1
|
Carroll SH, Schafer S, Richman AS, Tsay L, Wang P, Ahsan MU, Wang K, Liao EC. Genetic requirement for Esrp1/2 in vertebrate pituitary morphogenesis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.02.25325077. [PMID: 40236426 PMCID: PMC11998823 DOI: 10.1101/2025.04.02.25325077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The pituitary gland produces several hormones that regulate growth, metabolism, stress response, reproduction, and homeostasis. Congenital hypopituitarism is a deficiency in one or more pituitary hormones and encompasses a spectrum of clinical conditions. The pituitary has a complex embryonic origin with the oral ectoderm contributing the anterior lobe, and the neural ectoderm generating the posterior lobe. Pituitary abnormalities and growth deficiencies are associated with cleft palate however the developmental genetic connection between pituitary and orofacial cleft malformations remains to be determined. The epithelial RNA splicing regulators Esrp1 and Esrp2 are required for orofacial development in zebrafish, mice, and humans, and loss of function of these genes results in a cleft palate. Here we present a detailed developmental analysis of the genetic requirement for Esrp1/2 in pituitary morphogenesis in mouse and zebrafish. Further, we describe a patient with cleft palate and hypopituitarism that harbors a nucleotide variant in the RNA binding domain of ESRP2. The discovery of this key function for Esrp1/2 in pituitary formation has significant fundamental and clinical implications for understanding congenital hypopituitarism and craniofacial anomalies.
Collapse
Affiliation(s)
- Shannon H. Carroll
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Shriners Hospital for Children, Tampa, FL, USA
| | - Sogand Schafer
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariella S. Richman
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lisa Tsay
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Peng Wang
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mian Umair Ahsan
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kai Wang
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric C. Liao
- Center for Craniofacial Innovation, Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Shriners Hospital for Children, Tampa, FL, USA
| |
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
|
Santoro C, Aiello F, Farina A, Miraglia del Giudice E, Pascarella F, Licenziati MR, Improda N, Piluso G, Torella A, Del Vecchio Blanco F, Cirillo M, Nigro V, Grandone A. A Novel Missense Variant in LHX4 in Three Children with Multiple Pituitary Hormone Deficiency Belonging to Two Unrelated Families and Contribution of Additional GLI2 and IGFR1 Variant. CHILDREN (BASEL, SWITZERLAND) 2025; 12:364. [PMID: 40150646 PMCID: PMC11941417 DOI: 10.3390/children12030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/03/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Multiple genes can disrupt hypothalamic-pituitary axis development, causing multiple pituitary hormone deficiencies (MPHD). Despite advances in next-generation sequencing (NGS) identifying over 30 key genes, 85% of cases remain unsolved, indicating complex genotype-phenotype correlations and variable inheritance patterns. OBJECTIVE This study aimed to identify the MPHD genetics in three probands from two unrelated families. METHODS Family A had one affected child, while Family B had two affected siblings. All probands exhibited poor growth since birth, and family B's probands were born small for gestational age. Growth hormone deficiency was confirmed in all subjects. Family B's probands responded poorly to growth hormone treatment compared to the first patient. Furthermore, Family A's proband and Family B's younger sibling developed central hypothyroidism, while Family B's older sibling presented hypogonadotropic hypogonadism. Brain magnetic resonance imaging (MRI) revealed pituitary hypoplasia, ectopic posterior pituitary gland, and small sella turcica in all probands. Patients and their available relatives underwent NGS. RESULTS NGS identified the same novel and likely pathogenic LHX4 variant (c.481C>G) in all probands despite the families being unrelated. Additionally, Family A's proband carried a GLI2 variant (c.2105C>A), and Family B's probands carried an IGF1R variant (c.166G>A), both interpreted as being of uncertain significance. CONCLUSIONS This study confirms that heterozygous pathogenic variants of LHX4 can cause MPHD associated with a specific neuroradiological triad of abnormalities despite incomplete penetrance and variable phenotype. Moreover, the co-occurrence of the other two gene variants was debated. The IGF1R variant could explain the unusually poor response to growth hormone therapy in Family B, suggesting an oligogenic mechanism underlying the phenotype.
Collapse
Affiliation(s)
- Claudia Santoro
- Department of Child, Woman, General and Specialized Surgery, University of Campania “L. Vanvitelli”, L. De Crecchio 4 Street, 80138 Naples, Italy; (C.S.); (E.M.d.G.); (A.G.)
| | - Francesca Aiello
- Department of Child, Woman, General and Specialized Surgery, University of Campania “L. Vanvitelli”, L. De Crecchio 4 Street, 80138 Naples, Italy; (C.S.); (E.M.d.G.); (A.G.)
| | - Antonella Farina
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Sant’Andrea delle Dame Square L. De Crecchio 7 Street, 80138 Naples, Italy; (A.F.); (G.P.); (A.T.); (F.D.V.B.); (V.N.)
| | - Emanuele Miraglia del Giudice
- Department of Child, Woman, General and Specialized Surgery, University of Campania “L. Vanvitelli”, L. De Crecchio 4 Street, 80138 Naples, Italy; (C.S.); (E.M.d.G.); (A.G.)
| | - Filomena Pascarella
- Pediatric Endocrinology Unit, Sant’Anna e San Sebastiano Hospital, Palasciano Street, 81100 Caserta, Italy
| | - Maria Rosaria Licenziati
- Neuro-Endocrine Diseases and Obesity Unit, Department of Neurosciences, Santobono-Pausilipon Children’s Hospital, Via Egiziaca a Forcella, 18, 80139 Naples, Italy; (M.R.L.); (N.I.)
| | - Nicola Improda
- Neuro-Endocrine Diseases and Obesity Unit, Department of Neurosciences, Santobono-Pausilipon Children’s Hospital, Via Egiziaca a Forcella, 18, 80139 Naples, Italy; (M.R.L.); (N.I.)
| | - Giulio Piluso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Sant’Andrea delle Dame Square L. De Crecchio 7 Street, 80138 Naples, Italy; (A.F.); (G.P.); (A.T.); (F.D.V.B.); (V.N.)
| | - Annalaura Torella
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Sant’Andrea delle Dame Square L. De Crecchio 7 Street, 80138 Naples, Italy; (A.F.); (G.P.); (A.T.); (F.D.V.B.); (V.N.)
| | - Francesca Del Vecchio Blanco
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Sant’Andrea delle Dame Square L. De Crecchio 7 Street, 80138 Naples, Italy; (A.F.); (G.P.); (A.T.); (F.D.V.B.); (V.N.)
| | - Mario Cirillo
- Advanced MRI Neuroimaging Centre, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Sant’Andrea delle Dame Square, 80138 Naples, Italy;
- MRI Research Center SUN-FISM, University of Campania “Luigi Vanvitelli”, Sant’Andrea delle Dame Square, 80138 Naples, Italy
| | - Vincenzo Nigro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Sant’Andrea delle Dame Square L. De Crecchio 7 Street, 80138 Naples, Italy; (A.F.); (G.P.); (A.T.); (F.D.V.B.); (V.N.)
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Anna Grandone
- Department of Child, Woman, General and Specialized Surgery, University of Campania “L. Vanvitelli”, L. De Crecchio 4 Street, 80138 Naples, Italy; (C.S.); (E.M.d.G.); (A.G.)
| |
Collapse
|
4
|
Nguyen HT, Nguyen KN, Dien TM, Can TBN, Nguyen TTN, Lien NTK, Tung NV, Xuan NT, Tao NT, Nguyen NL, Tran VK, Mai TTC, Tran VA, Nguyen HH, Vu CD. Identification of POU1F1 Variants in Vietnamese Patients with Combined Pituitary Hormone Deficiency. Int J Mol Sci 2025; 26:2406. [PMID: 40141050 PMCID: PMC11941804 DOI: 10.3390/ijms26062406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Hypopituitarism is a condition characterized by the deficiency of several hormones produced by the pituitary gland. Genetic factors play an important role. Variants in the POU1F1 gene are associated with combined pituitary hormone deficiency 1 (CPHD1), which manifests as deficiencies in growth hormone (GH), thyroid-stimulating hormone (TSH), and prolactin (PRL). This study aimed to analyze the phenotype, genotype, treatment, and outcomes of Vietnamese patients with deficiency. Six patients from five unrelated families, initially diagnosed with hypopituitarism, were enrolled in this study. Data on physical characteristics, biochemical tests, treatment, outcomes, and follow-up were collected. Exome sequencing and Sanger sequencing were conducted to identify disease-causing variants in five probands and their families. All six patients exhibited anterior pituitary hypoplasia on brain magnetic resonance imaging and presented with TSH, GH, and PRL deficiencies. Exome sequencing identified three variants in the POU1F1 gene: c.428G>A p.(Arg143Gln), c.557T>G p.(Leu186Arg), and c.811C>T p.(Arg271Trp). The c.811C>T p.(Arg271Trp) variant was found in three patients, while c.557T>G p.(Leu186Arg) is a novel variant. Based on the ACMG classification, these variants were categorized as likely pathogenic or pathogenic variants. All patients were definitively diagnosed with CPHD1 caused by POU1F1 variants. All patients received levothyroxine and recombinant human growth hormone (rhGH) replacement therapy, leading to considerable growth. During the first year of treatment, all patients showed excellent growth response, with height increases ranging from 11 to 24 cm. After three years of treatment, two patients achieved normal height. One of the six patients developed scoliosis during treatment, which resolved after a one-year pause in rhGH therapy. Upon resuming treatment, no recurrence of scoliosis was observed. Our findings reveal the importance of early hormone testing and genetic analysis in improving the care and outcomes for patients with combined pituitary hormone deficiency.
Collapse
Affiliation(s)
- Ha Thu Nguyen
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (H.T.N.); (K.N.N.); (T.T.C.M.); (V.A.T.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Khanh Ngoc Nguyen
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (H.T.N.); (K.N.N.); (T.T.C.M.); (V.A.T.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Tran Minh Dien
- Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Thi Bich Ngoc Can
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Thi Thanh Ngan Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 10072, Vietnam; (T.T.N.N.); (N.T.K.L.); (N.V.T.); (N.T.X.); (N.T.T.)
| | - Nguyen Thi Kim Lien
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 10072, Vietnam; (T.T.N.N.); (N.T.K.L.); (N.V.T.); (N.T.X.); (N.T.T.)
| | - Nguyen Van Tung
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 10072, Vietnam; (T.T.N.N.); (N.T.K.L.); (N.V.T.); (N.T.X.); (N.T.T.)
| | - Nguyen Thi Xuan
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 10072, Vietnam; (T.T.N.N.); (N.T.K.L.); (N.V.T.); (N.T.X.); (N.T.T.)
| | - Nguyen Thien Tao
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 10072, Vietnam; (T.T.N.N.); (N.T.K.L.); (N.V.T.); (N.T.X.); (N.T.T.)
| | - Ngoc Lan Nguyen
- Center for Gene and Protein Research, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (N.L.N.); (V.K.T.)
| | - Van Khanh Tran
- Center for Gene and Protein Research, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (N.L.N.); (V.K.T.)
| | - Tran Thi Chi Mai
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (H.T.N.); (K.N.N.); (T.T.C.M.); (V.A.T.)
| | - Van Anh Tran
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (H.T.N.); (K.N.N.); (T.T.C.M.); (V.A.T.)
| | - Huy Hoang Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 10072, Vietnam; (T.T.N.N.); (N.T.K.L.); (N.V.T.); (N.T.X.); (N.T.T.)
| | - Chi Dung Vu
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (H.T.N.); (K.N.N.); (T.T.C.M.); (V.A.T.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| |
Collapse
|
5
|
Ji Z, Duan W, Wu J, Dai M, Chen P, Luo J, Xu M. A Pedigree With LHX4 and SOX3 Gene Variants Resulting in Gonadal Dysplasia. Clin Case Rep 2025; 13:e70118. [PMID: 39991535 PMCID: PMC11843472 DOI: 10.1002/ccr3.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/19/2024] [Accepted: 12/15/2024] [Indexed: 02/25/2025] Open
Abstract
Pituitary stalk interruption syndrome is a disease with multiple hormone deficiencies caused by pituitary stalk interruption. Identification of the pathogenic gene variant is helpful for early diagnosis. Early hormone replacement therapy is helpful to improve the prognosis and quality of life of patients. LHX4 (NM_033343.4):c.612G>C (p.Trp204Cys) variant may lead to PSIS.
Collapse
Affiliation(s)
- Zhaoyang Ji
- Department of Endocrinology and Metabolism, Shulan (Hangzhou) Affiliated Hospital of Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouZhejiangChina
- Department of General MedicineZhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of SciencesHangzhouZhejiangChina
| | - Wenyuan Duan
- Rare Disease CenterShulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical CollegeHangzhouZhejiangChina
- Yinfeng Gene Technology Co. Ltd.JinanShandongChina
| | - Jianbo Wu
- Department of Endocrinology and Metabolism, Shulan (Hangzhou) Affiliated Hospital of Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouZhejiangChina
| | - Mengting Dai
- Zhejiang University of MedicineHangzhouZhejiangChina
| | - Ping Chen
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
| | - Jialu Luo
- Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Mingzhi Xu
- Department of General MedicineZhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of SciencesHangzhouZhejiangChina
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical CollegeZhejiang Shuren UniversityHangzhouChina
| |
Collapse
|
6
|
Aouchiche K, Charmensat C, Morgane P, Teinturier C, Bretones P, Brac de la Perriere A, Layet V, Bouhours-Nouet N, Vantyghem MC, Haine E, Nunes-Sanchez ML, Camard O, Baron S, Castinetti F, Barlier A, Brue T, Reynaud R, Saveanu A. Phenotype and genotype of 23 patients with hypopituitarism and pathogenic GLI2 variants. Eur J Endocrinol 2025; 192:110-118. [PMID: 39938560 DOI: 10.1093/ejendo/lvaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/11/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025]
Abstract
OBJECTIVE To analyze the phenotype and genotype of patients with congenital hypopituitarism (CH) and pathogenic (P) GLI2 variants. METHODS A large cohort of patients with hypopituitarism was screened for GLI2 variants using a next-generation sequencing panel. Genotype-phenotype correlations were then assessed using GENHYPOPIT phenotypic data. RESULTS Of the 39 GLI2 variants identified in 717 index cases, 17 were classified as pathogenic and likely pathogenic. All these GLI2 variants were identified in 23 patients (17 index cases and 6 relatives) with associated pituitary stalk interruption syndrome or extrapituitary manifestations. GLI2 variants were the most frequently identified genetic cause in patients with syndromic hypopituitarism (68%): 88% (15/17) of mutations were truncating variants, and 45% were de novo. Most patients with a GLI2 variant (21/23, 91%) had hypopituitarism, including 21.7% (5/23) presenting isolated growth hormone deficiency. Two patients had Kallmann syndrome. Pituitary morphological abnormalities were present in 84% of the patients with P GLI2 variants (index cases and affected relatives). The remaining signs included neurocognitive disorders (38%), hexadactyly (27%), cardiac septal defects, and renal/vesical abnormalities. A possible digenic origin (GLI2/HESX1) is proposed in one family. CONCLUSION In this large multicentric international cohort, GLI2 was the most frequently identified genetic cause of syndromic CH with constant association of pituitary stalk interruption syndrome or extrapituitary clinical features. In addition to polydactyly and neurocognitive disorders, cardiac and renal abnormalities were also frequently observed and should be investigated further. The variable expression of GLI2-associated phenotypes justifies further research in this area.
