1
|
Toulia I, Savvidou P, Ververi A, Grammatikopoulou MG, Kosta K, Tziaferi V, Antachopoulos C, Goulis DG, Sotiriadis A, Tsiroukidou K. Clinical and genetic diagnosis and management of Silver-Russell syndrome: Report of four cases. World J Clin Pediatr 2025; 14:100330. [DOI: 10.5409/wjcp.v14.i2.100330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/19/2024] [Accepted: 01/07/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Silver-Russell syndrome (SRS) is a clinically heterogeneous entity characterized by intrauterine and postnatal growth restriction, relative macrocephaly at birth, distinct facial features, and body asymmetry combined with other malformations.
CASE SUMMARY Herein, we describe four individuals with SRS, focusing on their prenatal phenotype, postnatal presentation, diagnosis, and management. All cases had a typical phenotype, including postnatal growth failure, short stature (chronic malnutrition), and protruding forehead. Individually, they presented with feeding difficulties, leg length discrepancy, triangular face, or relative macrocephaly at birth, and each one exhibited distinct SRS features, including motor and/or speech delay, experiencing frequent hypoglycemic episodes. The fact that each patient exhibited a different combination of clinical findings underlines the heterogeneity of the syndrome.
CONCLUSION SRS is diagnosed clinically. However, only 60% of cases are genetically confirmed, while most are sporadic. Although SRS is a well-described syndrome, a delayed diagnosis can have grave consequences on a child’s growth. Recombinant human growth hormone treatment is often initiated shortly after the diagnosis. The follow-up requires a multidisciplinary approach.
Collapse
Affiliation(s)
- Ilektra Toulia
- Endocrine Unit, 3rd Pediatric Department, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Kentrikí Makedonía, Greece
| | - Parthena Savvidou
- Endocrine Unit, 3rd Pediatric Department, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Kentrikí Makedonía, Greece
| | - Athina Ververi
- Department of Genetics for Rare Diseases, Papageorgiou General Hospital, Thessaloniki GR-54124, Kentrikí Makedonía, Greece
| | - Maria G Grammatikopoulou
- Immunonutrition Unit, Department of Rheumatology & Clinical Immunology, University of Thessaly, Larissa University Hospital, Larissa GR-41223, Thessalía, Greece
| | - Konstantina Kosta
- Endocrine Unit, 3rd Pediatric Department, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Kentrikí Makedonía, Greece
| | - Vaya Tziaferi
- Pediatric Endocrinology Clinic, Thessaloniki GR-55535, Kentrikí Makedonía, Greece
| | - Charalampos Antachopoulos
- 3rd Department of Pediatrics, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Kentrikí Makedonía, Greece
| | - Dimitrios G Goulis
- First Department of Obstetrics and Gynecology, Medical School, Papageorgiou General Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54601, Kentrikí Makedonía, Greece
| | - Alexandros Sotiriadis
- 2nd Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki GR-54124, Greece
| | - Kyriaki Tsiroukidou
- Endocrine Unit, 3rd Pediatric Department, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Kentrikí Makedonía, Greece
| |
Collapse
|
2
|
Begemann M, Lengyel A, Pinti E, Kovács ÁF, Fekete G, Stratmann S, Krause J, Elbracht M, Kraft F, Eggermann T. Maternal uniparental disomy of chromosome 7: how chromosome 7-encoded imprinted genes contribute to the Silver-Russell phenotype. Clin Epigenetics 2025; 17:70. [PMID: 40307819 PMCID: PMC12042466 DOI: 10.1186/s13148-025-01867-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Silver-Russell syndrome (SRS) is a rare congenital growth disorder which is associated with molecular alterations affecting imprinted regions on chromosome 11p15 and maternal uniparental disomy of chromosome 7 (upd(7)mat). In 11p15, imprinted regions contributing to the SRS phenotype could be identified, whereas on chromosome 7 at least two regions in 7q32 and 7p13 are in discussion as SRS candidate regions. We report on DNA and RNA data from upd(7)mat patients and a monozygotic twin pair with a postnatal SRS phenotype carrying a small intragenic deletion within GRB10 to delineate the contribution of upd(7)mat and imprinted genes on this chromosome to the SRS phenotype. RESULTS Genome sequencing in the monozygotic twins revealed a 18 kb deletion within the paternal allele of the GRB10 gene. Expression of GRB10 in blood of the twins as well as in cells from upd(7)mat and upd(7q)mat patients was not altered, whereas RNAseq indicates noticeable changes of the expression of other genes encoded by chromosomes 7 and other genomic regions. CONCLUSIONS Our data indicate that intrauterine growth restriction as the prenatal phenotype of upd(7)mat is caused by defective paternal alleles of the 7q32 region, as well as by overexpression of the maternal GRB10 allele whereas a defective GRB10 paternal allele does not cause this feature. The altered expression of MEST in 7q32 by upd(7)mat is associated with the complete SRS phenotype, whereas maternalization or deletion of the paternal GRB10 copy and duplication of the chromosomal region 7p12 are associated with a postnatal SRS-like phenotype.
