogonadotropic hypogonadism (CHH) is a heterogenous group of genetic disorders that cause impairment in the production or action of gonadotropin releasing hormone (GnRH). These defects result in dysfunction of the hypothalamic-pituitary-gonadal hormone axis, leading to low testosterone levels and impaired fertility. Genetic testing techniques have expanded our knowledge of the underlying mechanisms contributing to CHH including over 30 genes to date implicated in the development of CHH. In some cases, non-reproductive signs or symptoms can give clues as to the putative genetic etiology, but many cases remain undiagnosed with less than 50% identified with a specific gene defect. This leads to many patients labelled as "idiopathic hypogonadotropic hypogonadism". Medical and family history as well as physical exam and laboratory features can aid in the identification of hypogonadotropic hypogonadism (HH) that is associated with specific medical syndromes or associated with other pituitary hormonal deficiencies. Genetic testing strategies are moving away from the classic practice of testing for only a few of the most commonly affected genes and instead utilizing next generation sequencing techniques that allow testing of numerous potential gene targets simultaneously. Treatment of CHH is dependent on the individual's desire to preserve fertility and commonly include human chorionic gonadotropin (hCG) and recombinant follicle stimulating hormone (rFSH) to stimulate testosterone production and spermatogenesis. In situations where fertility is not desired, testosterone replacement therapies are widely offered in order to maintain virilization and sexual function.
Collapse
Affiliation(s)
- Adam C. Millar
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Division of Endocrinology, Mount Sinai Hospital and Toronto General Hospital, Toronto, ON, Canada
| | - Hanna Faghfoury
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Fred A Litwin and Family Centre in Genetic Medicine, Mount Sinai Hospital and Toronto General Hospital, Toronto, ON, Canada
| | - Jared M. Bieniek
- Tallwood Urology & Kidney Institute, Hartford HealthCare, Hartford, CT, USA
| |
Collapse
|
42
|
Yan H, Ji H, Kubisiak T, Wu Y, Xiao J, Gu Q, Yang Y, Xie H, Ji T, Gao K, Li D, Xiong H, Shi Z, Li M, Zhang Y, Duan R, Bao X, Jiang Y, Burmeister M, Wang J. Genetic analysis of 20 patients with hypomyelinating leukodystrophy by trio-based whole-exome sequencing. J Hum Genet 2021; 66:761-768. [PMID: 33597727 PMCID: PMC8310791 DOI: 10.1038/s10038-020-00896-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 11/10/2020] [Indexed: 12/02/2022]
Abstract
Hypomyelinating leukodystrophies (HLDs) are a rare group of disorders characterized by myelin deficit of the brain-based on MRI. Here, we studied 20 patients with unexplained HLD to uncover their genetic etiology through whole-exome sequencing (WES). Trio-based WES was performed for 20 unresolved HLDs families after genetic tests for the PLP1 duplication and a panel of 115 known leukodystrophy-related genes. Variants in both known genes that related to HLDs and promising candidate genes were analyzed. Minigene splicing assay was conducted to confirm the effect of splice region variant. All 20 patients were diagnosed with HLDs clinically based on myelin deficit on MRI and impaired motor ability. Through WES, in 11 of 20 trios, 15 causative variants were detected in seven genes TUBB4A, POLR1C, POLR3A, SOX10, TMEM106B, DEGS1, and TMEM63A. The last three genes have just been discovered. Of 15 variants, six were novel. Using minigene splicing assay, splice variant POLR3A c.1770 + 5 G > C was proved to disrupt the normal splicing of intron 13 and led to a premature stop codon at position 618 (p.(P591Vfs*28)). Our analysis determined the molecular diagnosis of 11 HLDs patients. It emphasizes the heterogenicity of HLDs, the diagnostic power of trio-based WES for HLDs. Comprehensive analysis including a focus on candidate genes helps to discover novel disease-causing genes, determine the diagnosis for the first time, and improve the yield of WES. Moreover, novel mutations identified in TUBB4A, POLR3A, and POLR1C expand the mutation spectrum of these genes.