Collapse
Affiliation(s)
- Karine Aouchiche
- Department of Pediatrics Endocrinology, CHU Timone Enfants, Assistance Publique-Hôpitaux de Marseille (APHM), 13385 Marseille, France
- Aix Marseille University, INSERM, MMG, UMR 1251, 13385 Marseille, France
| | - Camille Charmensat
- Department of Pediatrics Endocrinology, CHU Timone Enfants, Assistance Publique-Hôpitaux de Marseille (APHM), 13385 Marseille, France
| | - Pertuit Morgane
- Laboratory of Molecular Biology GEnOPé, Assistance-Publique des Hôpitaux de Marseille (AP-HM), Hôpital de la Timone, 13385 Marseille, France
| | - Cécile Teinturier
- Department of Pediatric Endocrinology and Diabetes, Assistance Publique-Hôpitaux de Paris (AP-HP), Endocrinology and Diabetes for Children, Bicêtre Paris Sud Hospital (HUPS), 94270 Le Kremlin-Bicêtre, Val-de-Marne, France
| | - Patricia Bretones
- Department of Pediatric Endocrinology, Hopital Mère Enfant, Hospices Civils de Lyon (HCL), 69500 Bron, France
| | - Aude Brac de la Perriere
- Department of Endocrinology, Groupement Hospitalier Est, Hospices Civils de Lyon (HCL), 69500 Bron, France
| | - Valérie Layet
- Department of Genetics, Havre's Hospital, 76600 Le Havre, France
| | - Natacha Bouhours-Nouet
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49933 Angers, France
| | - Marie-Christine Vantyghem
- Department of Endocrinology, Diabetology, Metabolism, University Hospital of Lille, 59037 Lille, France
| | - Elsa Haine
- Department of Pediatrics, University Hospital of Nice-Lenval Hospital, 06200 Nice, France
| | | | - Odile Camard
- Department of Pediatrics, Niort Hospital, 79021 Niort, France
| | - Sabine Baron
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Nantes, 44093 Nantes, France
| | - Frederic Castinetti
- Department of Endocrinology, APHM, Aix Marseille Univ, INSERM, MMG, MarMaRa Institute, UMR 1251, La Conception University Hospital, 13385 Marseille, France
| | - Anne Barlier
- Department of Endocrinology, APHM, Aix Marseille Univ, INSERM, MMG, MarMaRa Institute, UMR 1251, La Conception University Hospital, 13385 Marseille, France
- Laboratory of Molécular Biology GenOpé, APHM, Aix Marseille Univ, INSERM, MMG, IUMR 1251, La Timone University Hospital, 13385 Marseille, France
| | - Thierry Brue
- Department of Endocrinology, APHM, Aix Marseille Univ, INSERM, MMG, MarMaRa Institute, UMR 1251, La Conception University Hospital, 13385 Marseille, France
| | - Rachel Reynaud
- Department of Pediatrics Endocrinology, CHU Timone Enfants, Assistance Publique-Hôpitaux de Marseille (APHM), 13385 Marseille, France
- Aix Marseille University, INSERM, MMG, UMR 1251, 13385 Marseille, France
| | - Alexandru Saveanu
- Laboratory of Molécular Biology GenOpé, APHM, Aix Marseille Univ, INSERM, MMG, IUMR 1251, La Timone University Hospital, 13385 Marseille, France
| |
Collapse
|
7
|
Garrelfs MR, Mooij CF, Boelen A, van Trotsenburg ASP, Zwaveling-Soonawala N. Newborn screening for central congenital hypothyroidism: past, present and future. Eur Thyroid J 2025; 14:e240329. [PMID: 39913280 PMCID: PMC11883867 DOI: 10.1530/etj-24-0329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/17/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Congenital hypothyroidism (CH) is defined as thyroid hormone deficiency at birth and constitutes one of the most common causes of preventable intellectual disability worldwide. Central CH is caused by insufficient pituitary or hypothalamic control of thyroid function, biochemically characterized by a low serum free thyroxine (fT4), in combination with a low, normal or mildly elevated thyroid-stimulating hormone (TSH). Central CH is less common than primary CH and is part of multiple pituitary hormone deficiencies (MPHD) in most of the cases. MPHD at birth, also known as 'congenital hypopituitarism', is a potentially life-threatening condition due to the possible co-occurrence of adrenocorticotropin hormone and growth hormone deficiency that can result in severe hypoglycemia and adrenal crisis. To date, central CH is the only pituitary hormone deficiency suitable for newborn screening (NBS), providing an opportunity for early detection of MPHD. Even though the first NBS programs utilized T4-based methods that were able to identify central CH, most countries have since transitioned to TSH-based approaches due to the high rate of false positives associated with T4-based strategies. Now, 50 years after the introduction of NBS for CH, only a few countries around the world have a screening program capable of detecting central CH. In this paper, we review the past, present and future of NBS for central CH. We will outline the importance of early detection of central CH and discuss the challenges and opportunities of screening for this condition.
Collapse
Affiliation(s)
- Mark R Garrelfs
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Christiaan F Mooij
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Anita Boelen
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A S Paul van Trotsenburg
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Nitash Zwaveling-Soonawala
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Besci Ö, Sevim RD, Acinikli KY, Demir K, Çatlı G, Özhan B, Ünüvar T, Anık A, Abacı A, Altıncık A. Clinical Characteristics of Children with Combined Pituitary Hormone Deficiency and the Effects of Growth Hormone Treatment. KLINISCHE PADIATRIE 2025; 237:11-20. [PMID: 38049102 DOI: 10.1055/a-2186-9304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
AIM We aimed to describe the clinical characteristics of patients with congenital combined pituitary hormone deficiency (CPHD) and evaluate the first-year growth responses of individuals with CPHD and isolated growth hormone deficiency (IGHD) in order to establish the influence of other hormone deficiencies on growth response. PATIENTS AND METHODS This retrospective study was conducted in four tertiary care centers in Turkey. The records of patients diagnosed with CPHD (n=39) and severe IGHD (n=50) were collected. Cases with acquired lesions or chronic diseases were not included in the study. Data are presented as median (interquartile range). RESULTS Among 39 patients (13 females; 33%) with a diagnosis of CPHD, the majority of patients (64%) presented initially with combined deficits at baseline examination, whereas isolated deficiencies (36%) were less prevalent. Among all patients with GH deficiency, TSH, ACTH, FSH/LH, and ADH deficiencies were present in 94%, 74%, 44%, and 9% of patients, respectively. Patients with CPHD were diagnosed at a younger age (4.9 (8.4) vs. 11.6 (4.1), p<0.001, respectively) and had lower peak GH concentrations (0.4 (1.8) vs. 3.7 (2.9), p<0.001, respectively) than patients with IGHD. Patients with IGHD and CPHD had similar first-year growth responses (Δheight SD score of 0.55 (0.63) vs. 0.76 (0.71), respectively, p=0.45). CONCLUSIONS We established the nature and timing of numerous hormonal deficits emerging over time. We also identified that the existence of CPHD did not hinder growth response.
Collapse
Affiliation(s)
- Özge Besci
- Pediatric Endocrinology, Dokuz Eylul University, İzmir, Turkey
| | | | | | - Korcan Demir
- Pediatric Endocrinology, Dokuz Eylul University, İzmir, Turkey
| | - Gönül Çatlı
- Pediatric Endocrinology, Istinye University, Istanbul, Turkey
| | - Bayram Özhan
- Pediatric Endocrinology, Pamukkale University, Denizli, Turkey
| | - Tolga Ünüvar
- Pediatric Endocrinology, Adnan Menderes University, Aydin, Turkey
| | - Ahmet Anık
- Pediatric Endocrinology, Adnan Menderes University, Aydin, Turkey
| | - Ayhan Abacı
- Pediatric Endocrinology, Dokuz Eylul University, İzmir, Turkey
| | - Ayça Altıncık
- Pediatric Endocrinology, Pamukkale University, Denizli, Turkey
| |
Collapse
|
9
|
Lee Y, Lee YA, Ko JM, Shin CH, Lee YJ. Clinical and genetic features of childhood-onset congenital combined pituitary hormone deficiency: a retrospective, single-center cohort study. Ann Pediatr Endocrinol Metab 2024; 29:379-386. [PMID: 39778407 PMCID: PMC11725638 DOI: 10.6065/apem.2448008.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/26/2024] [Accepted: 03/27/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE To investigate the clinical characteristics and genetic features of childhood-onset congenital combined pituitary hormone deficiency (cCPHD) in Korean patients. METHODS We retrospectively analyzed 444 patients diagnosed with childhood-onset CPHD at a tertiary center between 1994 and 2021. After excluding acquired case, 43 patients with cCPHD were enrolled. Anthropometric measurements, hormone evaluations, brain magnetic resonance imaging (MRI), extrapituitary phenotypes, and adult outcomes were analyzed. Genetic analyses were performed on 26 patients using a targeted gene panel or whole exome sequencing. RESULTS Mean age at diagnosis was 3.2 years, and 41.9% were diagnosed at less than 1 year old. Short stature was the most frequent (37.2%) initial presentation, and mean height z-score was -2.4. More than half (n=23, 53.5%) of patients had neonatal features suggestive of hypopituitarism; however, only 15 (65.2%) were diagnosed in infancy. Growth hormone deficiency (GHD) was prevalent in 42 (97.7%), and 33 (76.7%) had 3 or more hormone deficiencies. Extrapituitary phenotypes were identified in 31 (72.1%). Brain MRI abnormalities correlated with a higher number of hormone deficiencies (P for trend 0.049) and were present in 33 patients (80.5%). Adult GHD was diagnosed in all 17 investigated patients, and metabolic disturbances were noted in 10 (58.9%). Pathogenic variants in POU1F1, GLI2, HESX1, TBC1D32, and ROBO1 were found in 5 (19.2%). CONCLUSION Considering the high proportion of neonatal presentations, identification of the early neonatal features of hypopituitarism to manage pituitary and extrapituitary phenotypes is critical. The genetic etiology of cCPHD warrants further exploration.
Collapse
Affiliation(s)
- Yoonha Lee
- Department of Pediatrics, Hallym University Dongtan Sacred Heart Hospital, Hwasung, Korea
| | - Young Ah Lee
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Min Ko
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Choong Ho Shin
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Jeong Lee
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| |
Collapse
|
10
|
Finn BP, Dattani MT. The molecular basis of hypoprolactinaemia. Rev Endocr Metab Disord 2024; 25:967-983. [PMID: 39417960 DOI: 10.1007/s11154-024-09906-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/19/2024]
Abstract
Hypoprolactinaemia is an endocrinopathy which is typically encountered as part of a combined pituitary hormone deficiency picture. The vast majority of genetic causes identified to date have been in the context of congenital hypopituitarism with multiple co-existent endocrinopathies. This is primarily with its closest hormonal relation, namely growth hormone. Acquired hypoprolactinaemia is generally rare in paediatric patients, and usually occurs together with other hormonal deficiencies. Congenital hypopituitarism occurs with an incidence of 1:4,000-10,000 cases and mutations in the following transcription factors account for the majority of documented genetic causes: PROP-1, POU1F1, LHX3/4 as well as documented case reports for a smaller subset of transcription factors and other molecules implicated in lactotroph development and prolactin secretion. Isolated prolactin deficiency has been described in a number of sporadic case reports in the literature, but no cases of mutations in the gene have been described to date. A range of genetic polymorphisms affecting multiple components of the prolactin signalling pathway have been identified in the literature, ranging from RNA spliceosome mutations (RNPC3) to loss of function mutations in IGSF-1. As paediatricians gain a greater understanding of the long-term ramifications of hypoprolactinaemia in terms of metabolic syndrome, type 2 diabetes mellitus and impaired fertility, the expectation is that clinicians will measure prolactin more frequently over time. Ultimately, we will encounter further reports of hypoprolactinaemia-related clinical presentations with further genetic mutations, in turn leading to a greater insight into the molecular basis of hypoprolactinaemia in terms of signalling pathways and downstream mediators. In the interim, the greatest untapped reserve of genetic causes remains within the phenotypic spectrum of congenital hypopituitarism.
Collapse
Affiliation(s)
- Bryan Padraig Finn
- Department of Paediatric Endocrinology, Great Ormond Street Children's Hospital, London, UK.
| | - Mehul T Dattani
- Department of Paediatric Endocrinology, Great Ormond Street Children's Hospital, London, UK
- Genetics and Genomic Medicine Research and Teaching Department, UCL GOS Institute of Child Health, London, UK
| |
Collapse
|
11
|
Martinez-Mayer J, Vishnopolska S, Perticarari C, Iglesias Garcia L, Hackbartt M, Martinez M, Zaiat J, Jacome-Alvarado A, Braslavsky D, Keselman A, Bergadá I, Marino R, Ramírez P, Pérez Garrido N, Ciaccio M, Di Palma MI, Belgorosky A, Forclaz MV, Benzrihen G, D'Amato S, Cirigliano ML, Miras M, Paez Nuñez A, Castro L, Mallea-Gil MS, Ballarino C, Latorre-Villacorta L, Casiello AC, Hernandez C, Figueroa V, Alonso G, Morin A, Guntsche Z, Lee H, Lee E, Song Y, Marti MA, Perez-Millan MI. Exome Sequencing Has a High Diagnostic Rate in Sporadic Congenital Hypopituitarism and Reveals Novel Candidate Genes. J Clin Endocrinol Metab 2024; 109:3196-3210. [PMID: 38717911 DOI: 10.1210/clinem/dgae320] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 06/23/2024]
Abstract
CONTEXT The pituitary gland is key for childhood growth, puberty, and metabolism. Pituitary dysfunction is associated with a spectrum of phenotypes, from mild to severe. Congenital hypopituitarism (CH) is the most commonly reported pediatric endocrine dysfunction, with an incidence of 1:4000, yet low rates of genetic diagnosis have been reported. OBJECTIVE We aimed to unveil the genetic etiology of CH in a large cohort of patients from Argentina. METHODS We performed whole exome sequencing of 137 unrelated cases of CH, the largest cohort examined with this method to date. RESULTS Of the 137 cases, 19.1% and 16% carried pathogenic or likely pathogenic variants in known and new genes, respectively, while 28.2% carried variants of uncertain significance. This high yield was achieved through the integration of broad gene panels (genes described in animal models and/or other disorders), an unbiased candidate gene screen with a new bioinformatics pipeline (including genes with high loss-of-function intolerance), and analysis of copy number variants. Three novel findings emerged. First, the most prevalent affected gene encodes the cell adhesion factor ROBO1. Affected children had a spectrum of phenotypes, consistent with a role beyond pituitary stalk interruption syndrome. Second, we found that CHD7 mutations also produce a phenotypic spectrum, not always associated with full CHARGE syndrome. Third, we add new evidence of pathogenicity in the genes PIBF1 and TBC1D32, and report 13 novel candidate genes associated with CH (eg, PTPN6, ARID5B). CONCLUSION Overall, these results provide an unprecedented insight into the diverse genetic etiology of hypopituitarism.