Collapse
Affiliation(s)
- Matthias Begemann
- Medical Faculty, Centre for Human Genetics and Genome Medicine, RWTH University Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Anna Lengyel
- Pediatric Center Tuzolto Street Department, Semmelweis University, Budapest, Hungary
| | - Eva Pinti
- Pediatric Center Tuzolto Street Department, Semmelweis University, Budapest, Hungary
| | - Árpád Ferenc Kovács
- Pediatric Center Tuzolto Street Department, Semmelweis University, Budapest, Hungary
| | - György Fekete
- Pediatric Center Tuzolto Street Department, Semmelweis University, Budapest, Hungary
| | - Svea Stratmann
- Science for Life Laboratory, Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Jeremias Krause
- Medical Faculty, Centre for Human Genetics and Genome Medicine, RWTH University Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Miriam Elbracht
- Medical Faculty, Centre for Human Genetics and Genome Medicine, RWTH University Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Florian Kraft
- Medical Faculty, Centre for Human Genetics and Genome Medicine, RWTH University Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Thomas Eggermann
- Medical Faculty, Centre for Human Genetics and Genome Medicine, RWTH University Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
3
|
Fang HH, Lee CL, Chuang CK, Chiu HC, Chang YH, Tu YR, Lo YT, Wu JY, Chou YY, Wang CH, Lin SJ, Chu SY, Yang C, Ou TY, Lin HY, Lin SP. Functional Independence of Taiwanese Children with Silver-Russell Syndrome. Diagnostics (Basel) 2025; 15:1109. [PMID: 40361928 PMCID: PMC12071216 DOI: 10.3390/diagnostics15091109] [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: 03/22/2025] [Revised: 04/15/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Silver-Russell syndrome (SRS) is a genetic disorder characterized by prenatal and postnatal growth retardation. Affected individuals commonly present with low birth weight, intrauterine growth restriction, postnatal short stature, hemihypotrophy, characteristic facial features, and body asymmetry. Methods: This study includes 24 Taiwanese children with SRS aged 2 years to 13 years and 3 months who were recruited at MacKay Memorial Hospital and other Taiwan hospitals between January 2013 and December 2024. Functional independence was assessed using the Functional Independence Measure for Children (WeeFIM) to evaluate self-care, mobility, and cognition domains. Results: The mean total WeeFIM score was 106.9 ± 23.2 (range: 54-126), with mean self-care, mobility, and cognition scores of 44.4 ± 13.8 (maximum 56), 32.4 ± 5.1 (maximum 35), and 30.2 ± 6.0 (maximum 35), respectively. The results of the restricted cubic spline analysis reveal a clear positive linear correlation before school age (approximately 72 months), followed by a plateau (p for nonlinearity < 0.05). Traceable molecular data were available for thirteen participants, of whom nine (69%) had loss of methylation at chromosome 11p15 (11p15LOM), and four (31%) had maternal uniparental disomy of chromosome 7 (upd(7)mat). Of the 24 children, 46% required assistance with bathing, which was strongly correlated with self-care ability and body height. In contrast, most of the children had independence in mobility tasks such as walking and stair climbing. However, some required support in cognitive tasks, including problem-solving, comprehension, and expression. Overall, the included children reached a functional plateau later than the normative population, with the greatest delays in self-care and mobility domains. Conclusions: This study highlights that Taiwanese children with SRS require support in self-care and cognitive tasks. Functional independence in self-care and mobility domains was positively associated with body height. The WeeFIM questionnaire effectively identified strengths and limitations, emphasizing the need for individualized support in daily activities.
Collapse
Grants
- NSTC-113-2314-B-195-003, NSTC-113-2314-B-195-004, NSTC-113-2314-B-715-002, NSTC-113-2314-B-195-021, NSTC-113-2811-B-195-001, NSTC-112-2314-B-195-014-MY3, NSTC-112-2811-B-195-001, NSTC-112-2314-B-195-003, NSTC-111-2314-B-195-017, NSTC-111-2811-B-195-002, N Ministry of Science and Technology, Executive Yuan, Taiwan
Collapse
Affiliation(s)
- Hung-Hsiang Fang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 104, Taiwan; (H.-H.F.); (C.-L.L.); (H.-C.C.); (Y.-H.C.)
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 104, Taiwan; (H.-H.F.); (C.-L.L.); (H.-C.C.); (Y.-H.C.)
- Institute of Clinical Medicine, National Yang-Ming Chiao-Tung University, Taipei 112, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei 104, Taiwan; (Y.-T.L.); (J.-Y.W.)
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 112, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 104, Taiwan; (C.-K.C.); (Y.-R.T.)
- College of Medicine, Fu-Jen Catholic University, Taipei 242, Taiwan
| | - Huei-Ching Chiu
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 104, Taiwan; (H.-H.F.); (C.-L.L.); (H.-C.C.); (Y.-H.C.)
| | - Ya-Hui Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 104, Taiwan; (H.-H.F.); (C.-L.L.); (H.-C.C.); (Y.-H.C.)
- International Rare Disease Center, MacKay Memorial Hospital, Taipei 104, Taiwan; (Y.-T.L.); (J.-Y.W.)
| | - Yuan-Rong Tu
- Department of Medical Research, MacKay Memorial Hospital, Taipei 104, Taiwan; (C.-K.C.); (Y.-R.T.)
| | - Yun-Ting Lo
- International Rare Disease Center, MacKay Memorial Hospital, Taipei 104, Taiwan; (Y.-T.L.); (J.-Y.W.)
| | - Jun-Yi Wu
- International Rare Disease Center, MacKay Memorial Hospital, Taipei 104, Taiwan; (Y.-T.L.); (J.-Y.W.)
| | - Yen-Yin Chou
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Chung-Hsing Wang
- Division of Medical Genetics, Pediatric Endocrinology and Metabolism, China Medical University Children’s Hospital, China Medical University, Taichung 404, Taiwan;
| | - Shio-Jean Lin
- Department of Pediatrics, Genetic Counseling Center, Chi Mei Medical Center, Tainan 710, Taiwan;
| | - Shao-Yin Chu
- Department of Pediatrics, Buddhist Tzu-Chi General Hospital, Hualien 970, Taiwan;
| | - Chen Yang
- Department of Pediatrics, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Tsung-Ying Ou
- Department of Pediatrics, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan;
| | - Hsiang-Yu Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 104, Taiwan; (H.-H.F.); (C.-L.L.); (H.-C.C.); (Y.-H.C.)
- International Rare Disease Center, MacKay Memorial Hospital, Taipei 104, Taiwan; (Y.-T.L.); (J.-Y.W.)
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei 112, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei 104, Taiwan; (C.-K.C.); (Y.-R.T.)
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Shuan-Pei Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 104, Taiwan; (H.-H.F.); (C.-L.L.); (H.-C.C.); (Y.-H.C.)
- International Rare Disease Center, MacKay Memorial Hospital, Taipei 104, Taiwan; (Y.-T.L.); (J.-Y.W.)
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei 104, Taiwan; (C.-K.C.); (Y.-R.T.)