Collapse
Affiliation(s)
- Huifang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA.,Joint International Research Center of Translational and Clinical Research, Beijing, China
| | - Haoran Ji
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Thomas Kubisiak
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jiangxi Xiao
- Department of Radiology, Peking University First Hospital, Beijing, China
| | - Qiang Gu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Han Xie
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Taoyun Ji
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Dongxiao Li
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Hui Xiong
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Zhen Shi
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ming Li
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuehua Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ruoyu Duan
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xinhua Bao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Joint International Research Center of Translational and Clinical Research, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Margit Burmeister
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA. .,Departments of Computational Medicine & Bioinformatics, Psychiatry and Human Genetics, University of Michigan, Ann Arbor, MI, USA.
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China. .,Joint International Research Center of Translational and Clinical Research, Beijing, China. .,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.
| |
Collapse
|
43
|
Lin T, Luo L, Guo W, Ren W, Liu C, Wei H, Yang S, Wang Y. Phenotypic similarities in pigs with SOX10 c.321dupC and SOX10 c.325A>T mutations implied the correlation of SOX10 haploinsufficiency with Waardenburg syndrome. J Genet Genomics 2021; 47:770-780. [PMID: 33766494 DOI: 10.1016/j.jgg.2020.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 10/22/2022]
Abstract
SOX10 is a causative gene of Waardenburg syndrome (WS) that is a rare genetic disorder characterized by hearing loss and pigment disturbance. More than 100 mutations of SOX10 have been found in patients with Type 2 WS (WS2), Type 4 WS (WS4), and more complex syndromes. However, no mutation hotspot has been detected in SOX10, and most cases are sporadic, making it difficult to establish a correlation between the high phenotypic and genetic variability. In this study, a duplication of the 321th cytosine (c.321dupC) was introduced into SOX10 in pigs, which induced premature termination of the translation of SOX10 (p.K108QfsX45). The premature stop codon in Exon 3 triggered the degradation of mutant mRNA through nonsense-mediated mRNA decay. However, SOX10c.321dupC induced a highly similar phenotype of WS2 with heterogeneous inner ear malformation compared with its adjacent missense mutation SOX10c.325A>T. In addition, a site-saturation mutation analysis of the SOX10 N-terminal nuclear localization signal (n-NLS), where these two mutations located, revealed the correlation between SOX10 haploinsufficiency and WS by an in vitro reporter assay. The analysis combining the in vitro assay with clinical cases may provide a clue to clinical diagnoses.
Collapse
Affiliation(s)
- Tingting Lin
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Lihua Luo
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Weiwei Guo
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Wei Ren
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Chuanhong Liu
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China.
| | - Shiming Yang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China.
| | - Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Science, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
44
|
Barraud S, Delemer B, Poirsier-Violle C, Bouligand J, Mérol JC, Grange F, Higel-Chaufour B, Decoudier B, Zalzali M, Dwyer AA, Acierno JS, Pitteloud N, Millar RP, Young J. Congenital Hypogonadotropic Hypogonadism with Anosmia and Gorlin Features Caused by a PTCH1 Mutation Reveals a New Candidate Gene for Kallmann Syndrome. Neuroendocrinology 2021; 111:99-114. [PMID: 32074614 DOI: 10.1159/000506640] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/18/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Two loci (CHD7 and SOX10) underlying Kallmann syndrome (KS) were discovered through clinical and genetic analysis of CHARGE and Waardenburg syndromes, conditions that include congenital anosmia caused by olfactory bulb (CA/OBs) defects and congenital hypogonadotropic hypogonadism (CHH). We hypothesized that other candidate genes for KS could be discovered by analyzing rare syndromes presenting with these signs. Study Design, Size, Duration: We first investigated a family with Gorlin-Goltz syndrome (GGS) in which affected members exhibited clinical signs suggesting KS. Participants/Materials, Methods: Proband and family members underwent detailed clinical assessment. The proband received detailed neuroendocrine evaluation. Genetic analyses included sequencing the PTCH1 gene at diagnosis, followed by exome analyses of causative or candidate KS/CHH genes, in order to exclude contribution to the phenotypes of additional mutations. Exome analyses in additional 124 patients with KS/CHH probands with no additional GGS signs. RESULTS The proband exhibited CA, absent OBs on magnetic resonance imaging, and had CHH with unilateral cryptorchidism, consistent with KS. Pulsatile Gonadotropin-releasing hormone (GnRH) therapy normalized serum gonadotropins and increased testosterone levels, supporting GnRH deficiency. Genetic studies revealed 3 affected family members harbor a novel mutation of PTCH1 (c.838G> T; p.Glu280*). This unreported nonsense deleterious mutation results in either a putative truncated Ptch1 protein or in an absence of translated Ptch1 protein related to nonsense mediated messenger RNA decay. This heterozygous mutation cosegregates in the pedigree with GGS and CA with OBs aplasia/hypoplasia and with CHH in the proband suggesting a genetic linkage and an autosomal dominant mode of inheritance. No pathogenic rare variants in other KS/CHH genes cosegregated with these phenotypes. In additional 124 KS/CHH patients, 3 additional heterozygous, rare missense variants were found and predicted in silico to be damaging: p.Ser1203Arg, p.Arg1192Ser, and p.Ile108Met. CONCLUSION This family suggests that the 2 main signs of KS can be included in GGS associated with PTCH1 mutations. Our data combined with mice models suggest that PTCH1 could be a novel candidate gene for KS/CHH and reinforce the role of the Hedgehog signaling pathway in pathophysiology of KS and GnRH neuron migration.