Collapse
Affiliation(s)
- Julian Martinez-Mayer
- Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Sebastian Vishnopolska
- Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Catalina Perticarari
- Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Lucia Iglesias Garcia
- Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Martina Hackbartt
- Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Marcela Martinez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, C1428EHA, Ciudad de Buenos Aires, Argentina
| | - Jonathan Zaiat
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, C1428EHA, Ciudad de Buenos Aires, Argentina
| | - Andrea Jacome-Alvarado
- Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| | - Debora Braslavsky
- Centro de Investigaciones "Dr. Cesar Bergadá" (CEDIE)-CONICET-FEI-División Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Ana Keselman
- Centro de Investigaciones "Dr. Cesar Bergadá" (CEDIE)-CONICET-FEI-División Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Ignacio Bergadá
- Centro de Investigaciones "Dr. Cesar Bergadá" (CEDIE)-CONICET-FEI-División Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Roxana Marino
- Servicio de Endocrinología-CONICET, Hospital de Pediatría Prof. Dr. J. P. Garrahan, C1245AAM, Buenos Aires, Argentina
| | - Pablo Ramírez
- Servicio de Endocrinología-CONICET, Hospital de Pediatría Prof. Dr. J. P. Garrahan, C1245AAM, Buenos Aires, Argentina
| | - Natalia Pérez Garrido
- Servicio de Endocrinología-CONICET, Hospital de Pediatría Prof. Dr. J. P. Garrahan, C1245AAM, Buenos Aires, Argentina
| | - Marta Ciaccio
- Servicio de Endocrinología-CONICET, Hospital de Pediatría Prof. Dr. J. P. Garrahan, C1245AAM, Buenos Aires, Argentina
| | - Maria Isabel Di Palma
- Servicio de Endocrinología-CONICET, Hospital de Pediatría Prof. Dr. J. P. Garrahan, C1245AAM, Buenos Aires, Argentina
| | - Alicia Belgorosky
- Servicio de Endocrinología-CONICET, Hospital de Pediatría Prof. Dr. J. P. Garrahan, C1245AAM, Buenos Aires, Argentina
| | - Maria Veronica Forclaz
- Servicio de Endocrinología Pediátrica, Hospital Nacional Profesor Alejandro Posadas, 1684, Buenos Aires, Argentina
| | - Gabriela Benzrihen
- Servicio de Endocrinología Pediátrica, Hospital Nacional Profesor Alejandro Posadas, 1684, Buenos Aires, Argentina
| | - Silvia D'Amato
- Servicio de Endocrinología Pediátrica, Hospital Nacional Profesor Alejandro Posadas, 1684, Buenos Aires, Argentina
| | - Maria Lujan Cirigliano
- Servicio de Endocrinología Pediátrica, Hospital Nacional Profesor Alejandro Posadas, 1684, Buenos Aires, Argentina
| | - Mirta Miras
- Hospital De Niños de la Santísima Trinidad, CP5000, Córdoba, Argentina
- Centro Privado de Endocrinologia Infanto Juvenil Crecer, CP5000, Cordoba, Argentina
| | - Alejandra Paez Nuñez
- Centro Privado de Endocrinologia Infanto Juvenil Crecer, CP5000, Cordoba, Argentina
| | - Laura Castro
- Hospital De Niños de la Santísima Trinidad, CP5000, Córdoba, Argentina
| | | | - Carolina Ballarino
- Servicio de Endocrinología, Hospital Militar Central, C1426BOS, Buenos Aires, Argentina
| | | | - Ana Clara Casiello
- Servicio de Endocrinología, Hospital General de Niños Pedro de Elizalde, C1270AAN, Buenos Aires, Argentina
| | - Claudia Hernandez
- Servicio de Endocrinología, Hospital General de Niños Pedro de Elizalde, C1270AAN, Buenos Aires, Argentina
| | - Veronica Figueroa
- Servicio de Endocrinología, Hospital General de Niños Pedro de Elizalde, C1270AAN, Buenos Aires, Argentina
| | - Guillermo Alonso
- Sección Endocrinología Pediátrica, Hospital Italiano, C1199ABB, Buenos Aires, Argentina
| | - Analia Morin
- Sala de Endocrinología, Hospital de Niños Sor Maria Ludovica de La Plata, B1904, La Plata, Argentina
| | | | - Hane Lee
- 3Billion Inc., 14th, 416 Teheran-ro, Gangnam-gu, Seoul, South Korea
| | - Eugene Lee
- 3Billion Inc., 14th, 416 Teheran-ro, Gangnam-gu, Seoul, South Korea
| | - Yongjun Song
- 3Billion Inc., 14th, 416 Teheran-ro, Gangnam-gu, Seoul, South Korea
| | - Marcelo Adrian Marti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, C1428EHA, Ciudad de Buenos Aires, Argentina
| | - Maria Ines Perez-Millan
- Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EHA, Buenos Aires, Argentina
| |
Collapse
|
12
|
Galichet C, Rizzoti K, Lovell-Badge R. Hypopituitarism in Sox3 null mutants correlates with altered NG2-glia in the median eminence and is influenced by aspirin and gut microbiota. PLoS Genet 2024; 20:e1011395. [PMID: 39325695 PMCID: PMC11426531 DOI: 10.1371/journal.pgen.1011395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/13/2024] [Indexed: 09/28/2024] Open
Abstract
The median eminence (ME), located at the base of the hypothalamus, is an essential centre of information exchange between the brain and the pituitary. We and others previously showed that mutations and duplications affecting the transcription factor SOX3/Sox3 result in hypopituitarism, and this is likely of hypothalamic origin. We demonstrate here that the absence of Sox3 predominantly affects the ME with phenotypes that first occur in juvenile animals, despite the embryonic onset of SOX3 expression. In the pituitary, reduction in hormone levels correlates with a lack of endocrine cell maturation. In parallel, ME NG2-glia renewal and oligodendrocytic differentiation potential are affected. We further show that low-dose aspirin treatment, which is known to affect NG2-glia, or changes in gut microbiota, rescue both proliferative defects and hypopituitarism in Sox3 mutants. Our study highlights a central role of NG2-glia for ME function during a transitional period of post-natal development and indicates their sensitivity to extrinsic signals.
Collapse
Affiliation(s)
- Christophe Galichet
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
- Neurobiological Research Facility, UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, London, United Kingdom
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
| | - Robin Lovell-Badge
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
13
|
Masser BE, Brinkmeier ML, Lin Y, Liu Q, Miyazaki A, Nayeem J, Cheung LYM. Gene Misexpression in a Smoc2+ve/ Sox2-Low Population in Juvenile Prop1-Mutant Pituitary Gland. J Endocr Soc 2024; 8:bvae146. [PMID: 39253355 PMCID: PMC11382140 DOI: 10.1210/jendso/bvae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Indexed: 09/11/2024] Open
Abstract
Mutations in the pituitary-specific transcription factor Prophet of Pit-1 (PROP1) are the most common genetic etiology of combined pituitary hormone deficiency (CPHD). CPHD is associated with short stature, attributable to growth hormone deficiency and/or thyroid-stimulating hormone deficiency, as well as hypothyroidism and infertility. Pathogenic lesions impair pituitary development and differentiation of endocrine cells. We performed single-cell RNA sequencing of pituitary cells from a wild-type and a Prop1-mutant P4 female mouse to elucidate population-specific differential gene expression. We observed a Smoc2+ve population that expressed low Sox2, which trajectory analyses suggest are a transitional cell state as stem cells differentiate into endocrine cells. We also detected ectopic expression of Sox21 in these cells in the Prop1df/df mutant. Prop1-mutant mice are known to overexpress Pou3f4, which we now show to be also enriched in this Smoc2+ve population. We sought to elucidate the role of Pou3f4 during pituitary development and to determine the contributions of Pou3f4 upregulation to pituitary disease by utilizing double-mutant mice lacking both Prop1 and Pou3f4. However, our data showed that Pou3f4 is not required for normal pituitary development and function. Double mutants further demonstrated that the upregulation of Pou3f4 was not causative for the overexpression of Sox21. These data indicate loss of Pou3f4 is not a potential cause of CPHD, and further studies may investigate the functional consequence of upregulation of Pou3f4 and Sox21, if any, in the novel Smoc2+ve cell population.
Collapse
Affiliation(s)
- Bailey E Masser
- Department of Human Genetics, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48105, USA
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yuxuan Lin
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Qin Liu
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Aya Miyazaki
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jannatun Nayeem
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Leonard Y M Cheung
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
14
|
Lyra A, Rodart IF, Barros L, Silva TSE, da Rocha AJ, Kochi C, Longui CA. Trio-based whole exome sequencing in patients with ectopic posterior pituitary. Front Pediatr 2024; 12:1334610. [PMID: 39156017 PMCID: PMC11327137 DOI: 10.3389/fped.2024.1334610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 07/03/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Ectopic posterior pituitary (EPP) is a rare congenital abnormality, sometimes associated with other midline defects, such as pituitary stalk interruption syndrome (PSIS), in which thin or absent pituitary stalk and anterior pituitary hypoplasia are combined to EPP. Most cases are sporadic, with few reports of familial cases, and many congenital hypopituitarism (CH) cases remain unsolved. Objective To search for candidate genes associated with this condition, we performed trio-based whole-exome sequencing (WES) on patients with EPP, including two familial cases. Methods This study included subjects with EPP and PSIS diagnosed by a simple MRI protocol (FAST1.2). We performed two distinct analyses in the trio-based WES. We looked for previously described genes associated with pituitary development. Next, we investigated the whole exome for variants inherited in a pattern consistent with a monogenic etiology. Results Ten families were evaluated; eight were composed of a child with EPP and healthy parents, one has two affected siblings, and one family has a son and mother with EPP. When analyzing the previously described candidate variants associated with pituitary development, we found variants in GLI2 and FGFR1 in three families. We also found six other variants of interest in three patients: KMT2A, GALR3, RTN4R, SEMA3A, NIPBL, and DSCAML1. Conclusion The analysis allowed us to find previously reported and not reported GLI2 variants, all inherited from healthy parents, which reinforces the incomplete penetrance pattern of GLI2 variants in the development of EPP and draws attention to possible future functional studies of those variants that have a recurrent expression in CH. We also found novel FGFR1 and SEMA3A variants that suggest an oligogenic mechanism in PSIS and EPP, as seen in patients with hypogonadotropic hypogonadism. We report the first case of a patient with Wiedemann-Steiner syndrome and PSIS, suggesting that the KMT2A gene may be related to pituitary development. Furthermore, the trios' analysis allowed us to find five other variants of interest. Future investigations may clarify the roles of these variants in the etiology of EPP and PSIS.
Collapse
Affiliation(s)
- Arthur Lyra
- Pediatric Endocrinology Unit, Pediatric Department, Irmandade da Santa Casa de Misericórdia de São Paulo and Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Itatiana Ferreira Rodart
- Pediatric Endocrinology Unit, Pediatric Department, Irmandade da Santa Casa de Misericórdia de São Paulo and Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Lara Barros
- Pediatric Endocrinology Unit, Pediatric Department, Irmandade da Santa Casa de Misericórdia de São Paulo and Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Tatiane Sousa e Silva
- Pediatric Endocrinology Unit, Pediatric Department, Irmandade da Santa Casa de Misericórdia de São Paulo and Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Antônio José da Rocha
- Department of Radiology, Irmandade da Santa Casa de Misericórdia de São Paulo and Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Cristiane Kochi
- Pediatric Endocrinology Unit, Pediatric Department, Irmandade da Santa Casa de Misericórdia de São Paulo and Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| | - Carlos Alberto Longui
- Pediatric Endocrinology Unit, Pediatric Department, Irmandade da Santa Casa de Misericórdia de São Paulo and Santa Casa de Sao Paulo School of Medical Sciences, São Paulo, Brazil
| |
Collapse
|
15
|
Cheetham T, Wood C. Paediatric thyroid disease. Clin Endocrinol (Oxf) 2024. [PMID: 39072866 DOI: 10.1111/cen.15110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/30/2024]
Abstract
The spectrum of thyroid disorders presenting to paediatricians is different to that seen by adult physicians. Referrals reflect cases detected by the neonatal screening programme for congenital hypothyroidism and many of the inherited defects of thyroid hormone generation or action will be manifest in early life. Autoimmune thyroid disease can be particularly challenging to manage in the young and the potential impact of thyroid status on neurodevelopment and schooling are key considerations throughout childhood and adolescence.
Collapse
Affiliation(s)
- Timothy Cheetham
- Newcastle University and Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Claire Wood
- Newcastle University and Great North Children's Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
16
|
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
|
17
|
Castets S, Albarel F, Bachelot A, Brun G, Bouligand J, Briet C, Bui Quoc E, Cazabat L, Chabbert-Buffet N, Christin-Maitre S, Courtillot C, Cuny T, De Filippo G, Donadille B, Illouz F, Pellegrini I, Reznik Y, Saveanu A, Teissier N, Touraine P, Vantyghem MC, Vergier J, Léger J, Brue T, Reynaud R. Position statement on the diagnosis and management of congenital pituitary deficiency in adults: The French National Diagnosis and Treatment Protocol (NDTP). ANNALES D'ENDOCRINOLOGIE 2024; 85:327-339. [PMID: 38452869 DOI: 10.1016/j.ando.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Pituitary deficiency, or hypopituitarism, is a rare chronic disease. It is defined by insufficient synthesis of one or more pituitary hormones (growth hormone, TSH, ACTH, LH-FSH, prolactin), whether or not associated with arginine vasopressin deficiency (formerly known as diabetes insipidus). In adult patients, it is usually acquired (notably during childhood), but can also be congenital, due to abnormal pituitary development. The present study focuses on congenital pituitary deficiency in adults, from diagnosis to follow-up, including special situations such as pregnancy or the elderly. The clinical presentation is highly variable, ranging from isolated deficit to multiple deficits, which may be part of a syndromic form or not. Diagnosis is based on a combination of clinical, biological (assessment of all hormonal axes), radiological (brain and hypothalamic-pituitary MRI) and genetic factors. Treatment consists in hormonal replacement therapy, adapted according to the period of life and the deficits, which may be progressive. Comorbidities, risk of complications and acute decompensation, and the impact on fertility and quality of life all require adaptative multidisciplinary care and long-term monitoring.