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei 112, Taiwan
| |
Collapse
|
4
|
Biesmans K, van Aken L, Wetzels-Meertens S, Schreurs L, Wingbermühle E, de Graaff L, Kleefstra T, Egger J. Intelligence, Cognition, and Psychopathology in Adults with Silver-Russell Syndrome: Overview of the Literature and Description of Three Clinical Cases. Arch Clin Neuropsychol 2025:acaf026. [PMID: 40257940 DOI: 10.1093/arclin/acaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/08/2025] [Accepted: 03/02/2025] [Indexed: 04/23/2025] Open
Abstract
OBJECTIVE Research on the neuropsychological profile in individuals with Silver-Russell Syndrome (SRS), is scarce. The current study aims to enhance common clinical knowledge of SRS by exploring the syndrome related neuropsychological specificities previously described by literature and comparing those with neuropsychological findings in three adults with SRS. METHODS Literature search on intelligence, cognition, and psychopathology in SRS was carried out. Included articles defined SRS adults with known genetic etiology and provided objective measures available within the investigated domains. Analyzes of the latter was conducted through available data from neuropsychological assessment in three persons with SRS; one male with maternal 11p duplication and two females with H19 hypomethylation. RESULTS Analysis of the literature suggested specific cognitive profiles for individuals with different genetic variants. As to the presented cases, for the maternal 11p duplication, a higher variability of overall intellectual abilities and stronger verbal comprehension was found. Across all cases, overall intellectual abilities varied from mild intellectual disability to average level. No specific cognitive profile was found. Psychopathology presented itself either as externalizing or internalizing, and all cases reported negative life and/or learning experiences and self-esteem issues directly related to SRS. CONCLUSION Clinical cases' results were consistent with literature. Next to the prevailing focus on somatic aspects, current results support in-depth analysis of neuropsychological functioning as necessary to optimize care and reduce the risk of psychopathology during the life course in SRS. Further research and tailored selection of neuropsychological batteries is recommended to improve the understanding of the cognitive profile of SRS.
Collapse
Affiliation(s)
- Kim Biesmans
- STEVIG Department of Specialized and Forensic Care for People with Intellectual Disabilities, Dichterbij, Oostrum, The Netherlands
- Centre of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Loes van Aken
- Centre of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Sascha Wetzels-Meertens
- STEVIG Department of Specialized and Forensic Care for People with Intellectual Disabilities, Dichterbij, Oostrum, The Netherlands
| | - Lisanne Schreurs
- STEVIG Department of Specialized and Forensic Care for People with Intellectual Disabilities, Dichterbij, Oostrum, The Netherlands
| | - Ellen Wingbermühle
- Centre of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Laura de Graaff
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tjitske Kleefstra
- Centre of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jos Egger
- STEVIG Department of Specialized and Forensic Care for People with Intellectual Disabilities, Dichterbij, Oostrum, The Netherlands
- Centre of Excellence for Neuropsychiatry, Vincent van Gogh Institute for Psychiatry, Venray, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
5
|
O'Leary EM, Bonthuis PJ. Mom genes and dad genes: genomic imprinting in the regulation of social behaviors. Epigenomics 2025:1-19. [PMID: 40249667 DOI: 10.1080/17501911.2025.2491294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/31/2025] [Indexed: 04/20/2025] Open
Abstract
Genomic imprinting is an epigenetic phenomenon in mammals that affects brain development and behavior. Imprinting involves the regulation of allelic expression for some genes in offspring that depends on whether alleles are inherited from mothers compared to fathers, and is thought to provide parental control over offspring social behavior phenotypes. Imprinted gene expression is prevalent in the mammalian brain, and human imprinted gene mutations are associated with neurodevelopmental disorders and neurodivergent social behavior in Prader-Willi Syndrome, Angelman Syndrome, and autism. Here, we provide a review of the evidence that imprinted genes influence social behaviors across major neurodevelopmental stages in humans and mouse animal models that include parent-infant interactions, juvenile sociability, and adult aggression, dominance, and sexual behavior.
Collapse
Affiliation(s)
- Erin M O'Leary
- Neuroscience Program, University of Illinois, Urbana, IL, USA
| | - Paul J Bonthuis
- Neuroscience Program, University of Illinois, Urbana, IL, USA
- Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Gene Networks in Neural & Development Plasticity Theme at Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
6
|
Braga BL, da Cunha Scalco R, Homma TK, Freire BL, Cellin LDP, Canton APM, Lerario AM, de Assis Funari MF, de Souza V, Bertola DR, Malaquias AC, Mendonca BB, de Lima Jorge AA. Rare Causes and Differential Diagnosis in Patients With Silver-Russell Syndrome. Clin Genet 2025; 107:441-445. [PMID: 39586716 DOI: 10.1111/cge.14659] [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: 08/30/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Silver-Russell syndrome (SRS) is an imprinting disorder mainly characterized by pre- and postnatal growth restriction. Most SRS cases are due to 11p15.5 loss of methylation (11p15.5 LOM) or maternal uniparental disomy of chromosome 7 [UPD(7)mat], but several patients remain molecularly undiagnosed. This study describes the molecular investigation of children with a clinical diagnosis or suspicion of SRS at a tertiary center specialized in growth disorders. Thirty-nine patients were evaluated with multiplex ligation-dependent probe amplification, chromosomal microarray and/or massively parallel sequencing. The most common result was 11p15.5 LOM (n = 17; 43.5%), followed by UPD(7)mat (n = 2; 5.1%). Additionally, we found maternal duplications of the imprinting centers in 11p15.5 (n = 2; 5.1%), and genetic defects in SRS-causing genes (IGF2 and HMGA2) (n = 3; 7.7%; two mutations and one deletion). Alternative molecular diagnoses included UPD(14)mat (n = 1; 2,6%), UPD(20)mat (n = 1;2,6%), copy number variants (n = 2; 5.1%), and mutations in genes associated with other growth disorders (n = 4; 10.3%), leading to diagnoses of Temple syndrome, Mulchandani-Bhoj-Conlin syndrome, IGF-1 resistance (IGF1R), Bloom syndrome (BLM), Gabriele-De Vries syndrome (YY1), Intellectual developmental disorder autosomal dominant 50 with behavioral abnormalities (NAA15), and Intellectual developmental disorder 64 (ZNF292). These findings underscore the importance of establishing the molecular diagnosis of SRS and its differential diagnoses to guide appropriate management and genetic counseling.