Collapse
Affiliation(s)
- Sara Barraud
- Department of Endocrinology, Reims University Hospital, Reims, France
- University of Reims Champagne-Ardenne, Reims, France
| | - Brigitte Delemer
- Department of Endocrinology, Reims University Hospital, Reims, France
- University of Reims Champagne-Ardenne, Reims, France
| | | | - Jérôme Bouligand
- Department of Molecular Genetics, Pharmacogenomics, and Hormonology, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- University Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM U1185, Paris Saclay Medical School, Le Kremlin-Bicêtre, France
| | - Jean-Claude Mérol
- Department of Otolaryngology, Reims University Hospital, Reims, France
| | - Florent Grange
- Department of Dermatology, Reims University Hospital, Reims, France
| | | | | | - Mohamad Zalzali
- Department of Endocrinology, Reims University Hospital, Reims, France
| | - Andrew A Dwyer
- Boston College, William F. Connell School of Nursing, Chestnut Hill, Massachusetts, USA
| | - James S Acierno
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Robert P Millar
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jacques Young
- University Paris-Saclay, Le Kremlin-Bicêtre, France,
- Department of Reproductive Endocrinology, Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin-Bicêtre, France,
- INSERM U1185, Paris Saclay Medical School, Le Kremlin-Bicêtre, France,
| |
Collapse
|
45
|
Thongpradit S, Jinawath N, Javed A, Jensen LT, Chunsuwan I, Rojnueangnit K, Tim-Aroon T, Lertsukprasert K, Shiao MS, Sirachainan N, Wattanasirichaigoon D. Novel SOX10 Mutations in Waardenburg Syndrome: Functional Characterization and Genotype-Phenotype Analysis. Front Genet 2020; 11:589784. [PMID: 33362852 PMCID: PMC7756068 DOI: 10.3389/fgene.2020.589784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Waardenburg syndrome (WS) is a prevalent hearing loss syndrome, concomitant with focal skin pigmentation abnormalities, blue iris, and other abnormalities of neural crest-derived cells, including Hirschsprung’s disease. WS is clinically and genetically heterogeneous and it is classified into four major types WS type I, II, III, and IV (WS1, WS2, WS3, and WS4). WS1 and WS3 have the presence of dystopia canthorum, while WS3 also has upper limb anomalies. WS2 and WS4 do not have the dystopia canthorum, but the presence of Hirschsprung’s disease indicates WS4. There is a more severe subtype of WS4 with peripheral nerve and/or central nervous system involvement, namely peripheral demyelinating neuropathy, central dysmyelinating leukodystrophy, WS, and Hirschsprung’s disease or PCW/PCWH. We characterized the genetic defects underlying WS2, WS4, and the WS4-PCW/PCWH) using Sanger and whole-exome sequencing and cytogenomic microarray in seven patients from six unrelated families, including two with WS2 and five with WS4. We also performed multiple functional studies and analyzed genotype–phenotype correlations. The cohort included a relatively high frequency (80%) of individuals with neurological variants of WS4. Six novel SOX10 mutations were identified, including c.89C > A (p.Ser30∗), c.207_8 delCG (p.Cys71Hisfs∗62), c.479T > C (p.Leu160Pro), c.1379 delA (p.Tyr460Leufs∗42), c.425G > C (p.Trp142Ser), and a 20-nucleotide insertion, c.1155_1174dupGCCCCACTATGGCTCAGCCT (p.Phe392Cysfs∗117). All pathogenic variants were de novo. The results of reporter assays, western blotting, immunofluorescence, and molecular modeling supported the deleterious effects of the identified mutations and their correlations with phenotypic severity. The prediction of genotype–phenotype correlation and functional pathology, and dominant negative effect vs. haploinsufficiency in SOX10-related WS were influenced not only by site (first two vs. last coding exons) and type of mutation (missense vs. truncation/frameshift), but also by the protein expression level, molecular weight, and amino acid content of the altered protein. This in vitro analysis of SOX10 mutations thus provides a deeper understanding of the mechanisms resulting in specific WS subtypes and allows better prediction of the phenotypic manifestations, though it may not be always applicable to in vivo findings without further investigations.