Collapse
Affiliation(s)
- Sarah Castets
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France.
| | - Frédérique Albarel
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Conception, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Anne Bachelot
- IE3M, ICAN, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Centre de Référence des Pathologies Gynécologiques Rares, hôpital Pitié-Salpêtrière, AP-HP, Paris, France; Sorbonne université, Paris, France
| | - Gilles Brun
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Hôpital Européen, Pôle imagerie médicale, 13003, Marseille, France
| | - Jérôme Bouligand
- Molecular Genetic, Pharmacogenetic and Hormonology, Kremlin-Bicêtre Hospital, Paris-Saclay University, AP-HP, Le Kremlin-Bicêtre, France
| | - Claire Briet
- Département d'endocrinologie-diabétologie nutrition, Centre de référence des maladies rares de la Thyroïde et des Récepteurs Hormonaux, Endo-ERN centre for rare endocrine diseases, CHU d'Angers, 4, rue larrey, 49100 Angers, France; Laboratoire MITOVASC, UMR CNRS 6015, Inserm 1083, Université d'Angers, rue Roger Amsler, 49100 Angers, France
| | - Emmanuelle Bui Quoc
- Ophthalmology Department, Robert-Debré University Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Laure Cazabat
- Department of Endocrinology, Diabetology and Nutrition, Ambroise Paré Hospital, AP-HP, UVSQ, Boulogne-Billancourt, France
| | - Nathalie Chabbert-Buffet
- Department of Gynecology and Obstetrics, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, 75020 Paris, France
| | - Sophie Christin-Maitre
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Carine Courtillot
- IE3M, ICAN, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Centre de Référence des Pathologies Gynécologiques Rares, hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Thomas Cuny
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Gianpaolo De Filippo
- Service d'endocrinologie et diabétologie pédiatrique, centre de référence des maladies endocriniennes de la croissance et du développement, hôpital universitaire Robert-Debré, université Paris Cité, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Bruno Donadille
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Frédéric Illouz
- Département d'endocrinologie-diabétologie nutrition, Centre de référence des maladies rares de la Thyroïde et des Récepteurs Hormonaux, Endo-ERN centre for rare endocrine diseases, CHU d'Angers, 4, rue larrey, 49100 Angers, France; Laboratoire MITOVASC, UMR CNRS 6015, Inserm 1083, Université d'Angers, rue Roger Amsler, 49100 Angers, France
| | - Isabelle Pellegrini
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Conception, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Yves Reznik
- Endocrinology and Diabetes Department, CHU Côte de Nacre and Unicaen, Caen Cedex, France
| | - Alexandru Saveanu
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France
| | - Natacha Teissier
- Department of Pediatric Otolaryngology, Robert Debré Hospital, AP-HP Nord, Paris, France
| | - Philippe Touraine
- Service d'endocrinologie et médecine de la reproduction, centre de maladies endocrinennes rares de la croissance et du développement, médecine-hôpital Pitié-Salpêtrière, Sorbonne université, Paris, France
| | - Marie-Christine Vantyghem
- Service d'endocrinologie, diabétologie et maladies métaboliques, CHRU de Lille, rue Polonowski, Lille cedex, France
| | - Julia Vergier
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Julianne Léger
- Service d'endocrinologie et diabétologie pédiatrique, centre de référence des maladies endocriniennes de la croissance et du développement, hôpital universitaire Robert-Debré, université Paris Cité, Assistance publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1141, Paris, France
| | - Thierry Brue
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France; Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Inserm, MMG, Laboratory of Molecular Biology, Hospital La Conception, Aix-Marseille University, AP-HM, Marseille, France
| | - Rachel Reynaud
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France; Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Inserm, MMG, Laboratory of Molecular Biology, Hospital La Conception, Aix-Marseille University, AP-HM, Marseille, France
| |
Collapse
|
18
|
Fleseriu M, Christ-Crain M, Langlois F, Gadelha M, Melmed S. Hypopituitarism. Lancet 2024; 403:2632-2648. [PMID: 38735295 DOI: 10.1016/s0140-6736(24)00342-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 05/14/2024]
Abstract
Partial or complete deficiency of anterior or posterior pituitary hormone production leads to central hypoadrenalism, central hypothyroidism, hypogonadotropic hypogonadism, growth hormone deficiency, or arginine vasopressin deficiency depending on the hormones affected. Hypopituitarism is rare and likely to be underdiagnosed, with an unknown but rising incidence and prevalence. The most common cause is compressive growth or ablation of a pituitary or hypothalamic mass. Less common causes include genetic mutations, hypophysitis (especially in the context of cancer immunotherapy), infiltrative and infectious disease, and traumatic brain injury. Clinical features vary with timing of onset, cause, and number of pituitary axes disrupted. Diagnosis requires measurement of basal circulating hormone concentrations and confirmatory hormone stimulation testing as needed. Treatment is aimed at replacement of deficient hormones. Increased mortality might persist despite treatment, particularly in younger patients, females, and those with arginine vasopressin deficiency. Patients with complex diagnoses, pregnant patients, and adolescent pituitary-deficient patients transitioning to adulthood should ideally be managed at a pituitary tumour centre of excellence.
Collapse
Affiliation(s)
- Maria Fleseriu
- Department of Medicine, Division of Endocrinology, Diabetes and Clinical Nutrition, Oregon Health and Science University, Portland, OR, USA; Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, USA; Pituitary Center, Oregon Health and Science University, Portland, OR, USA.
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Fabienne Langlois
- Department of Medicine, Division of Endocrinology, Centre intégré universitaire de santé et de services sociaux de l'Estrie, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Mônica Gadelha
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Shlomo Melmed
- Department of Medicine and Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
19
|
List EO, Basu R, Berryman DE, Duran-Ortiz S, Martos-Moreno GÁ, Kopchick JJ. Common and uncommon mouse models of growth hormone deficiency. Endocr Rev 2024:bnae017. [PMID: 38853618 DOI: 10.1210/endrev/bnae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/22/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Mouse models of growth hormone deficiency (GHD) have provided important tools for uncovering the various actions of GH. Nearly 100 years of research using these mouse lines has greatly enhanced our knowledge of the GH/IGF-1 axis. Some of the shared phenotypes of the five "common" mouse models of GHD include reduced body size, delayed sexual maturation, decreased fertility, reduced muscle mass, increased adiposity, and enhanced insulin sensitivity. Since these common mouse lines outlive their normal-sized littermates - and have protection from age-associated disease - they have become important fixtures in the aging field. On the other hand, the twelve "uncommon" mouse models of GHD described herein have tremendously divergent health outcomes ranging from beneficial aging phenotypes (similar to those described for the common models) to extremely detrimental features (such as improper development of the CNS, numerous sensory organ defects, and embryonic lethality). Moreover, advancements in next generation sequencing technologies have led to the identification of an expanding array of genes that are recognized as causative agents to numerous rare syndromes with concomitant GHD. Accordingly, this review provides researchers with a comprehensive up-to-date collection of the common and uncommon mouse models of GHD that have been used to study various aspects of physiology and metabolism associated with multiple forms of GHD. For each mouse line presented, the closest comparable human syndromes are discussed providing important parallels to the clinic.
Collapse
Affiliation(s)
- Edward O List
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Specialty Medicine, Heritage College of Osteopathic Medicine, Athens Ohio
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens Ohio
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
| | - Gabriel Á Martos-Moreno
- Department of Endocrinology & Pediatrics, Hospital Infantil Universitario Niño Jesús, IIS La Princesa & Universidad Autónoma de Madrid. CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens Ohio, 45701, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens Ohio
| |
Collapse
|
20
|
Bangalore Krishna K, Fuqua JS, Witchel SF. Hypogonadotropic Hypogonadism. Endocrinol Metab Clin North Am 2024; 53:279-292. [PMID: 38677870 DOI: 10.1016/j.ecl.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Delayed puberty is defined as absent testicular enlargement in boys or breast development in girls at an age that is 2 to 2.5 SDS later than the mean age at which these events occur in the population (traditionally, 14 years in boys and 13 years in girls). One cause of delayed/absent puberty is hypogonadotropic hypogonadism (HH), which refers to inadequate hypothalamic/pituitary function leading to deficient production of sex steroids in males and females. Individuals with HH typically have normal gonads, and thus HH differs from hypergonadotropic hypogonadism, which is associated with primary gonadal insufficiency.
Collapse
Affiliation(s)
- Kanthi Bangalore Krishna
- Division of Pediatric Endocrinology and Diabetes, UPMC Childrens Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - John S Fuqua
- Division of Pediatric Endocrinology, Indiana University School of Medicine, 705 Riley Hospital Drive, Room 5960, Indianapolis, IN 46202, USA
| | - Selma F Witchel
- Division of Pediatric Endocrinology and Diabetes, UPMC Childrens Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| |
Collapse
|
21
|
Basu R, Goswami S, Sengupta N, Baidya A, Mondal S, Swapnil K, Deb R, Khare VR, Datta J. Rare coexistence of hypopituitarism with osteogenesis imperfecta - A double-trouble for bone. Bone Rep 2024; 21:101768. [PMID: 38706521 PMCID: PMC11068520 DOI: 10.1016/j.bonr.2024.101768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Osteogenesis imperfecta (OI) commonly involving defects in COL1A1 and COL1A2 is a rare hereditary disease of bone fragility affecting 6-7 per 100,000 population. On the other hand, hypopituitarism is a separate entity that manifests with reduced levels of pituitary hormones. The cooccurrence of these two is seldom reported previously in literature as a deviation from Occam's razor. Here, we reported a case of pathological fracture in a 31-year-old male who had blue sclera and secondary adrenal insufficiency, hypogonadotropic hypogonadism, and growth hormone deficiency along with primary autoimmune hypothyroidism. Diagnosis of OI was suggested by heterozygous missense variant in exon 40 of the COL1A2 gene (chr7: g.94423092G > A; Depth: 99×) that resulted in the amino acid substitution of Serine for Glycine at codon 847. Replacement of glucocorticoid, levothyroxine, and testosterone was started along with antiresorptive therapy for better bone health outcomes.
Collapse
Affiliation(s)
- Rajdeep Basu
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Soumik Goswami
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Nilanjan Sengupta
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Arjun Baidya
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Sunetra Mondal
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Kumar Swapnil
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Rajat Deb
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Vibhu Ranjan Khare
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Joydip Datta
- Department of Endocrinology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| |
Collapse
|
22
|
Martinez-Mayer J, Brinkmeier ML, O'Connell SP, Ukagwu A, Marti MA, Miras M, Forclaz MV, Benzrihen MG, Cheung LYM, Camper SA, Ellsworth BS, Raetzman LT, Pérez-Millán MI, Davis SW. Knockout mice with pituitary malformations help identify human cases of hypopituitarism. Genome Med 2024; 16:75. [PMID: 38822427 PMCID: PMC11140907 DOI: 10.1186/s13073-024-01347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Congenital hypopituitarism (CH) and its associated syndromes, septo-optic dysplasia (SOD) and holoprosencephaly (HPE), are midline defects that cause significant morbidity for affected people. Variants in 67 genes are associated with CH, but a vast majority of CH cases lack a genetic diagnosis. Whole exome and whole genome sequencing of CH patients identifies sequence variants in genes known to cause CH, and in new candidate genes, but many of these are variants of uncertain significance (VUS). METHODS The International Mouse Phenotyping Consortium (IMPC) is an effort to establish gene function by knocking-out all genes in the mouse genome and generating corresponding phenotype data. We used mouse embryonic imaging data generated by the Deciphering Mechanisms of Developmental Disorders (DMDD) project to screen 209 embryonic lethal and sub-viable knockout mouse lines for pituitary malformations. RESULTS Of the 209 knockout mouse lines, we identified 51 that have embryonic pituitary malformations. These genes not only represent new candidates for CH, but also reveal new molecular pathways not previously associated with pituitary organogenesis. We used this list of candidate genes to mine whole exome sequencing data of a cohort of patients with CH, and we identified variants in two unrelated cases for two genes, MORC2 and SETD5, with CH and other syndromic features. CONCLUSIONS The screening and analysis of IMPC phenotyping data provide proof-of-principle that recessive lethal mouse mutants generated by the knockout mouse project are an excellent source of candidate genes for congenital hypopituitarism in children.
Collapse
Affiliation(s)
- Julian Martinez-Mayer
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Sean P O'Connell
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA
| | - Arnold Ukagwu
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Marcelo A Marti
- Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mirta Miras
- Hospital De Niños de La Santísima Trinidad, Córdoba, Argentina
| | - Maria V Forclaz
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Maria G Benzrihen
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Leonard Y M Cheung
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
- Department of Physiology and Biophyscis, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Buffy S Ellsworth
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Lori T Raetzman
- Department of Molecular and Integrative Physiology, University of Illinois, Champaign-Urbana, Urbana, IL, 61801, USA
| | - Maria I Pérez-Millán
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina.
| | - Shannon W Davis
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA.
| |
Collapse
|
23
|
Attia N, Moussa K, Altwaim A, Al-Agha AE, Amir AA, Almuhareb A. Tackling access and payer barriers for growth hormone therapy in Saudi Arabia: a consensus statement for the Saudi Working Group for Pediatric Endocrinology. J Pediatr Endocrinol Metab 2024; 37:387-399. [PMID: 38547465 DOI: 10.1515/jpem-2024-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/13/2024] [Indexed: 05/05/2024]
Abstract
Prompt diagnosis and early treatment are key goals to optimize the outcomes of children with growth hormone deficiency (GHD) and attain the genetically expected adult height. Nonetheless, several barriers can hinder prompt diagnosis and treatment of GHD, including payer-related issues. In Saudi Arabia, moderate-to-severe short stature was reported in 13.1 and 11.7 % of healthy boys and girls, respectively. Several access and payer barriers can face pediatric endocrinologists during the diagnosis and treatment of GHD in Saudi Arabia. Insurance coverage policies can restrict access to diagnostic tests for GHD and recombinant human growth hormone (rhGH) due to their high costs and lack of gold-standard criteria. Some insurance policies may limit the duration of treatment with rhGH or the amount of medication covered per month. This consensus article gathered the insights of pediatric endocrinologists from Saudi Arabia to reflect the access and payer barriers to the diagnostic tests and treatment options of children with short stature. We also discussed the current payer-related challenges endocrinologists face during the investigations of children with short stature. The consensus identified potential strategies to overcome these challenges and optimize patient management.
Collapse
Affiliation(s)
- Najya Attia
- Department of Pediatric Endocrinology, 4917 King Abdulaziz Medical City/King Saud bin Abdulaziz University for Health Sciences/King Abdullah International Medical Research Center , Jeddah, Saudi Arabia
| | | | - Abdulaziz Altwaim
- King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- International Diabetes Care Center, Jeddah, Saudi Arabia
| | - Abdulmoein Eid Al-Agha
- Pediatric Department, Pediatric Endocrinology & Diabetes Section, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | | | | |
Collapse
|
24
|
Ge X, Weis K, Raetzman L. Glycoprotein hormone subunit alpha 2 (GPHA2): A pituitary stem cell-expressed gene associated with NOTCH2 signaling. Mol Cell Endocrinol 2024; 586:112163. [PMID: 38246572 DOI: 10.1016/j.mce.2024.112163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024]
Abstract
NOTCH2 is expressed in pituitary stem cells and is necessary for stem cell maintenance, proliferation, and differentiation. However, the pathways NOTCH2 engages to affect pituitary development remain unclear. In this study, we hypothesized that glycoprotein hormone subunit A2 (GPHA2), a corneal stem cell factor and ligand for the thyroid stimulating hormone receptor (TSHR), is downstream of NOTCH2 signaling. We found Gpha2 is expressed in quiescent pituitary stem cells by RNAscope in situ hybridization and scRNA seq. In Notch2 conditional knockout pituitaries, Gpha2 mRNA is reduced compared with control littermates. We then investigated the possible functions of GPHA2. Pituitaries treated with a GPHA2 peptide do not have a change in proliferation. However, in dissociated adult pituitary cells, GPHA2 increased pCREB expression and this induction was reversed by co-treatment with a TSHR inhibitor. These data suggest GPHA2 is a NOTCH2 related stem cell factor that activates TSHR signaling, potentially impacting pituitary development.