Collapse
Affiliation(s)
- Barbara Leitao Braga
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Renata da Cunha Scalco
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thais Kataoka Homma
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Lucheze Freire
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Laurana De Polli Cellin
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Pinheiro Machado Canton
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Antônio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, USA
| | - Mariana Ferreira de Assis Funari
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vinicius de Souza
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Debora Romeo Bertola
- Unidade de Genética, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Alexsandra Christianne Malaquias
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Unidade de Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - Berenice Bilharinho Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Alexander Augusto de Lima Jorge
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM42), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Narusawa H, Ogawa T, Yagasaki H, Nagasaki K, Urakawa T, Saito T, Soneda S, Kinjo S, Sano S, Mamada M, Terashita S, Dateki S, Narumi S, Naiki Y, Horikawa R, Ogata T, Fukami M, Kagami M. Comprehensive Study on Central Precocious Puberty: Molecular and Clinical Analyses in 90 Patients. J Clin Endocrinol Metab 2025; 110:1023-1036. [PMID: 39324648 DOI: 10.1210/clinem/dgae666] [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: 06/06/2024] [Revised: 08/25/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
CONTEXT Defects in MKRN3, DLK1, KISS1, and KISS1R and some disorders, such as Temple syndrome (TS14), cause central precocious puberty (CPP). Recently, pathogenic variants (PVs) in MECP2 have been reported to be associated with CPP. OBJECTIVE We aimed to clarify the contribution of (epi)genetic abnormalities to CPP and clinical and hormonal features in each etiology. METHODS We conducted targeted sequencing for MKRN3, DLK1, MECP2, KISS1, and KISS1R and methylation analysis for screening of imprinting disorders such as TS14 associated with CPP in 90 patients with CPP (no history of brain injuries and negative brain magnetic resonance imaging) and collected their clinical and laboratory data. We measured serum DLK1 levels in 3 patients with TS14 and serum MKRN3 levels in 2 patients with MKRN3 genetic defects, together with some etiology-unknown patients with CPP and controls. RESULTS We detected 8 patients with TS14 (6, epimutation; 1, mosaic maternal uniparental disomy chromosome 14; 1, microdeletion) and 3 patients with MKRN3 genetic defects (1, PV; 1, 13-bp deletion in the 5'-untranslated region [5'-UTR]; 1, microdeletion) with family histories of paternal early puberty. There were no patients with PVs identified in MECP2, KISS1, or KISS1R. We confirmed low serum MKRN3 level in the patient with a deletion in 5'-UTR. The median height at initial evaluation of TS14 patients was lower than that of all patients. Six patients with TS14 were born small for gestational age (SGA). CONCLUSION (Epi)genetic causes were identified in 12.2% of patients with CPP at our center. For patients with CPP born SGA or together with family histories of paternal early puberty, (epi)genetic testing for TS14 and MKRN3 genetic defects should be considered.
Collapse
Affiliation(s)
- Hiromune Narusawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Tomoe Ogawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Hideaki Yagasaki
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo 409-3898, Japan
| | - Keisuke Nagasaki
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8510, Japan
| | - Tatsuki Urakawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Tomohiro Saito
- Department of Pediatrics, Yamanashi Prefectural Central Hospital, Kofu 400-0027, Japan
| | - Shun Soneda
- Tanaka Growth Clinic, Tokyo 158-0097, Japan
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-0015, Japan
| | - Saori Kinjo
- Department of Pediatrics, Okinawa Chubu Hospital, Uruma 904-2293, Japan
| | - Shinichiro Sano
- Department of Pediatric Endocrinology and Metabolism, Shizuoka Children's Hospital, Shizuoka 420-0953, Japan
| | - Mitsukazu Mamada
- Department of Pediatrics, Japanese Red Cross Wakayama Medical Center, Wakayama 640-8558, Japan
| | - Shintaro Terashita
- Department of Pediatrics, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Sumito Dateki
- Department of Pediatrics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Yasuhiro Naiki
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Reiko Horikawa
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Tsutomu Ogata
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
- Department of Pediatrics, Hamamatsu Medical Center, Hamamatsu 432-8580, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
8
|
Vimercati A, Tannorella P, Guzzetti S, Calzari L, Gentilini D, Manfredini E, Gori G, Gaudino R, Antona V, Piccione M, Daolio C, Auricchio R, Sirchia F, Minelli A, Rossi E, Bellini M, Biasucci G, Raucci AR, Pozzobon G, Patti G, Napoli F, Larizza L, Maghnie M, Russo S. Distinguishing Genetic Alterations Versus (Epi)Mutations in Silver-Russell Syndrome and Focus on the IGF1R Gene. J Clin Endocrinol Metab 2025; 110:e932-e944. [PMID: 39412159 PMCID: PMC11913091 DOI: 10.1210/clinem/dgae730] [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: 06/26/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 01/03/2025]
Abstract
CONTEXT Silver-Russell Syndrome (SRS) is a growth retardation disorder characterized by pre- and postnatal growth failure, relative macrocephaly at birth, prominent forehead, body asymmetry, and feeding difficulties. The main molecular mechanisms are imprinting alterations at multiple loci, though a small number of pathogenic variants have been reported in the SRS genes IGF2-PLAG1-HMGA2 and CDKN1C. However, around 40% of clinically suspected SRS cases do not achieve a molecular diagnosis, highlighting the necessity to uncover the underlying mechanism in unsolved cases. OBJECTIVE Evaluate the frequency of genetic variants in undiagnosed SRS patients [Netchine-Harbison Clinical Scoring System (NH-CSS) ≥ 4], and investigate whether (epi)genetic patients may be distinguished from genetic patients. METHODS One hundred thirty-two clinically SRS patients without (epi)genetic deregulations were investigated by whole-exome (n = 15) and targeted (n = 117) Sequencing. Clinical data from our cohort and from an extensive revision of the literature were compared. RESULTS Pathogenic variants were identified in 9.1% of this cohort: 3% in IGF2, PLAG1, and HMGA2 genes and 3% in the IGF1R gene, associated with IGF-1 resistance (IGF1RES), an SRS differential diagnosis. Overall, IGF2-PLAG1-HMGA2 and IGF1R account for 3.6% of SRS with NH-CSS score ≥ 4. A clinical cross-comparison of (epi)genetic vs genetic SRS underlined (epi)genotype-phenotype correlation highlighted the prevalence of body asymmetry and relative macrocephaly in mosaic (epi)genetic SRS and recurrence of genetic familial cases. Furthermore, overlapping features were evidenced in (epi)genetic SRS and IGF1RES patients. CONCLUSION Our study explores the frequency of genetic SRS, underscores body asymmetry as a distinctive phenotype in (epi)genetic SRS and suggests IGF1R sequencing in a SRS diagnostic flowchart.