Collapse
Affiliation(s)
- Supranee Thongpradit
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Integrative Computational BioScience Center (ICBS), Mahidol University, Salaya, Thailand
| | - Asif Javed
- Computational and Systems Biology Group, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore.,School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Laran T Jensen
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Issarapa Chunsuwan
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Kitiwan Rojnueangnit
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Thipwimol Tim-Aroon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Krisna Lertsukprasert
- Department of Communication Sciences and Disorders, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Meng-Shin Shiao
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nongnuch Sirachainan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duangrurdee Wattanasirichaigoon
- Division of Medical Genetics, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
46
|
Perera SN, Williams RM, Lyne R, Stubbs O, Buehler DP, Sauka-Spengler T, Noda M, Micklem G, Southard-Smith EM, Baker CVH. Insights into olfactory ensheathing cell development from a laser-microdissection and transcriptome-profiling approach. Glia 2020; 68:2550-2584. [PMID: 32857879 PMCID: PMC7116175 DOI: 10.1002/glia.23870] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022]
Abstract
Olfactory ensheathing cells (OECs) are neural crest-derived glia that ensheath bundles of olfactory axons from their peripheral origins in the olfactory epithelium to their central targets in the olfactory bulb. We took an unbiased laser microdissection and differential RNA-seq approach, validated by in situ hybridization, to identify candidate molecular mechanisms underlying mouse OEC development and differences with the neural crest-derived Schwann cells developing on other peripheral nerves. We identified 25 novel markers for developing OECs in the olfactory mucosa and/or the olfactory nerve layer surrounding the olfactory bulb, of which 15 were OEC-specific (that is, not expressed by Schwann cells). One pan-OEC-specific gene, Ptprz1, encodes a receptor-like tyrosine phosphatase that blocks oligodendrocyte differentiation. Mutant analysis suggests Ptprz1 may also act as a brake on OEC differentiation, and that its loss disrupts olfactory axon targeting. Overall, our results provide new insights into OEC development and the diversification of neural crest-derived glia.
Collapse
Affiliation(s)
- Surangi N. Perera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ruth M. Williams
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rachel Lyne
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Oliver Stubbs
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Dennis P. Buehler
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Tatjana Sauka-Spengler
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Japan
| | - Gos Micklem
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - E. Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Clare V. H. Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Abstract
The understanding of male factors of infertility has grown exponentially in the past ten years. While clear guidelines for obstructive azoospermia have been developed, management of non-obstructive azoospermia has lagged. Specifically, management of Kallmann Syndrome and central non-obstructive azoospermia has been limited by a lack of understanding of the molecular pathogenesis and investigational trials exploring the best option for management and fertility in these patients. This review aims to summarize our current understanding of the causes of central hypogonadotropic hypogonadism with a focus on genetic etiologies while also discussing options that endocrinologists and urologists can utilize to successfully treat this group of infertile men.
Collapse
Affiliation(s)
| | | | - Bobby B Najari
- NYU Langone Department of Urology, Department of Population Health, New York, NY, USA.