Collapse
Affiliation(s)
- Xiyu Ge
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, 524 Burrill Hall, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Karen Weis
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, 524 Burrill Hall, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Lori Raetzman
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, 524 Burrill Hall, 407 South Goodwin Avenue, Urbana, IL, 61801, USA; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 W. Gregory Drive, Urbana, IL, 61801, USA.
| |
Collapse
|
25
|
Balen AH, Tamblyn J, Skorupskaite K, Munro MG. A comprehensive review of the new FIGO classification of ovulatory disorders. Hum Reprod Update 2024; 30:355-382. [PMID: 38412452 DOI: 10.1093/humupd/dmae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND The World Health Organization (WHO) system for the classification of disorders of ovulation was produced 50 years ago and, by international consensus, has been updated by the International Federation of Gynecology and Obstetrics (FIGO). OBJECTIVE AND RATIONALE This review outlines in detail each component of the FIGO HyPO-P (hypothalamic, pituitary, ovarian, PCOS) classification with a concise description of each cause, and thereby provides a systematic method for diagnosis and management. SEARCH METHODS We searched the published articles in the PubMed database in the English-language literature until October 2022, containing the keywords ovulatory disorders; ovulatory dysfunction; anovulation, and each subheading in the FIGO HyPO-P classification. We did not include abstracts or conference proceedings because the data are usually difficult to assess. OUTCOMES We present the most comprehensive review of all disorders of ovulation, published systematically according to the logical FIGO classification. WIDER IMPLICATIONS Improving the diagnosis of an individual's ovulatory dysfunction will significantly impact clinical practice by enabling healthcare practitioners to make a precise diagnosis and plan appropriate management.
Collapse
Affiliation(s)
- Adam H Balen
- Leeds Centre for Reproductive Medicine, The University of Leeds, Leeds, UK
| | - Jennifer Tamblyn
- Leeds Centre for Reproductive Medicine, The University of Leeds, Leeds, UK
| | | | - Malcolm G Munro
- Department of Obstetrics and Gynecology, The University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
26
|
Castets S, Thomas-Teinturier C, Villanueva C, Amsellem J, Barat P, Brun G, Quoc EB, Carel JC, De Filippo GP, Kipnis C, Martinerie L, Vergier J, Saveanu A, Teissier N, Coutant R, Léger J, Reynaud R. Diagnosis and management of congenital hypopituitarism in children. Arch Pediatr 2024; 31:165-171. [PMID: 38538470 DOI: 10.1016/j.arcped.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 04/07/2024]
Abstract
Hypopituitarism (or pituitary deficiency) is a rare disease with an estimated prevalence of between 1/16,000 and 1/26,000 individuals, defined by insufficient production of one or several anterior pituitary hormones (growth hormone [GH], thyroid-stimulating hormone [TSH], adrenocorticotropic hormone [ACTH], luteinizing hormone [LH], follicle-stimulating hormone [FSH], prolactin), in association or not with diabetes insipidus (antidiuretic hormone [ADH] deficiency). While in adults hypopituitarism is mostly an acquired disease (tumors, irradiation), in children it is most often a congenital condition, due to abnormal pituitary development. Clinical symptoms vary considerably from isolated to combined deficiencies and between syndromic and non-syndromic forms. Early signs are non-specific but should not be overlooked. Diagnosis is based on a combination of clinical, laboratory (testing of all hormonal axes), imaging (brain magnetic resonance imaging [MRI] with thin slices centered on the hypothalamic-pituitary region), and genetic (next-generation sequencing of genes involved in pituitary development, array-based comparative genomic hybridization, and/or genomic analysis) findings. Early brain MRI is crucial in neonates or in cases of severe hormone deficiency for differential diagnosis and to inform syndrome workup. This article presents recommendations for hormone replacement therapy for each of the respective deficient axes. Lifelong follow-up with an endocrinologist is required, including in adulthood, with multidisciplinary management for patients with syndromic forms or comorbidities. Treatment objectives include alleviating symptoms, preventing comorbidities and acute complications, and optimal social and educational integration.
Collapse
Affiliation(s)
- Sarah Castets
- Assistance Publique Hôpitaux de Marseille, Hôpital la Timone, service de pédiatrie multidisciplinaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France.
| | - Cécile Thomas-Teinturier
- Assistance Publique-Hôpitaux de Paris, Université Paris-Saclay, Hôpital Bicêtre, service d'Endocrinologie et diabète de l'enfant, Le Kremlin Bicêtre, France; INSERM UMR 1018, Equipe d'épidémiologie des radiations, CESP, 94800 Villejuif, France
| | - Carine Villanueva
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, Bron, France
| | - Jessica Amsellem
- CHU Angers, Service d'endocrinologie diabétologie pédiatrique, Angers, France
| | - Pascal Barat
- Centre hospitalier universitaire de Bordeaux, unite d'endocrinologie pédiatrique, Bordeaux, France
| | - Gilles Brun
- Hôpital Européen, neuroradiologie, Marseille, France
| | - Emmanuel Bui Quoc
- Assistance Publique-Hôpitaux de Paris, Hôpital universitaire Robert Debré, service d'ophtalmologie, Paris, France
| | - Jean-Claude Carel
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1141, Paris, France
| | - Gian Paolo De Filippo
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France
| | - Clara Kipnis
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France
| | - Laetitia Martinerie
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France
| | - Julia Vergier
- Assistance Publique Hôpitaux de Marseille, Hôpital la Timone, service de pédiatrie multidisciplinaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France
| | - Alexandru Saveanu
- Assistance Publique Hôpitaux de Marseille, Hôpital Conception, laboratoire de bioloie moléculaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France; Aix Marseille Université, INSERM, MMG, U 1251, Marseille, France
| | - Natacha Teissier
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service de Chirurgie ORL et cervico-faciale pédiatrique, Paris, France
| | - Régis Coutant
- CHU Angers, Service d'endocrinologie diabétologie pédiatrique, Angers, France
| | - Juliane Léger
- Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Hôpital universitaire Robert Debré, service d'Endocrinologie et Diabétologie Pédiatrique, Centre de Référence des Maladies Endocriniennes de la Croissance et du Développement, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1141, Paris, France
| | - Rachel Reynaud
- Assistance Publique Hôpitaux de Marseille, Hôpital la Timone, service de pédiatrie multidisciplinaire, Centre de Référence des Maladies Rares d'Origine Hypophysaire HYPO, Marseille, France; Aix Marseille Université, INSERM, MMG, U 1251, Marseille, France
| |
Collapse
|
27
|
Ruggiero-Ruff RE, Le BH, Villa PA, Lainez NM, Athul SW, Das P, Ellsworth BS, Coss D. Single-Cell Transcriptomics Identifies Pituitary Gland Changes in Diet-Induced Obesity in Male Mice. Endocrinology 2024; 165:bqad196. [PMID: 38146776 PMCID: PMC10791142 DOI: 10.1210/endocr/bqad196] [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: 07/31/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023]
Abstract
Obesity is a chronic disease with increasing prevalence worldwide. Obesity leads to an increased risk of heart disease, stroke, and diabetes, as well as endocrine alterations, reproductive disorders, changes in basal metabolism, and stress hormone production, all of which are regulated by the pituitary. In this study, we performed single-cell RNA sequencing of pituitary glands from male mice fed control and high-fat diet (HFD) to determine obesity-mediated changes in pituitary cell populations and gene expression. We determined that HFD exposure is associated with dramatic changes in somatotrope and lactotrope populations, by increasing the proportion of somatotropes and decreasing the proportion of lactotropes. Fractions of other hormone-producing cell populations remained unaffected. Gene expression changes demonstrated that in HFD, somatotropes became more metabolically active, with increased expression of genes associated with cellular respiration, and downregulation of genes and pathways associated with cholesterol biosynthesis. Despite a lack of changes in gonadotrope fraction, genes important in the regulation of gonadotropin hormone production were significantly downregulated. Corticotropes and thyrotropes were the least affected in HFD, while melanotropes exhibited reduced proportion. Lastly, we determined that changes in plasticity and gene expression were associated with changes in hormone levels. Serum prolactin was decreased corresponding to reduced lactotrope fraction, while lower luteinizing hormone and follicle-stimulating hormone in the serum corresponded to a decrease in transcription and translation. Taken together, our study highlights diet-mediated changes in pituitary gland populations and gene expression that play a role in altered hormone levels in obesity.
Collapse
Affiliation(s)
- Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences; School of Medicine, University of California, Riverside, CA 92521, USA
| | - Brandon H Le
- Institute for Integrative Genome Biology Bioinformatics Core Facility, University of California, Riverside, CA 92521, USA
| | - Pedro A Villa
- Division of Biomedical Sciences; School of Medicine, University of California, Riverside, CA 92521, USA
| | - Nancy M Lainez
- Division of Biomedical Sciences; School of Medicine, University of California, Riverside, CA 92521, USA
| | - Sandria W Athul
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL 62901, USA
| | - Pratyusa Das
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL 62901, USA
| | - Buffy S Ellsworth
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL 62901, USA
| | - Djurdjica Coss
- Division of Biomedical Sciences; School of Medicine, University of California, Riverside, CA 92521, USA
| |
Collapse
|
28
|
Kırkgöz T, Gürsoy S, Acar S, Nalbantoğlu Ö, Özkaya B, Anıl Korkmaz H, Hazan F, Özkan B. Genetic diagnosis of congenital hypopituitarism in Turkish patients by a target gene panel: novel pathogenic variants in GHRHR, GLI2, LHX4 and POU1F1 genes. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 68:e220254. [PMID: 37948564 PMCID: PMC10916835 DOI: 10.20945/2359-4292-2022-0254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 05/02/2023] [Indexed: 11/12/2023]
Abstract
Objective Congenital hypopituitarism (CH) is a rare disease characterized by one or more hormone deficiencies of the pituitary gland. To date, many genes have been associated with CH. In this study, we identified the allelic variant spectrum of 11 causative genes in Turkish patients with CH. Materials and methods This study included 47 patients [21 girls (44.6%) and 26 boys (55.4%)] from 45 families. To identify the genetic etiology, we screened 11 candidate genes associated with CH using next-generation sequencing. To confirm and detect the status of the specific familial variant in relatives, Sanger sequencing was also performed. Results We identified 12 possible pathogenic variants in GHRHR, GH1, GLI2, PROP-1, POU1F1, and LHX4 in 11 patients (23.4%), of which six were novel variants: two in GHRHR, two in POU1F1, one in GLI2, and one in LHX4. In all patients, these variants were most frequently found in GLI2, followed by PROP-1 and GHRHR. Conclusion Genetic causes were determined in only 23.4% of all patients with CH and 63% of molecularly diagnosed patients (7/11) from consanguineous families. Despite advances in genetics, we were unable to identify the genetic etiology of most patients with CH, suggesting the effect of unknown genes or environmental factors. More genetic studies are necessary to understand the etiology of CH.
Collapse
Affiliation(s)
- Tarık Kırkgöz
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey,
| | - Semra Gürsoy
- Division of Pediatric Genetics, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Sezer Acar
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Özlem Nalbantoğlu
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Beyhan Özkaya
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Hüseyin Anıl Korkmaz
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Filiz Hazan
- Department of Medical Genetics, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Behzat Özkan
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| |
Collapse
|
29
|
Ho KKY, Kaiser UB, Chanson P, Gadelha M, Wass J, Nieman L, Little A, Aghi MK, Raetzman L, Post K, Raverot G, Borowsky AD, Erickson D, Castaño JP, Laws ER, Zatelli MC, Sisco J, Esserman L, Yuen KCJ, Reincke M, Melmed S. Pituitary adenoma or neuroendocrine tumour: the need for an integrated prognostic classification. Nat Rev Endocrinol 2023; 19:671-678. [PMID: 37592077 DOI: 10.1038/s41574-023-00883-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 08/19/2023]
Abstract
In the 2022 fifth edition of the WHO Classification of Endocrine Tumours and of Central Nervous System Tumours, pituitary adenomas are reclassified as neuroendocrine tumours (NETs). This change confers an oncology label to neoplasms that are overwhelmingly benign. A comprehensive clinical classification schema is required to guide prognosis, therapy and outcomes for all patients with pituitary adenomas. Pituitary adenomas and NETs exhibit some morphological and ultrastructural similarities. However, unlike NETs, pituitary adenomas are highly prevalent, yet indolent and rarely become malignant. This Perspective presents the outcomes of an interdisciplinary international workshop that addressed the merit and clinical implications of the classification change of pituitary adenoma to NET. Many non-histological factors provide mechanistic insight and influence the prognosis and treatment of pituitary adenoma. We recommend the development of a comprehensive classification that integrates clinical, genetic, biochemical, radiological, pathological and molecular information for all anterior pituitary neoplasms.
Collapse
Affiliation(s)
- Ken K Y Ho
- The Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- The University of New South Wales, Sydney, New South Wales, Australia.
| | - Ursula B Kaiser
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Phillippe Chanson
- Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Monica Gadelha
- Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Lynnette Nieman
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | | | - Manish K Aghi
- University of California, San Francisco, San Francisco, CA, USA
| | - Lori Raetzman
- University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Kalmon Post
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gerald Raverot
- Hospices Civils de Lyon, Groupement Hospitalier Est, Université Claude Bernard Lyon 1, Bron, France
| | | | | | - Justo P Castaño
- Maimónides Biomedical Research Institute of Córdoba, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | | | | | - Jill Sisco
- The Acromegaly Community, Grove, OK, USA
| | - Laura Esserman
- University of California, San Francisco, San Francisco, CA, USA
| | - Kevin C J Yuen
- Barrow Neurological Institute, Phoenix, AZ, USA
- University of Arizona College of Medicine and Creighton School of Medicine, Phoenix, AZ, USA
| | - Martin Reincke
- Klinikum der Universität, Ludwig-Maximilians-Universität, München, Germany
| | | |
Collapse
|
30
|
Gregory LC, Cionna C, Cerbone M, Dattani MT. Identification of genetic variants and phenotypic characterization of a large cohort of patients with congenital hypopituitarism and related disorders. Genet Med 2023; 25:100881. [PMID: 37165954 DOI: 10.1016/j.gim.2023.100881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023] Open
Abstract
PURPOSE Congenital hypopituitarism (CH) disorders are phenotypically variable. Variants in multiple genes are associated with these disorders, with variable penetrance and inheritance. METHODS We screened a large cohort (N = 1765) of patients with or at risk of CH using Sanger sequencing, selected according to phenotype, and conducted next-generation sequencing (NGS) in 51 families within our cohort. We report the clinical, hormonal, and neuroradiological phenotypes of patients with variants in known genes associated with CH. RESULTS We identified variants in 178 patients: GH1/GHRHR (51 patients of 414 screened), PROP1 (17 of 253), POU1F1 (15 of 139), SOX2 (13 of 59), GLI2 (7 of 106), LHX3/LHX4 (8 of 110), HESX1 (8 of 724), SOX3 (9 of 354), OTX2 (5 of 59), SHH (2 of 64), and TCF7L1, KAL1, FGFR1, and FGF8 (2 of 585, respectively). NGS identified 26 novel variants in 35 patients (from 24 families). Magnetic resonance imaging showed prevalent hypothalamo-pituitary abnormalities, present in all patients with PROP1, GLI2, SOX3, HESX1, OTX2, LHX3, and LHX4 variants. Normal hypothalamo-pituitary anatomy was reported in 24 of 121, predominantly those with GH1, GHRHR, POU1F1, and SOX2 variants. CONCLUSION We identified variants in 10% (178 of 1765) of our CH cohort. NGS has revolutionized variant identification, and careful phenotypic patient characterization has improved our understanding of CH. We have constructed a flow chart to guide genetic analysis in these patients, which will evolve upon novel gene discoveries.