Collapse
Affiliation(s)
- Alessandro Vimercati
- Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| | - Pierpaola Tannorella
- Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| | - Sara Guzzetti
- Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| | - Luciano Calzari
- Bioinformatics and Statistical Genomic Unit, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| | - Davide Gentilini
- Bioinformatics and Statistical Genomic Unit, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Emanuela Manfredini
- Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| | - Giulia Gori
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Rossella Gaudino
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37129 Verona, Italy
| | - Vincenzo Antona
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D'Alessandro,” University of Palermo, 90127 Palermo, Italy
| | - Maria Piccione
- Medical Genetics Unit Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Cecilia Daolio
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Renata Auricchio
- European Laboratory for the Investigation of Food Induced Diseases, Department of Translational Medical Science, Section of Pediatrics, University Federico II, 80131 Naples, Italy
| | - Fabio Sirchia
- Medical Genetic Unit, Department of Diagnostic Medicine, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Antonella Minelli
- Medical Genetic Unit, Department of Diagnostic Medicine, IRCCS San Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Elena Rossi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Melissa Bellini
- Pediatrics and Neonatology Unit, Gugliemo da Saliceto Hospital, 29121 Piacenza, Italy
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Gugliemo da Saliceto Hospital, 29121 Piacenza, Italy
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Annalisa Russo Raucci
- Division of Genetics and Cell Biology and Laboratory of Clinical Molecular Biology and Cytogenetics, Unit of Genomics for Human Disease Diagnosis, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Giuseppa Patti
- Paediatric Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16147 Genoa, Italy
| | - Flavia Napoli
- Paediatric Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Lidia Larizza
- Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| | - Mohamad Maghnie
- Paediatric Endocrinology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16147 Genoa, Italy
| | - Silvia Russo
- Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy
| |
Collapse
|
9
|
Kessler L, Krause J, Kraft F, Amin AK, Fekete G, Lengyel A, Pinti E, Kovacs A, Lischka A, Eggermann K, Kurth I, Knopp C, Elbracht M, Begemann M, Eggermann T. Diagnostic Use of Genome Sequencing in Patients With 11p15.5 Imprinting Disorder Features: A Pilot Study. Clin Genet 2025; 107:278-291. [PMID: 39663844 PMCID: PMC11790513 DOI: 10.1111/cge.14649] [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: 08/14/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
To assess the suitability of genome sequencing (GS) as the second step in the diagnostics of patients with the features of 11p15.5-associated imprinting disorders (ImpDis: Silver-Russell syndrome [SRS], Beckwith-Wiedemann syndrome [BWS]), we performed short-read GS in patients negatively tested for imprinting disturbances. Obtaining a genetic diagnosis for patients with the features of these syndromes is challenging due to the clinical and molecular heterogeneity and overlap, and many patients remain undiagnosed after the currently suggested stepwise diagnostic workup. GS was conducted in 48 patients (SRS features: n = 37 and BWS features: n = 11). The detection rate differed markedly between the ImpDis: although a genetic cause could be identified in 51% of patients referred with SRS features, no pathogenic variants were detected in patients with BWS features. Thus, GS substantially improves the diagnostic yield and broadens the spectrum of overlapping disorders with SRS features. Obtaining a precise molecular diagnosis provides the basis for a personalized clinical management. Our findings support the use of GS as a second-tier diagnostic tool for patients with growth disturbances, as it addresses all currently known variant types and shortens the diagnostic odyssey.
Collapse
Affiliation(s)
- Luise Kessler
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Jeremias Krause
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Florian Kraft
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Asmaa K. Amin
- Department of Human Genetics, Medical Research InstituteAlexandria UniversityAlexandriaEgypt
| | - Gyorgy Fekete
- 2nd Department of PediatricsSemmelweis UniversityBudapestHungary
| | - Anna Lengyel
- 2nd Department of PediatricsSemmelweis UniversityBudapestHungary
| | - Eva Pinti
- 2nd Department of PediatricsSemmelweis UniversityBudapestHungary
| | - Arpad Kovacs
- 2nd Department of PediatricsSemmelweis UniversityBudapestHungary
| | - Annette Lischka
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Katja Eggermann
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Cordula Knopp
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Miriam Elbracht
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Matthias Begemann
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Thomas Eggermann
- Institute for Human Genetics and Genomic Medicine, Medical FacultyRWTH Aachen UniversityAachenGermany
| |
Collapse
|
10
|
Muz N, Petersson M, Saalman R, Dahlgren J. Percutaneous endoscopic gastrostomy helped to normalise feeding problems and gastrointestinal symptoms in Silver-Russell syndrome. Acta Paediatr 2025; 114:569-577. [PMID: 39460380 PMCID: PMC11828731 DOI: 10.1111/apa.17474] [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: 06/20/2024] [Revised: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
AIM This study evaluated feeding problems and gastrointestinal symptoms in children with Silver-Russell syndrome (SRS), which is a rare epigenetic disorder. It also compared the symptoms experienced during different feeding methods, including percutaneous endoscopic gastrostomy (PEG). METHODS The national expert team for children with SRS at Queen Silvia Children's Hospital, Gothenburg, studied 46 referrals (63% male) who were born with SRS in Sweden from 1984 to 2018. Patient data were extracted from the Paediatric National Growth Hormone Registry. RESULTS The medical records covered a median of 68% of the time of the patients' childhood, with a median follow-up of 9 years. Their symptoms were most prevalent during infancy and decreased when they were toddlers. Feeding problems and gastrointestinal symptoms were reported in 91% of the 46 patients, with vomiting in 57% and constipation in 46%. There were 19 children who relied on enteral feeding for their nutrition and 13 of those received PEG. Their body mass index (BMI) increased significantly 2 years after PEG started (p = 0.005). CONCLUSION Feeding problems and gastrointestinal symptoms were very common in children with SRS, but partly disappeared during childhood. Providing treatment, such as PEG, normalised the BMIs of children with SRS and reduced their symptoms.