| |
Collapse
|
48
|
Taroc EZM, Katreddi RR, Forni PE. Identifying Isl1 Genetic Lineage in the Developing Olfactory System and in GnRH-1 Neurons. Front Physiol 2020; 11:601923. [PMID: 33192618 PMCID: PMC7609815 DOI: 10.3389/fphys.2020.601923] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/30/2020] [Indexed: 01/04/2023] Open
Abstract
During embryonic development, symmetric ectodermal thickenings [olfactory placodes (OP)] give rise to several cell types that comprise the olfactory system, such as those that form the terminal nerve ganglion (TN), gonadotropin releasing hormone-1 neurons (GnRH-1ns), and other migratory neurons in rodents. Even though the genetic heterogeneity among these cell types is documented, unidentified cell populations arising from the OP remain. One candidate to identify placodal derived neurons in the developing nasal area is the transcription factor Isl1, which was recently identified in GnRH-3 neurons of the terminal nerve in fish, as well as expression in neurons of the nasal migratory mass (MM). Here, we analyzed the Isl1 genetic lineage in chemosensory neuronal populations in the nasal area and migratory GnRH-1ns in mice using in situ hybridization, immunolabeling a Tamoxifen inducible Isl1CreERT and a constitutive Isl1Cre knock-in mouse lines. In addition, we also performed conditional Isl1 ablation in developing GnRH neurons. We found Isl1 lineage across non-sensory cells of the respiratory epithelium and sustentacular cells of OE and VNO. We identified a population of transient embryonic Isl1 + neurons in the olfactory epithelium and sparse Isl1 + neurons in postnatal VNO. Isl1 is expressed in almost all GnRH neurons and in approximately half of the other neuron populations in the MM. However, Isl1 conditional ablation alone does not significantly compromise GnRH-1 neuronal migration or GnRH-1 expression, suggesting compensatory mechanisms. Further studies will elucidate the functional and mechanistic role of Isl1 in development of migratory endocrine neurons.
Collapse
Affiliation(s)
- Ed Zandro M Taroc
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| | - Raghu Ram Katreddi
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| | - Paolo E Forni
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| |
Collapse
|
49
|
Hamada J, Ochi F, Sei Y, Takemoto K, Hirai H, Honda M, Shibata H, Hasegawa T, Eguchi M. A novel SOX10 variant in a Japanese girl with Waardenburg syndrome type 4C and Kallmann syndrome. Hum Genome Var 2020; 7:30. [PMID: 33082981 PMCID: PMC7522263 DOI: 10.1038/s41439-020-00118-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/01/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
We report the first case of Waardenburg syndrome type 4C and Kallmann syndrome in the same person. The patient, a Japanese girl, presented with bilateral iris depigmentation, bilateral sensorineural hearing loss, Hirschsprung disease, hypogonadotropic hypogonadism, and anosmia. We identified a novel SOX10 variant, c.124delC, p.Leu42Cysfs*67.
Collapse
Affiliation(s)
- Junpei Hamada
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Fumihiro Ochi
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yuka Sei
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Takemoto
- Department of Pediatrics, Ehime Prefectural Niihama Hospital, Ehime, Japan
| | - Hiroki Hirai
- Department of Pediatrics, Ehime Prefectural Central Hospital, Ehime, Japan
| | - Misa Honda
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Hironori Shibata
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Mariko Eguchi
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
50
|
Zhang L, Wu X, Lin X. Gene therapy for genetic mutations affecting non-sensory cells in the cochlea. Hear Res 2020; 394:107858. [PMID: 31791650 DOI: 10.1016/j.heares.2019.107858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/13/2019] [Accepted: 11/22/2019] [Indexed: 01/15/2023]
Abstract
Congenital hearing loss (HL) affects about 1 in every 500 infants. Among those affected more than half are caused by genetic mutations. According to the cellular sites affected by mutations in the cochlea, deafness genes could be classified into three major groups: those affecting the function of hair cells and synapses, cochlear supporting cells, and cells in the stria vascularis (SV) as well as in the lateral wall. The second and third groups account for more than half of all sensorineural hearing loss (SNHL) cases caused by genetic mutations. Current major treatment options for SNHL patients are hearing aids and cochlear implants (CIs). Hearing aids can only help patients with moderate to severe HL. Resolution of CIs is still improving and these devices are quite expensive especially when lifetime rehabilitation and maintenance costs are included. Tremendous efforts have been made to find novel treatments that are expected to restore hearing with higher-resolution and more natural quality, and to have a significantly lower cost over the lifetime of uses. Gene therapy studies have made impressive progresses in preclinical trials. This review focuses on deafness genes that affect supporting cells and cells in the SV of the cochlea. We will discuss recent progresses and remaining challenges for gene therapies targeting mutations in deafness genes belonging to this category.
Collapse
Affiliation(s)
- Li Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Department of Otolaryngology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322-3030, USA
| | - Xuewen Wu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China; Department of Otolaryngology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322-3030, USA
| | - Xi Lin
- Department of Otolaryngology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322-3030, USA.
| |
Collapse
|