Collapse
Affiliation(s)
- Louise C Gregory
- Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Cecilia Cionna
- Pediatric Unit, Department of Mother and Child Health, G. Salesi Children's Hospital, Ancona, Italy
| | - Manuela Cerbone
- Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Department of Endocrinology, Great Ormond Street Hospital for Children, Great Ormond Street, United Kingdom
| | - Mehul T Dattani
- Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Department of Endocrinology, Great Ormond Street Hospital for Children, Great Ormond Street, United Kingdom.
| |
Collapse
|
31
|
Kodytková A, Amaratunga SA, Zemková D, Maratová K, Dušátková P, Plachý L, Průhová Š, Koloušková S, Lebl J. SALL4 Phenotype in Four Generations of One Family: An Interplay of the Upper Limb, Kidneys, and the Pituitary. Horm Res Paediatr 2023; 97:203-210. [PMID: 37611564 DOI: 10.1159/000531996] [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: 11/29/2022] [Accepted: 07/01/2023] [Indexed: 08/25/2023] Open
Abstract
INTRODUCTION The SALL4 gene encodes a transcription factor that is essential for early embryonic cellular differentiation of the epiblast and primitive endoderm. It is required for the development of neural tissue, kidney, heart, and limbs. Pathogenic SALL4 variants cause Duane-radial ray syndrome (Okihiro syndrome), acro-renal-ocular syndrome, and Holt-Oram syndrome. We report a family with vertical transmission of a SALL4 pathogenic variant leading to radial hypoplasia and kidney dystopia in several generations with additional growth hormone deficiency (GHD) in the proband. CASE PRESENTATION Our male proband was born at the 39th week of gestation. He was born small for gestational age (SGA; birth weight 2,550 g, -2.2 SDS; length 47 cm, -2.0 SDS). He had bilateral asymmetrical radial ray malformation (consisting of radial hypoplasia, ulnar flexure, and bilateral aplasia of the thumb) and pelvic kidney dystopia, but no cardiac malformations, clubfoot, ocular coloboma, or Duane anomaly. He was examined for progressive short stature at the age of 3.9 years, where his IGF-1 was 68 μg/L (-1.0 SD), and growth hormone (GH) after stimulation 6.2 μg/L. Other pituitary hormones were normal. A brain CT revealed normal morphology of the cerebral midline and the pituitary. He had a dental anomaly - a central mandibular ectopic canine. MRI could not be done due to the presence of metal after multiple corrective plastic surgeries of his hands. His mother's and father's heights are 152.3 cm (-2.4 SD) and 177.8 cm (-0.4 SD), respectively. His father has a milder malformation of the forearm. The affected paternal grandfather (height 164 cm; -2.3 SD) has a radial ray defect with missing opposition of the thumb. The family reports a similar phenotype of radial dysplasia in the paternal grandfather's mother. The proband started GH therapy at age 6.5 years when his height was 109 cm (-2.8 SDS) and he experienced catch-up growth as expected in GHD. Puberty started spontaneously at the age of 12.5 years. At age 13, his height was 158.7 cm (-0.2 SDS). Whole-exome sequencing revealed a nonsense variant in the SALL4 gene c.1717C>T (p.Arg573Ter) in the proband, his father, and paternal grandfather. CONCLUSION This is the first observation of a patient with a congenital upper limb defect due to a pathogenic SALL4 variant who has isolated GHD with no apparent cerebral or facial midline anomaly and has been successfully treated with growth hormone.
Collapse
Affiliation(s)
- Aneta Kodytková
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Shenali Anne Amaratunga
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Daniela Zemková
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Klára Maratová
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Petra Dušátková
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Lukáš Plachý
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Štěpánka Průhová
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Stanislava Koloušková
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jan Lebl
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia,
| |
Collapse
|
32
|
Ferreira NGBP, Madeira JLO, Gergics P, Kertsz R, Marques JM, Trigueiro NSS, Benedetti AFF, Azevedo BV, Fernandes BHV, Bissegatto DD, Biscotto IP, Fang Q, Ma Q, Ozel AB, Li J, Camper SA, Jorge AAL, Mendonça BB, Arnhold IJP, Carvalho LR. Homozygous CDH2 variant may be associated with hypopituitarism without neurological disorders. Endocr Connect 2023; 12:e220473. [PMID: 37166408 PMCID: PMC10388658 DOI: 10.1530/ec-22-0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/11/2023] [Indexed: 05/12/2023]
Abstract
Context Congenital hypopituitarism is a genetically heterogeneous condition. Whole exome sequencing (WES) is a promising approach for molecular diagnosis of patients with this condition. Objectives The aim of this study is to conduct WES in a patient with congenital hypopituitarism born to consanguineous parents, CDH2 screening in a cohort of patients with congenital hypopituitarism, and functional testing of a novel CDH2 variant. Design Genomic DNA from a proband and her consanguineous parents was analyzed by WES. Copy number variants were evaluated. The genetic variants were filtered for population frequency (ExAC, 1000 genomes, gnomAD, and ABraOM), in silico prediction of pathogenicity, and gene expression in the pituitary and/or hypothalamus. Genomic DNA from 145 patients was screened for CDH2 by Sanger sequencing. Results One female patient with deficiencies in growth hormone, thyroid-stimulating hormone, adrenocorticotropic hormone, luteinizing hormone, and follicle-stimulating hormone and ectopic posterior pituitary gland contained a rare homozygous c.865G>A (p.Val289Ile) variant in CDH2. To determine whether the p.Val289Ile variant in CDH2 affects cell adhesion properties, we stably transfected L1 fibroblast lines, labeled the cells with lipophilic dyes, and quantified aggregation. Large aggregates formed in cells expressing wildtype CDH2, but aggregation was impaired in cells transfected with variant CDH2 or non-transfected. Conclusion A homozygous CDH2 allelic variant was found in one hypopituitarism patient, and the variant impaired cell aggregation function in vitro. No disease-causing variants were found in 145 other patients screened for CDH2 variants. Thus, CDH2 is a candidate gene for hypopituitarism that needs to be tested in different populations. Significance statement A female patient with hypopituitarism was born from consanguineous parents and had a homozygous, likely pathogenic, CDH2 variant that impairs cell aggregation in vitro. No other likely pathogenic variants in CDH2 were identified in 145 hypopituitarism patients.
Collapse
Affiliation(s)
- Nathalia G B P Ferreira
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Joao L O Madeira
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Peter Gergics
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Renata Kertsz
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Juliana M Marques
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Nicholas S S Trigueiro
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | | | - Bruna V Azevedo
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Bianca H V Fernandes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Universidade de São Paulo, Zebrafish Facility, São Paulo, São Paulo, Brazil
| | - Debora D Bissegatto
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Isabela P Biscotto
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Qing Fang
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Qianyi Ma
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Asye B Ozel
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Jun Li
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Sally A Camper
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genética, Laboratório de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Berenice B Mendonça
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Ivo J P Arnhold
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Luciani R Carvalho
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM42, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| |
Collapse
|
33
|
Cheung LYM, Menage L, Rizzoti K, Hamilton G, Dumontet T, Basham K, Daly AZ, Brinkmeier ML, Masser BE, Treier M, Cobb J, Delogu A, Lovell-Badge R, Hammer GD, Camper SA. Novel Candidate Regulators and Developmental Trajectory of Pituitary Thyrotropes. Endocrinology 2023; 164:bqad076. [PMID: 37183548 PMCID: PMC10227867 DOI: 10.1210/endocr/bqad076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
The pituitary gland regulates growth, metabolism, reproduction, the stress response, uterine contractions, lactation, and water retention. It secretes hormones in response to hypothalamic input, end organ feedback, and diurnal cues. The mechanisms by which pituitary stem cells are recruited to proliferate, maintain quiescence, or differentiate into specific cell types, especially thyrotropes, are not well understood. We used single-cell RNA sequencing in juvenile P7 mouse pituitary cells to identify novel factors in pituitary cell populations, with a focus on thyrotropes and rare subtypes. We first observed cells coexpressing markers of both thyrotropes and gonadotropes, such as Pou1f1 and Nr5a1. This was validated in vivo by both immunohistochemistry and lineage tracing of thyrotropes derived from Nr5a1-Cre; mTmG mice and demonstrates that Nr5a1-progenitors give rise to a proportion of thyrotropes during development. Our data set also identifies novel factors expressed in pars distalis and pars tuberalis thyrotropes, including the Shox2b isoform in all thyrotropes and Sox14 specifically in Pou1f1-negative pars tuberalis thyrotropes. We have therefore used single-cell transcriptomics to determine a novel developmental trajectory for thyrotropes and potential novel regulators of thyrotrope populations.
Collapse
Affiliation(s)
- Leonard Y M Cheung
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lucy Menage
- School of Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Karine Rizzoti
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Greg Hamilton
- Department of Biological Sciences, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Typhanie Dumontet
- Training Program in Organogenesis, Center for Cell Plasticity and Organ Design, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kaitlin Basham
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Current affiliation: Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandre Z Daly
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
- Current affiliation is Vanguard, Valley Forge, PA 19482, USA
| | | | - Bailey E Masser
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mathias Treier
- Max Delbrϋck Center for Molecular Medicine (MDC), 13092 Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - John Cobb
- Department of Biological Sciences, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Alessio Delogu
- School of Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
34
|
Camper SA, Smith C, Kitzman JO. Disruption of RNA Splicing Is an Important Contributor to Congenital Hypopituitarism and Other Human Genetic Diseases. Endocrinology 2023; 164:bqad039. [PMID: 36857601 PMCID: PMC10282917 DOI: 10.1210/endocr/bqad039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/03/2023]
Affiliation(s)
- Sally A Camper
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109-5618, USA
| | - Cathy Smith
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109-5618, USA
| | - Jacob O Kitzman
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109-5618, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109-5618, USA
| |
Collapse
|
35
|
Cassin J, Stamou MI, Keefe KW, Sung KE, Bojo CC, Tonsfeldt KJ, Rojas RA, Ferreira Lopes V, Plummer L, Salnikov KB, Keefe DL, Ozata M, Genel M, Georgopoulos NA, Hall JE, Crowley WF, Seminara SB, Mellon PL, Balasubramanian R. Heterozygous mutations in SOX2 may cause idiopathic hypogonadotropic hypogonadism via dominant-negative mechanisms. JCI Insight 2023; 8:e164324. [PMID: 36602867 PMCID: PMC9977424 DOI: 10.1172/jci.insight.164324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Pathogenic SRY-box transcription factor 2 (SOX2) variants typically cause severe ocular defects within a SOX2 disorder spectrum that includes hypogonadotropic hypogonadism. We examined exome-sequencing data from a large, well-phenotyped cohort of patients with idiopathic hypogonadotropic hypogonadism (IHH) for pathogenic SOX2 variants to investigate the underlying pathogenic SOX2 spectrum and its associated phenotypes. We identified 8 IHH individuals harboring heterozygous pathogenic SOX2 variants with variable ocular phenotypes. These variant proteins were tested in vitro to determine whether a causal relationship between IHH and SOX2 exists. We found that Sox2 was highly expressed in the hypothalamus of adult mice and colocalized with kisspeptin 1 (KISS1) expression in the anteroventral periventricular nucleus of adult female mice. In vitro, shRNA suppression of mouse SOX2 protein in Kiss-expressing cell lines increased the levels of human kisspeptin luciferase (hKiss-luc) transcription, while SOX2 overexpression repressed hKiss-luc transcription. Further, 4 of the identified SOX2 variants prevented this SOX2-mediated repression of hKiss-luc. Together, these data suggest that pathogenic SOX2 variants contribute to both anosmic and normosmic forms of IHH, attesting to hypothalamic defects in the SOX2 disorder spectrum. Our study describes potentially novel mechanisms contributing to SOX2-related disease and highlights the necessity of SOX2 screening in IHH genetic evaluation irrespective of associated ocular defects.
Collapse
Affiliation(s)
- Jessica Cassin
- Department of Obstetrics, Gynecology, and Reproductive Sciences; Center for Reproductive Science and Medicine; and
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
| | - Maria I. Stamou
- Massachusetts General Hospital Harvard Center for Reproductive Medicine and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kimberly W. Keefe
- Center for Infertility and Reproductive Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Kaitlin E. Sung
- Department of Obstetrics, Gynecology, and Reproductive Sciences; Center for Reproductive Science and Medicine; and
| | - Celine C. Bojo
- Department of Obstetrics, Gynecology, and Reproductive Sciences; Center for Reproductive Science and Medicine; and
| | - Karen J. Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences; Center for Reproductive Science and Medicine; and
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
| | - Rebecca A. Rojas
- Massachusetts General Hospital Harvard Center for Reproductive Medicine and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Vanessa Ferreira Lopes
- Massachusetts General Hospital Harvard Center for Reproductive Medicine and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lacey Plummer
- Center for Infertility and Reproductive Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Kathryn B. Salnikov
- Massachusetts General Hospital Harvard Center for Reproductive Medicine and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David L. Keefe
- Center for Infertility and Reproductive Surgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - Myron Genel
- Section of Pediatric Endocrinology, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Neoklis A. Georgopoulos
- Division of Endocrinology, Department of Medicine, University of Patras Medical School, Patras, Greece
| | - Janet E. Hall
- National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - William F. Crowley
- Endocrine Unit, Department of Medicine, and Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephanie B. Seminara
- Massachusetts General Hospital Harvard Center for Reproductive Medicine and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Pamela L. Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences; Center for Reproductive Science and Medicine; and
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
| | - Ravikumar Balasubramanian
- Massachusetts General Hospital Harvard Center for Reproductive Medicine and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Bando H, Brinkmeier ML, Castinetti F, Fang Q, Lee MS, Saveanu A, Albarel F, Dupuis C, Brue T, Camper SA. Heterozygous variants in SIX3 and POU1F1 cause pituitary hormone deficiency in mouse and man. Hum Mol Genet 2023; 32:367-385. [PMID: 35951005 PMCID: PMC9851746 DOI: 10.1093/hmg/ddac192] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/22/2022] [Accepted: 08/09/2022] [Indexed: 01/24/2023] Open
Abstract
Congenital hypopituitarism is a genetically heterogeneous condition that is part of a spectrum disorder that can include holoprosencephaly. Heterozygous mutations in SIX3 cause variable holoprosencephaly in humans and mice. We identified two children with neonatal hypopituitarism and thin pituitary stalk who were doubly heterozygous for rare, likely deleterious variants in the transcription factors SIX3 and POU1F1. We used genetically engineered mice to understand the disease pathophysiology. Pou1f1 loss-of-function heterozygotes are unaffected; Six3 heterozygotes have pituitary gland dysmorphology and incompletely ossified palate; and the Six3+/-; Pou1f1+/dw double heterozygote mice have a pronounced phenotype, including pituitary growth through the palate. The interaction of Pou1f1 and Six3 in mice supports the possibility of digenic pituitary disease in children. Disruption of Six3 expression in the oral ectoderm completely ablated anterior pituitary development, and deletion of Six3 in the neural ectoderm blocked the development of the pituitary stalk and both anterior and posterior pituitary lobes. Six3 is required in both oral and neural ectodermal tissues for the activation of signaling pathways and transcription factors necessary for pituitary cell fate. These studies clarify the mechanism of SIX3 action in pituitary development and provide support for a digenic basis for hypopituitarism.