Collapse
Affiliation(s)
- Nataliia Muz
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Miriam Petersson
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Robert Saalman
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Region Västra Götaland, Department of Pediatric MedicineQueen Silvia Children's HospitalGothenburgSweden
| | - Jovanna Dahlgren
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Region Västra Götaland, Department of Pediatric MedicineQueen Silvia Children's HospitalGothenburgSweden
| |
Collapse
|
11
|
Cheng L, Yin Z, Liu H, Shi S, Lv L, Wang Y, Zhou M, Li M, Guo T, Guo X, Yang G, Ma J, Yu J, Zhang Y, Duo S, Zhao L, Li R. Inhibition of LncRNA H19 Attenuates Testicular Torsion-Induced Apoptosis and Preserves Blood-Testis Barrier Integrity. Int J Mol Sci 2025; 26:2134. [PMID: 40076761 PMCID: PMC11899958 DOI: 10.3390/ijms26052134] [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: 01/21/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Testicular torsion is a common emergency in adolescents, and can lead to severe ischemia reperfusion injury (IRI). LncRNA H19 has been shown to increase during ischemia, but its role in testicular IRI remains unknown. Focusing on this research gap, we utilized H19 biallelic mutant mice and Sertoli cell line (TM4) to construct in vivo and in vitro models of ischemia/reperfusion (I/R) and oxygen-glucose deprivation/reperfusion (OGD/R). Compared to WT I/R mice, H19-/- I/R mice showed milder tissue disorganization and cell loss, with a more intact blood-testis barrier (BTB). The cell viability decreased, ROS levels and apoptosis-related factors such as Bax/Bcl-2 increased in TM4 cells after OGD/R, whereas these changes were reversed when H19 was knocked down followed by OGD/R (si-H19+OGD/R). In contrast, over-expression of H19 in TM4 cells exacerbates OGD/R-induced cell apoptosis. Through in-depth analysis of KEGG-enriched pathways, the PI3K/AKT pathway was identified as a potential target of H19 modulation. Western blotting confirmed that, in OGD/R cells, elevated H19 levels were accompanied by the excessive AKT phosphorylation and the tight junction marker ZO-1 degradation; and in si-H19+OGD/R cells, the decreased AKT phosphorylation was recovered and the up-regulated ZO-1 expression was weakened simultaneously via using the AKT activator SC79. These results suggest that inhibiting H19 in OGD/R cells might preserve the integrity of the BTB by reversing the excessive phosphorylation of AKT. Moreover, H19 deficiency in si-H19+OGD/R cells alleviated the disturbances in glycolysis, fatty acid biosynthesis, and amino acid metabolism. Our study indicates that H19 might be a potential therapeutic target for clinic testicular I/R treatment.
Collapse
Affiliation(s)
- Linxin Cheng
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhibao Yin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Han Liu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Sijing Shi
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Limin Lv
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China;
| | - Yixi Wang
- Laboratory Animal Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (Y.W.); (S.D.)
| | - Meng Zhou
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Meishuang Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Tianxu Guo
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiyun Guo
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Guang Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Junjun Ma
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jinbo Yu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yu Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shuguang Duo
- Laboratory Animal Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (Y.W.); (S.D.)
| | - Lihua Zhao
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Rongfeng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; (L.C.); (Z.Y.); (H.L.); (S.S.); (M.Z.); (M.L.); (T.G.); (X.G.); (G.Y.); (J.M.); (J.Y.); (Y.Z.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China;
| |
Collapse
|
12
|
Aghili ZS, Khoshnevisan G, Mostoli R, Alibaglouei M, Zarkesh-Esfahani SH. Growth hormone signaling and clinical implications: from molecular to therapeutic perspectives. Mol Biol Rep 2025; 52:202. [PMID: 39904816 DOI: 10.1007/s11033-025-10304-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/24/2025] [Indexed: 02/06/2025]
Abstract
Growth hormone (GH) is a key polypeptide hormone secreted by somatotroph cells in the anterior pituitary gland, essential for postnatal growth, metabolism, and systemic homeostasis. Its secretion is regulated by hypothalamic neuropeptides, including GH-releasing hormone and somatostatin. GH exerts effects through direct interaction with the growth hormone receptor and indirect pathways mediated by the GH-IGF-I axis. GHR activation triggers signaling pathways, such as JAK-STAT, PI3K/AKT, and MAPK, promoting cellular proliferation, differentiation, and metabolic balance. The GH-IGF-I axis is critical for bone growth, lipid and carbohydrate metabolism, and organ-specific physiological functions. Dysregulation of GH results in diverse disorders. Congenital deficiencies, like isolated GH deficiency and syndromic conditions (e.g., Turner syndrome), stem from genetic mutations. Acquired deficiencies arise from trauma, tumors, infections, or autoimmune damage, while GH overproduction causes gigantism in children and acromegaly in adults, often due to pituitary adenomas. Idiopathic deficiencies, lacking identifiable causes, complicate management further. Advances in therapy have transformed outcomes for GH disorders. Recombinant human growth hormone provides effective replacement therapy for deficiencies. Somatostatin analogs, dopamine receptor agonists, and GH receptor antagonists are pivotal for managing GH excess. Surgical and radiotherapeutic interventions remain essential for pituitary adenomas. However, GH therapy requires close monitoring to prevent side effects like insulin resistance and metabolic complications. This review provides a comprehensive evaluation of the molecular mechanisms underlying GH action, its physiological roles, GH-related disorders, and therapeutic approaches to optimize patient outcomes.
Collapse
Affiliation(s)
- Zahra Sadat Aghili
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Golnoosh Khoshnevisan
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Rezvan Mostoli
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Alibaglouei
- Department of Biological Sciences, University of Cincinnati, Cincinnati, USA
| | - Sayyed Hamid Zarkesh-Esfahani
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
13
|
Huo X, Lu X, Lu D, Liu H, Liu Y, Zhao Q, Sun Y, Dai W, Qiu W, Yu Y, Fan Y. Clinical utility of regions of homozygosity (ROH) identified in exome sequencing: when to pursue confirmatory uniparental disomy testing for imprinting disorders? Clin Chem Lab Med 2025; 63:87-96. [PMID: 39022805 DOI: 10.1515/cclm-2024-0239] [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: 02/22/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES Regions of homozygosity (ROH) could implicate uniparental disomy (UPD) on specific chromosomes associated with imprinting disorders. Though the algorithms for ROH detection in exome sequencing (ES) have been developed, optimal reporting thresholds and when to pursue confirmatory UPD testing for imprinting disorders remain in ambiguity. This study used a data-driven approach to assess optimal reporting thresholds of ROH in clinical practice. METHODS ROH analysis was performed using Automap in a retrospective cohort of 8,219 patients and a prospective cohort of 1,964 patients with ES data. Cases with ROH on imprinting-disorders related chromosomes were selected for additional methylation-specific confirmatory testing. The diagnostic yield, the ROH pattern of eventually diagnosed cases and optimal thresholds for confirmatory testing were analyzed. RESULTS In the retrospective analysis, 15 true UPD cases of imprinting disorders were confirmed among 51 suspected cases by ROH detection. Pattern of ROH differed between confirmed UPD and non-UPD cases. Maximized yield and minimized false discovery rate of confirmatory UPD testing was achieved at the thresholds of >20 Mb or >25 % chromosomal coverage for interstitial ROH, and >5 Mb for terminal ROH. Current recommendation by ACMG was nearly optimal, though refined thresholds as proposed in this study could reduce the workload by 31 % without losing any true UPD diagnosis. Our refined thresholds remained optimal after independent evaluation in a prospective cohort. CONCLUSIONS ROH identified in ES could implicate the presence of clinically relevant UPD. This study recommended size and coverage thresholds for confirmatory UPD testing after ROH detection in ES, contributing to the development of evidence-based reporting guidelines.