Collapse
Affiliation(s)
- Hironori Bando
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Frederic Castinetti
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares de l’hypophyse HYPO, Marseille, France
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille, Maladies Rares (MarMaRa), Marseille, France
| | - Qing Fang
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Mi-Sun Lee
- Michigan Neuroscience Institute, Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Alexandru Saveanu
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares de l’hypophyse HYPO, Marseille, France
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille, Maladies Rares (MarMaRa), Marseille, France
| | - Frédérique Albarel
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares de l’hypophyse HYPO, Marseille, France
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille, Maladies Rares (MarMaRa), Marseille, France
| | - Clémentine Dupuis
- Department of Pediatrics, Centre Hospitalier Universitaire de Grenoble-Alpes, site Nord, Hôpital Couple Enfants, Grenoble, France
| | - Thierry Brue
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares de l’hypophyse HYPO, Marseille, France
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille, Maladies Rares (MarMaRa), Marseille, France
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Winningham AH, Camper SA. Pituitary Stem Cell Regulation by Zeb2 and BMP Signaling. Endocrinology 2023; 164:bqad016. [PMID: 36683433 PMCID: PMC10091485 DOI: 10.1210/endocr/bqad016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023]
Abstract
Epithelial to mesenchymal transition (EMT) is important for many developing organs, and for wound healing, fibrosis, and cancer. Pituitary stem cells undergo an EMT-like process as they migrate and initiate differentiation, but little is known about the input of signaling pathways or the genetic hierarchy of the transcriptional cascade. Prop1 mutant stem cells fail to undergo changes in cellular morphology, migration, and transition to the Pou1f1 lineage. We used Prop1 mutant mice to identify the changes in gene expression that are affiliated with EMT-like processes. BMP and TGF-β family gene expression was reduced in Prop1 mutants and Elf5, a transcription factor that characteristically suppresses EMT, had elevated expression. Genes involved in cell-cell contact such as cadherins and claudins were elevated in Prop1 mutants. To establish the genetic hierarchy of control, we manipulated gene expression in pituitary stem cell colonies. We determined that the EMT inducer, Zeb2, is necessary for robust BMP signaling and repression of Elf5. We demonstrated that inhibition of BMP signaling affects expression of target genes in the Id family, but it does not affect expression of other EMT genes. Zeb2 is necessary for expression of the SHH effector gene Gli2. However, knock down of Gli2 has little effect on the EMT-related genes, suggesting that it acts through a separate pathway. Thus, we have established the genetic hierarchy involved in the transition of pituitary stem cells to differentiation.
Collapse
Affiliation(s)
- Amanda H Winningham
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109-5618, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109-5618, USA
| |
Collapse
|
38
|
Zheng X, Zhang C, Zheng D, Guo Q, Maierhaba M, Xue L, Zeng X, Wu Y, Gao W. An original cuproptosis-related genes signature effectively influences the prognosis and immune status of head and neck squamous cell carcinoma. Front Genet 2023; 13:1084206. [PMID: 36685880 PMCID: PMC9845781 DOI: 10.3389/fgene.2022.1084206] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Recently, a non-apoptotic cell death pathway that is dependent on the presence of copper ions was proposed, named as cuproptosis. Cuproptosis have been found to have a strong association with the clinical progression and prognosis of several cancers. Head and neck squamous cell carcinoma (HNSC) are among the most common malignant tumors, with a 5-year relative survival rate ranging between 40% and 50%. The underlying mechanisms and clinical significance of cuproptosis-related genes (CRGs) in HNSC progression have not been clarified. Methods: In this study, expression pattern, biological functions, Immunohistochemistry (IHC), gene variants and immune status were analyzed to investigate the effects of CRGs on HNSC progression. Moreover, a 12-CRGs signature and nomogram were also constructed for prognosis prediction of HNSC. Results: The results revealed that some CRGs were dysregulated, had somatic mutations, and CNV in HNSC tissues. Among them, ISCA2 was found to be upregulated in HNSC and was strongly correlated with the overall survival (OS) of HNSC patients (HR = 1.13 [1.01-1.26], p-value = 0.0331). Functionally, CRGs was mainly associated with the TCA cycle, cell cycle, iron-sulfur cluster assembly, p53 signaling pathway, chemical carcinogenesis, and carbon metabolism in cancer. A 12-CRGs signature for predicting the OS was constructed which included, CAT, MTFR1L, OXA1L, POLE, NTHL1, DNA2, ATP7B, ISCA2, GLRX5, NDUFA1, and NDUFB2. This signature showed good prediction performance on the OS (HR = 5.3 [3.4-8.2], p-value = 3.4e-13) and disease-specific survival (HR = 6.4 [3.6-11], p-value = 2.4e-10). Furthermore, 12-CRGs signature significantly suppressed the activation of CD4+ T cells and antigen processing and presentation. Finally, a nomogram based on a 12-CRGs signature and clinical features was constructed which showed a significantly adverse effect on OS (HR = 1.061 [1.042-1.081], p-value = 1.6e-10) of HNSC patients. Conclusion: This study reveals the association of CRGs with the progression of HNSC based on multi-omics analysis. The study of CRGs is expected to improve clinical diagnosis, immunotherapeutic responsiveness and prognosis prediction of HNSC.
Collapse
Affiliation(s)
- Xiwang Zheng
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Defei Zheng
- Department of Hematology/Oncology, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qingbo Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mijiti Maierhaba
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lingbin Xue
- Department of Otolaryngology Head and Neck Surgery, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
- Shenzhen Institute of Otolaryngology and Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
| | - Xianhai Zeng
- Department of Otolaryngology Head and Neck Surgery, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
- Shenzhen Institute of Otolaryngology and Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
| | - Yongyan Wu
- Department of Otolaryngology Head and Neck Surgery, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
- Shenzhen Institute of Otolaryngology and Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
| | - Wei Gao
- Department of Otolaryngology Head and Neck Surgery, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
- Shenzhen Institute of Otolaryngology and Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
39
|
Sun C, Chen S. Disease-causing mutations in genes encoding transcription factors critical for photoreceptor development. Front Mol Neurosci 2023; 16:1134839. [PMID: 37181651 PMCID: PMC10172487 DOI: 10.3389/fnmol.2023.1134839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Photoreceptor development of the vertebrate visual system is controlled by a complex transcription regulatory network. OTX2 is expressed in the mitotic retinal progenitor cells (RPCs) and controls photoreceptor genesis. CRX that is activated by OTX2 is expressed in photoreceptor precursors after cell cycle exit. NEUROD1 is also present in photoreceptor precursors that are ready to specify into rod and cone photoreceptor subtypes. NRL is required for the rod fate and regulates downstream rod-specific genes including the orphan nuclear receptor NR2E3 which further activates rod-specific genes and simultaneously represses cone-specific genes. Cone subtype specification is also regulated by the interplay of several transcription factors such as THRB and RXRG. Mutations in these key transcription factors are responsible for ocular defects at birth such as microphthalmia and inherited photoreceptor diseases such as Leber congenital amaurosis (LCA), retinitis pigmentosa (RP) and allied dystrophies. In particular, many mutations are inherited in an autosomal dominant fashion, including the majority of missense mutations in CRX and NRL. In this review, we describe the spectrum of photoreceptor defects that are associated with mutations in the above-mentioned transcription factors, and summarize the current knowledge of molecular mechanisms underlying the pathogenic mutations. At last, we deliberate the outstanding gaps in our understanding of the genotype-phenotype correlations and outline avenues for future research of the treatment strategies.
Collapse
Affiliation(s)
- Chi Sun
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Chi Sun,
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
40
|
Moalla M, Mnif-Feki M, Safi W, Charfi N, Mejdoub-Rekik N, Abid M, Hadj Kacem F, Hadj Kacem H. Analysis of ProP1 Gene in a Cohort of Tunisian Patients with Congenital Combined Pituitary Hormone Deficiency. J Clin Med 2022; 11:7525. [PMID: 36556141 PMCID: PMC9787973 DOI: 10.3390/jcm11247525] [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: 11/03/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Non-syndromic combined pituitary hormone deficiency (CPHD) occurs due to defects in transcription factors that govern early pituitary development and the specification of hormone-producing cells. The most common mutations are in the Prophet of Pit-1 (ProP1) gene. This work aims to (1) report findings of genetic analyses of Tunisian patients with non-syndromic CPHD and (2) describe their phenotype patterns and their evolution through life. Methods: Fifteen patients from twelve unrelated families with variable clinical phenotypes were included after excluding autoimmune and acquired forms of non-syndromic CPHD. Detailed pedigree charts and auxological, hormonal, radiological, and therapeutic details were recorded. Sanger sequencing was performed, and sequences were analyzed with a specific focus on coding and splice site regions of the ProP1 gene. Retained variants were classified using several in silico pathogenicity prediction tools and the VarSome platform. Results: We identified the common p.Arg73Cys mutation in seven patients from four unrelated pedigrees. We found a novel homozygous mutation (c.340C>T) in one sporadic case. This mutation generates a truncated ProP1 protein, predicted to be non-functional, lacking the last 112 codons (p.(Gln114Ter)). We confirmed by polymerase chain reaction (PCR) the absence of large exon deletions or insertions in the remaining sporadic patients (7/8). Conclusions: We report two mutations {one newly identified [p.(Gln114Ter)] and one previously reported (p.Arg73Cys)} in five unrelated Tunisian families with non-syndromic CPHD. This work is of clinical importance as it reports the high frequency of the p.Arg73Cys mutation in Tunisian CPHD families. Our study also illuminated the involvement of novel gene(s) in the emergence of non-syndromic CPHD.
Collapse
Affiliation(s)
- Mariam Moalla
- Laboratory of Molecular and Cellular Screening Processes, Center of Biotechnology of Sfax, Sfax 3018, Tunisia
- Laboratory of Human Molecular Genetics, Faculty of Medicine of Sfax, University of Sfax, Sfax 3029, Tunisia
| | - Mouna Mnif-Feki
- Endocrinology Department, Hedi Chaker Hospital, Sfax 3029, Tunisia
| | - Wajdi Safi
- Endocrinology Department, Hedi Chaker Hospital, Sfax 3029, Tunisia
| | - Nadia Charfi
- Endocrinology Department, Hedi Chaker Hospital, Sfax 3029, Tunisia
| | | | - Mohamed Abid
- Endocrinology Department, Hedi Chaker Hospital, Sfax 3029, Tunisia
| | - Faten Hadj Kacem
- Endocrinology Department, Hedi Chaker Hospital, Sfax 3029, Tunisia
| | - Hassen Hadj Kacem
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| |
Collapse
|
41
|
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
|
42
|
Lin SZ, Ma QJ, Pang QM, Chen QD, Wang WQ, Li JY, Zhang SL. Novel compound heterozygous variants in the LHX3 gene caused combined pituitary hormone deficiency: A case report. World J Clin Cases 2022; 10:11486-11492. [PMID: 36387827 PMCID: PMC9649574 DOI: 10.12998/wjcc.v10.i31.11486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/14/2022] [Accepted: 09/29/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Combined pituitary hormone deficiency 3 (CPHD3; OMIM: 221750) is caused by mutations within the LHX3 gene (OMIM: 600577), which located on the subtelomeric region of chromosome 9 at band 9q34.3, has seven coding exons and six introns. LIM homeobox (LHX) 3 protein is the key regulator of pituitary development in fetal life.
CASE SUMMARY We have diagnosed and treate an 11-year-old boy with combined pituitary hormone deficiency (CPHD). The main clinical manifestations were pituitary hormone deficiency, hydrocele of the tunica vaginalis, pituitary dwarfism, gonadal dysplasia, micropenis, clonic convulsion, and mild facial dysmorphic features. We collected peripheral blood from the patient, the patient's older brother, as well as their parents, and sequenced them by using high-throughput whole-exosome sequencing, which was verified by Sanger sequencing. The results showed that there were two compound heterozygous variants of c.613G>C (p.V205L) and c.220T>C (p.C74R) in the LHX3 gene. c.613G>C (p.V205L) was inherited from his mother and c.220T>C (p.C74R) from his father. His brother also has both variants and symptoms.
CONCLUSION This study reported ununreported genetic mutations of LHX3, and recorded the treatment process of the patients, providing data for the diagnosis and treatment of CPHD.
Collapse
Affiliation(s)
- Shuang-Zhu Lin
- Diagnosis and Treatment Center for Children, The First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Qi-Ji Ma
- The First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, Jilin Province, China
| | - Qi-Ming Pang
- Department of Neonatology, Hainan Women and Children's Medical Center, Haikou 570100, Hainan Province, China
| | - Qian-Dui Chen
- Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Wan-Qi Wang
- Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Jia-Yi Li
- Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Su-Li Zhang
- Department of Neonatology, Hainan Women and Children's Medical Center, Haikou 570100, Hainan Province, China
| |
Collapse
|
43
|
Hietamäki J, Kärkinen J, Iivonen AP, Vaaralahti K, Tarkkanen A, Almusa H, Huopio H, Hero M, Miettinen PJ, Raivio T. Presentation and diagnosis of childhood-onset combined pituitary hormone deficiency: A single center experience from over 30 years. EClinicalMedicine 2022; 51:101556. [PMID: 35875813 PMCID: PMC9304914 DOI: 10.1016/j.eclinm.2022.101556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Childhood-onset combined pituitary hormone deficiency (CPHD) has a wide spectrum of etiologies and genetic causes for congenital disease. We aimed to describe the clinical spectrum and genetic etiologies of CPHD in a single tertiary center and estimate the population-level incidence of congenital CPHD. METHODS The retrospective clinical cohort comprised 124 CPHD patients (48 with congenital CPHD) treated at the Helsinki University Hospital (HUH) Children's Hospital between 1985 and 2018. Clinical data were collected from the patient charts. Whole exome sequencing was performed in 21 patients with congenital CPHD of unknown etiology. FINDINGS The majority (61%;76/124) of the patients had acquired CPHD, most frequently due to craniopharyngiomas and gliomas. The estimated incidence of congenital CPHD was 1/16 000 (95%CI, 1/11 000-1/24 000). The clinical presentation of congenital CPHD in infancy included prolonged/severe neonatal hypoglycaemia, prolonged jaundice, and/or micropenis/bilateral cryptorchidism in 23 (66%) patients; despite these clinical cues, only 76% of them were referred to endocrine investigations during the first year of life. The median delay between the first violation of the growth screening rules and the initiation of GH Rx treatment among all congenital CPHD patients was 2·2 years, interquartile range 1·2-3·7 years. Seven patients harbored pathogenic variants in PROP1, SOX3, TBC1D32, OTX2, and SOX2, and one patient carried a likely pathogenic variant in SHH (c.676G>A, p.(Ala226Thr)). INTERPRETATION Our study suggests that congenital CPHD can occur in 1/16 000 children, and that patients frequently exhibit neonatal cues of hypopituitarism and early height growth deflection. These results need to be corroborated in future studies and might inform clinical practice. FUNDING Päivikki and Sakari Sohlberg Foundation, Biomedicum Helsinki Foundation, and Emil Aaltonen Foundation research grants.