Collapse
Affiliation(s)
- Xiaoyan Huo
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Lu
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Deyun Lu
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huili Liu
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Liu
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianfeng Zhao
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqian Dai
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjuan Qiu
- Department of Pediatric Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongguo Yu
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjie Fan
- Clinical Genetics Center, 91603 Shanghai Institute for Pediatric Research , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Walker V. The Molecular Biology of Placental Transport of Calcium to the Human Foetus. Int J Mol Sci 2025; 26:383. [PMID: 39796238 PMCID: PMC11720126 DOI: 10.3390/ijms26010383] [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: 10/30/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
From fertilisation to delivery, calcium must be transported into and within the foetoplacental unit for intracellular signalling. This requires very rapid, precisely located Ca2+ transfers. In addition, from around the eighth week of gestation, increasing amounts of calcium must be routed directly from maternal blood to the foetus for bone mineralisation through a flow-through system, which does not impact the intracellular Ca2+ concentration. These different processes are mediated by numerous membrane-sited Ca2+ channels, transporters, and exchangers. Understanding the mechanisms is essential to direct interventions to optimise foetal development and postnatal bone health and to protect the mother and foetus from pre-eclampsia. Ethical issues limit the availability of human foetal tissue for study. Our insight into the processes of placental Ca2+ handling is advancing rapidly, enabled by developing genetic, analytical, and computer technology. Because of their diverse sources, the reports of new findings are scattered. This review aims to pull the data together and to highlight areas of uncertainty. Areas needing clarification include trafficking, membrane expression, and recycling of channels and transporters in the placental microvilli; placental metabolism of vitamin D in gestational diabetes and pre-eclampsia; and the vascular effects of increased endothelial Orai expression by pregnancy-specific beta-1-glycoproteins PSG1 and PSG9.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton SO16 6YD, UK
| |
Collapse
|
15
|
Giabicani E, Perrière A, Netchine I. Silver-Russell Syndrome in 2025: Is It Still a Distinct Diagnostic Entity? J Clin Endocrinol Metab 2025:dgae902. [PMID: 39745816 DOI: 10.1210/clinem/dgae902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Affiliation(s)
- Eloïse Giabicani
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, APHP, Hôpital Armand Trousseau, Explorations Fonctionnelles Endocriniennes, F-75012, Paris, France
| | - Amélie Perrière
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, APHP, Hôpital Armand Trousseau, Explorations Fonctionnelles Endocriniennes, F-75012, Paris, France
| | - Irène Netchine
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, APHP, Hôpital Armand Trousseau, Explorations Fonctionnelles Endocriniennes, F-75012, Paris, France
| |
Collapse
|
16
|
Liu P, Han X, Li X, Dai S, Xu Y, Jiao L, Du H, Zhao L, Li R, Teng Z, Yang Y, Liu C. An EED/PRC2-H19 Loop Regulates Cerebellar Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403591. [PMID: 39498824 PMCID: PMC11714151 DOI: 10.1002/advs.202403591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/29/2024] [Indexed: 11/07/2024]
Abstract
EED (embryonic ectoderm development) is a core subunit of the polycomb repressive complex 2 (PRC2), which senses the trimethylation of histone H3 lysine 27 (H3K27). However, its biological function in cerebellar development remains unknown. Here, we show that EED deletion from neural stem cells (NSCs) or cerebellar granule cell progenitors (GCPs) leads to reduced GCPs proliferation, cell death, cerebellar hypoplasia, and motor deficits in mice. Joint profiling of transcripts and ChIP-seq analysis in cerebellar granule cells reveals that EED regulates bunches of genes involved in cerebellar development. EED ablation exhibits overactivation of a developmental repressor long non-coding RNA H19. Importantly, an obvious H3K27ac enrichment is found at Ctcf, a trans-activator of H19, and H3K27me3 enrichment at the H19 imprinting control region (ICR), suggesting that EED regulates H19 in an H3K27me3-dependent manner. Intriguingly, H19 deletion reduces EED expression and the reprogramming of EED-mediated H3K27me3 profiles, resulting in increased proliferation, differentiation, and decreased apoptosis of GCPs. Finally, molecular and genetic evidence provides that increased H19 expression is responsible for cerebellar hypoplasia and motor defects in EED mutant mice. Thus, this study demonstrates that EED, H19 forms a negative feedback loop, which plays a crucial role in cerebellar morphogenesis and controls cerebellar development.