Collapse
Affiliation(s)
- Johanna Hietamäki
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
| | - Juho Kärkinen
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
| | - Anna-Pauliina Iivonen
- Department of Physiology, Medicum Unit, Faculty of Medicine, and Stem Cells and Metabolism Research Program, Research Programs Unit, University of Helsinki, Helsinki 00014, Finland
| | - Kirsi Vaaralahti
- Department of Physiology, Medicum Unit, Faculty of Medicine, and Stem Cells and Metabolism Research Program, Research Programs Unit, University of Helsinki, Helsinki 00014, Finland
| | - Annika Tarkkanen
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
- Department of Physiology, Medicum Unit, Faculty of Medicine, and Stem Cells and Metabolism Research Program, Research Programs Unit, University of Helsinki, Helsinki 00014, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Hanna Huopio
- Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Matti Hero
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
| | - Päivi J. Miettinen
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
| | - Taneli Raivio
- Helsinki University Hospital, New Children's Hospital, Pediatric Research Center, Helsinki 00014, Finland
- Department of Physiology, Medicum Unit, Faculty of Medicine, and Stem Cells and Metabolism Research Program, Research Programs Unit, University of Helsinki, Helsinki 00014, Finland
- Corresponding author at: Faculty of Medicine University of Helsinki, Medicum/Physiology, P.O. Box 63 (Haartmaninkatu 8), FI-00014 Helsinki, Finland.
| |
Collapse
|
44
|
Sertedaki A, Tatsi EB, Vasilakis IA, Fylaktou I, Nikaina E, Iacovidou N, Siahanidou T, Kanaka-Gantenbein C. Whole Exome Sequencing Points towards a Multi-Gene Synergistic Action in the Pathogenesis of Congenital Combined Pituitary Hormone Deficiency. Cells 2022; 11:cells11132088. [PMID: 35805171 PMCID: PMC9265573 DOI: 10.3390/cells11132088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/21/2022] [Accepted: 06/28/2022] [Indexed: 12/21/2022] Open
Abstract
Combined pituitary hormone deficiency (CPHD) is characterized by deficiency of growth hormone and at least one other pituitary hormone. Pathogenic variants in more than 30 genes expressed during the development of the head, hypothalamus, and/or pituitary have been identified so far to cause genetic forms of CPHD. However, the etiology of around 85% of the cases remains unknown. The aim of this study was to unveil the genetic etiology of CPHD due to congenital hypopituitarism employing whole exome sequencing (WES) in two newborn patients, initially tested and found to be negative for PROP1, LHX3, LHX4 and HESX1 pathogenic variants by Sanger sequencing and for copy number variations by MLPA. In this study, the application of WES in these CPHD newborns revealed the presence of three different heterozygous gene variants in each patient. Specifically in patient 1, the variants BMP4; p.Ala42Pro, GNRH1; p.Arg73Ter and SRA1; p.Gln32Glu, and in patient 2, the SOX9; p.Val95Ile, HS6ST1; p.Arg306Gln, and IL17RD; p.Pro566Ser were identified as candidate gene variants. These findings further support the hypothesis that CPHD constitutes an oligogenic rather than a monogenic disease and that there is a genetic overlap between CPHD and congenital hypogonadotropic hypogonadism.
Collapse
Affiliation(s)
- Amalia Sertedaki
- Division of Endocrinology, Diabetes and Metabolism, Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.B.T.); (I.A.V.); (I.F.); (C.K.-G.)
- Correspondence:
| | - Elizabeth Barbara Tatsi
- Division of Endocrinology, Diabetes and Metabolism, Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.B.T.); (I.A.V.); (I.F.); (C.K.-G.)
| | - Ioannis Anargyros Vasilakis
- Division of Endocrinology, Diabetes and Metabolism, Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.B.T.); (I.A.V.); (I.F.); (C.K.-G.)
| | - Irene Fylaktou
- Division of Endocrinology, Diabetes and Metabolism, Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.B.T.); (I.A.V.); (I.F.); (C.K.-G.)
| | - Eirini Nikaina
- Neonatology Unit, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.N.); (T.S.)
| | - Nicoletta Iacovidou
- Department of Neonatology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Tania Siahanidou
- Neonatology Unit, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.N.); (T.S.)
| | - Christina Kanaka-Gantenbein
- Division of Endocrinology, Diabetes and Metabolism, Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.B.T.); (I.A.V.); (I.F.); (C.K.-G.)
| |
Collapse
|
45
|
Griffero M, Benedetti AFF, Pérez M, Carvalho L, Jorge A, Latronico AC, Mendonca B, Arnhold I, Mericq V. Novel OTX2 loss of function variant associated with congenital hypopituitarism without eye abnormalities. J Pediatr Endocrinol Metab 2022; 35:831-835. [PMID: 35320640 DOI: 10.1515/jpem-2021-0719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/18/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The normal development of the pituitary gland requires multiple induction signals and transcription factors encoded by more than 30 genes, including OTX2. OTX2 mutations have been described with eye abnormalities and variable congenital hypopituitarism, but rarely with hypopituitarism without ocular manifestations. CASE PRESENTATION We report a girl with hypopituitarism associated with pituitary hypoplasia and pituitary stalk atrophy, without ocular manifestations. NGS revealed a novel heterozygous mutation in OTX2 c.426dupC:p.(Ser143Leufs*2). CONCLUSIONS Mutations in the transcription factor OTX2 have been associated with ocular, craniofacial, and pituitary development anomalies. Here we describe a novel mutation in OTX2 associated with hypopituitarism without an ocular phenotype.
Collapse
Affiliation(s)
- Mariana Griffero
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Anna Flavia Figueredo Benedetti
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marcela Pérez
- Department of Ophthalmology, Clínica Las Condes and Hospital Salvador, Santiago, Chile
| | - Luciani Carvalho
- Disciplina de Endocrinologia e Metabologia, Departamento de Clinica Medica, LIM/42, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alexander Jorge
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil.,Disciplina de Endocrinologia e Metabologia, Departamento de Clinica Medica, LIM/42, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Claudia Latronico
- Disciplina de Endocrinologia e Metabologia, Departamento de Clinica Medica, LIM/42, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Berenice Mendonca
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil.,Disciplina de Endocrinologia e Metabologia, Departamento de Clinica Medica, LIM/42, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ivo Arnhold
- Disciplina de Endocrinologia e Metabologia, Departamento de Clinica Medica, LIM/42, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Verónica Mericq
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
46
|
Cerbone M, Katugampola H, Simpson HL, Dattani MT. Approach to the Patient: Management of Pituitary Hormone Replacement Through Transition. J Clin Endocrinol Metab 2022; 107:2077-2091. [PMID: 35262704 PMCID: PMC9202712 DOI: 10.1210/clinem/dgac129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 11/19/2022]
Abstract
Hypopituitarism in childhood is a rare, complex disorder that can present with highly variable phenotypes, which may continue into adult life. Pituitary deficits can evolve over time, with unpredictable patterns resulting in significant morbidity and mortality. Hypopituitarism and hypothalamic dysfunction may be associated with challenging comorbidities such as obesity, learning difficulties, behavioral issues, sleep disturbance, and visual impairment. Transition is the purposeful planned movement of adolescents and young adults with chronic conditions from child-centered to adult-oriented health care systems with a shift from parent- to patient-focused care. To achieve effective transition within a health care setting, the inherent challenges involved in the evolution from a dependent child to an independent adult must be recognized. Transition is a critical time medically for patients with hypopituitarism. Complex issues with respect to puberty, attainment of optimal stature, adherence to treatment, and acceptance of the need for life-sustaining medications need to be addressed. For health care professionals, transition is an opportunity for reassessment of the pituitary deficits and the need for lifelong replacement therapies, often against a background of complex psychological issues. We present 4 illustrative cases of hypopituitarism of differing etiologies with diverse clinical presentations. Diagnostic and management processes from clinical presentation to young adulthood are discussed, with a particular focus on needs and outcomes through transition.
Collapse
Affiliation(s)
- Manuela Cerbone
- London Centre for Paediatric Endocrinology and Diabetes at Great Ormond Street Children’s Hospital and University College London Hospitals, London WC1N 1EH, UK
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Programme, University College London Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Harshini Katugampola
- London Centre for Paediatric Endocrinology and Diabetes at Great Ormond Street Children’s Hospital and University College London Hospitals, London WC1N 1EH, UK
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Programme, University College London Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Helen L Simpson
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Programme, University College London Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Department of Diabetes and Endocrinology, University College London Hospitals NHS Trust, London NW1 2BU, UK
| | - Mehul T Dattani
- London Centre for Paediatric Endocrinology and Diabetes at Great Ormond Street Children’s Hospital and University College London Hospitals, London WC1N 1EH, UK
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Programme, University College London Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| |
Collapse
|
47
|
Abstract
Pituitary autoimmunity is one of the principal causes of hypopituitarism. Additionally, hypophysitis is one of the immune-related adverse events associated with immunotherapy. Recent case-oriented research has revealed a novel type of autoimmune hypophysitis, anti-PIT-1 hypophysitis, related to isolated adrenocorticotropic hormone (ACTH) deficiency and immune checkpoint inhibitor-related hypophysitis, as a form of paraneoplastic syndrome. Under these conditions, the ectopic expression of pituitary antigens present in tumors evokes a breakdown of immune tolerance, resulting in the production of autoantibodies and autoreactive cytotoxic T cells that specifically harm pituitary cells. Consequently, an innovative clinical entity of paraneoplastic autoimmune hypophysitis has been purported. This novel concept and its underlying mechanisms provide clues for understanding the pathogenesis of autoimmune pituitary diseases and can be applied to other autoimmune diseases. This review discusses the etiology of paraneoplastic autoimmune hypophysitis and its future.
Collapse
Affiliation(s)
- Hironori Bando
- Division of Development of Advanced Therapy for Metabolic Diseases, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Keitaro Kanie
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Department of Diabetes and Endocrinology, Nara Medical University, Kashihara, Japan.
| |
Collapse
|
48
|
Abstract
The epidemiology of male hypogonadism has been understudied. Of the known causes of endogenous androgen deficiency, only Klinefelter syndrome is common with a likely population prevalence of greater than 5:10,000 men (possibly as high as 10-25:10,000). Mild traumatic injury might also be a common cause of androgen deficiency (prevalence 5-10:10,000 men), but large, long-term studies must be completed to confirm this prevalence estimation that might be too high. The classic causes of male androgen deficiency-hyperprolactinemia, pituitary macroadenoma, endogenous Cushing syndrome, and iron overload syndrome-are rare (prevalence < 10,000 men).
Collapse
Affiliation(s)
- Arthi Thirumalai
- Department of Medicine, University of Washington School of Medicine, Box 356420, 1959 Northeast Pacific Avenue, Seattle, WA 98195, USA
| | - Bradley D Anawalt
- Department of Medicine, University of Washington School of Medicine, Box 356420, 1959 Northeast Pacific Avenue, Seattle, WA 98195, USA.
| |
Collapse
|
49
|
Susaimanickam PJ, Kiral FR, Park IH. Region Specific Brain Organoids to Study Neurodevelopmental Disorders. Int J Stem Cells 2022; 15:26-40. [PMID: 35220290 PMCID: PMC8889336 DOI: 10.15283/ijsc22006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 01/17/2022] [Indexed: 12/03/2022] Open
Abstract
Region specific brain organoids are brain organoids derived by patterning protocols using extrinsic signals as opposed to cerebral organoids obtained by self-patterning. The main focus of this review is to discuss various region-specific brain organoids developed so far and their application in modeling neurodevelopmental disease. We first discuss the principles of neural axis formation by series of growth factors, such as SHH, WNT, BMP signalings, that are critical to generate various region-specific brain organoids. Then we discuss various neurodevelopmental disorders modeled so far with these region-specific brain organoids, and findings made on mechanism and treatment options for neurodevelopmental disorders (NDD).
Collapse
Affiliation(s)
- Praveen Joseph Susaimanickam
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Ferdi Ridvan Kiral
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
50
|
Huang Q, Mao J, Wang X, Yu B, Ma W, Ji W, Zhu Y, Zhang R, Sun B, Zhang J, Nie M, Wu X. Efficacy of Pulsatile Gonadotropin-Releasing Hormone Therapy in Male Patients: Comparison between Pituitary Stalk Interruption Syndrome and Congenital Hypogonadotropic Hypogonadism. Endocr Pract 2022; 28:521-527. [PMID: 35218954 DOI: 10.1016/j.eprac.2022.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Pulsatile gonadotropin-releasing hormone (GnRH), widely used to induce spermatogenesis in congenital hypogonadotropic hypogonadism (CHH) patients, can restore the pituitary-testis axis function in males with pituitary stalk interruption syndrome (PSIS). This retrospective study aimed to compare the long-term efficacy of pulsatile GnRH therapy between PSIS and CHH. METHODS Patients of PSIS (n=25) or CHH (n=64) who received pulsatile GnRH therapy ≥ 3 months were analyzed in this retrospective study. The rate of successful spermatogenesis, the median time to achieve spermatogenesis, serum gonadotropins, total testosterone (TT), and testicular size were compared. RESULTS Baseline characteristics were comparable except for the lower basal testosterone, triptorelin stimulated peak luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in patients with PSIS. Within similar treatment durations, significantly higher GnRH dose (p < 0.001) but lower increase in LH [2.82 (1.4, 4.55) vs. 5.89 (3.88, 8.02) IU/L, p < 0.001], TT [0.38 (0, 1.34) vs. 2.34 (1.34, 3.66) ng/mL, p < 0.001], and testicular volume (5.3 ± 4.5 vs. 8.8 ± 4.8 mL, p < 0.05) were observed in PSIS. However, spermatogenesis rate (52.0% vs. 70.3%, p > 0.05), median time of sperm appearance (14 vs. 11 months, p > 0.05), sperm concentration and progressive motility were comparable. Basal testicular volume (HR 1.13, 95% CI 1.01-1.27) and peak LH levels (HR 1.11, 95% CI 1.0-1.23) were predictors for early sperm appearance. CONCLUSIONS Pulsatile GnRH therapy can improve gonad function and induce spermatogenesis in male PSIS patients, however, its efficacy may be inferior to that in CHH.
Collapse
Affiliation(s)
- Qibin Huang
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jiangfeng Mao
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xi Wang
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Bingqing Yu
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Wanlu Ma
- Department of Endocrinology, China-Japan Friendship Hospital, Beijing, 100730, China
| | - Wen Ji
- Department of Endocrinology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Yiyi Zhu
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Rui Zhang
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Bang Sun
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Junyi Zhang
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Nie
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xueyan Wu
- NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital; Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|