Collapse
Affiliation(s)
- Pei‐Pei Liu
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Xiao Han
- University of Chinese Academy of SciencesBeijing100049China
- Key Laboratory of Genomic and Precision MedicineCollaborative Innovation Center of Genetics and DevelopmentCollege of Future TechnologyBeijing Institute of GenomicsChinese Academy of SciencesBeijing100101China
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xiao Li
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Shang‐Kun Dai
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Ya‐Jie Xu
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Lin‐Fei Jiao
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Hong‐Zhen Du
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Li‐Hua Zhao
- Jiangsu Key Laboratory of XenotransplantationNanjing Medical UniversityNanjing211166China
| | - Rong‐Feng Li
- Jiangsu Key Laboratory of XenotransplantationNanjing Medical UniversityNanjing211166China
- Key Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166China
| | - Zhao‐Qian Teng
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Yun‐Gui Yang
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
- Key Laboratory of Genomic and Precision MedicineCollaborative Innovation Center of Genetics and DevelopmentCollege of Future TechnologyBeijing Institute of GenomicsChinese Academy of SciencesBeijing100101China
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100049China
- China National Center for BioinformationBeijing100101China
| | - Chang‐Mei Liu
- Key Laboratory of Organ Regeneration and ReconstructionState Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| |
Collapse
|
17
|
Noda M, Matsumoto A, Ito H, Kagami M, Tajima T, Matsumura T, Yamagata T, Nagata KI. An unstable variant of GAP43 leads to neurodevelopmental deficiency. Sci Rep 2024; 14:31911. [PMID: 39738362 PMCID: PMC11686380 DOI: 10.1038/s41598-024-83445-w] [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: 09/06/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Growth-associated protein 43 (GAP43) is a membrane-associated phosphoprotein predominantly expressed in the nervous systems, and controls axonal growth, branching, and pathfinding. While the association between GAP43 and human neurological disorders have been reported, the underlying mechanisms remain largely unknown. We performed whole exome sequencing on a patient with intellectual disability (ID), neurodevelopmental disorders, short stature, and skeletal abnormalities such as left-right difference in legs and digital deformities, and identified a heterozygous missense variation in the GAP43 gene [NM_001130064.2: c.436G > A/p.(E146K)]. The variant GAP43 protein was unstable in primary cultured cortical neurons and hippocampal neurons in vitro. In utero electroporation of the variant protein also confirmed its instability in vivo, suggesting that the variant led to a condition similar with haploinsufficiency in the patient. Silencing of GAP43 via in utero electroporation of RNAi vectors demonstrated that loss of GAP43 suppressed axon elongation into the contralateral hemisphere and impaired the dendritic arbor formation as shown by decreased dendritic branch points and shortened total dendritic lengths. Collectively, these findings confirmed the critical roles of GAP43 in brain development and the pathological basis of GAP43-associated diseases. Our study will contribute to a better understanding of how dysregulation of GAP43 leads to human diseases.
Collapse
Affiliation(s)
- Mariko Noda
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan
| | - Ayumi Matsumoto
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
- Division of Cardiovascular and Genetic Research, Center for Molecular Medicine, Jichi Medical University, 3311-1 YakushijiShimotsuke-Shi, Tochigi-Ken, 329-0498, Japan
| | - Hidenori Ito
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan
| | - Masayo Kagami
- Clinical Endocrine Research Division, Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Toshihiro Tajima
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Takayoshi Matsumura
- Division of Cardiovascular and Genetic Research, Center for Molecular Medicine, Jichi Medical University, 3311-1 YakushijiShimotsuke-Shi, Tochigi-Ken, 329-0498, Japan.
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan.
| | | | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan.
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
18
|
Piekoszewska-Ziętek P, Witt-Porczyk A, Turska-Szybka A, Olczak-Kowalczyk D. Hygienic behaviors and use of dental care in patients with genetic syndromes. Sci Rep 2024; 14:30756. [PMID: 39730420 DOI: 10.1038/s41598-024-80922-0] [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: 04/05/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Patients with genetic syndromes require special dental attention because they have symptoms that promote plaque accumulation, dental erosion, dental caries and gingival diseases. The aim of the study was to assess hygienic behaviors, use of dental care and frequency of professional preventive procedures among Polish children and adolescents with Prader-Willi, Down, Angelman, Silver-Russell and Smith-Lemli-Opitz syndromes. Parents/legal guardians of children and adolescents with genetic syndromes were included. A questionnaire survey was conducted regarding socioeconomic factors, hygienic procedures performed at home and use of dental care as well as use of preventive treatments. The percentage of patients with genetic syndromes who received dental care was statistically significantly lower compared to the control group. Oral hygiene measures were most frequently used by participants with Silver-Russel syndrome, and less commonly by patients with Prader-Willi and Down syndrome. Dental treatment under general anesthesia was provided in 26 (38.2%) of the 68 children with genetic syndromes receiving dental care. Hygienic neglect and inadequate use of dental care due to limited access to certain preventive and therapeutic procedures among patients with genetic syndromes are worrying. It is necessary to educate and intensify caries prevention in this group of patients.
Collapse
Affiliation(s)
- Paula Piekoszewska-Ziętek
- Department of Paediatric Dentistry, Medical University of Warsaw, St. Binieckiego 6, 02-097, Warsaw, Poland.
| | | | - Anna Turska-Szybka
- Department of Paediatric Dentistry, Medical University of Warsaw, St. Binieckiego 6, 02-097, Warsaw, Poland
| | - Dorota Olczak-Kowalczyk
- Department of Paediatric Dentistry, Medical University of Warsaw, St. Binieckiego 6, 02-097, Warsaw, Poland.
| |
Collapse
|
19
|
Hong S, Wei H, Zhuang X, Huang W, Zhang Y. Prenatal diagnosis of a silver-russell syndrome caused by 11p15 duplication and pedigree analysis. Front Genet 2024; 15:1465521. [PMID: 39741906 PMCID: PMC11686225 DOI: 10.3389/fgene.2024.1465521] [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: 07/16/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Silver-Russell syndrome (SRS) is an imprinting disorder characterized by intrauterine and postnatal growth retardation. The pathogenic alterations and phenotypes are heterogeneous. Methods Here, we present a rare pedigree of duplications with different methylation patterns in 11p15.5, which caused SRS or a normal phenotype across three generations. Results Duplications of maternal IC2 (copy number of 3) with enhanced methylation (methylation index of 0.62) resulted in typical SRS. Conclusion The result added to the complexity of the molecular genetics of SRS.
Collapse
Affiliation(s)
- Shurong Hong
- Department of Molecular Genetic Center, Zhangzhou Municipal Hospital Affiliated to Fujian Medical University, Zhangzhou, China
| | - Hua Wei
- Department of Molecular Genetic Center, Zhangzhou Municipal Hospital Affiliated to Fujian Medical University, Zhangzhou, China
| | - Xueyi Zhuang
- Department of Obstetrics, Zhangzhou Municipal Hospital Affiliated to Fujian Medical University, Zhangzhou, China
| | - Weirong Huang
- Department of Molecular Genetic Center, Zhangzhou Municipal Hospital Affiliated to Fujian Medical University, Zhangzhou, China
| | - Yu Zhang
- Department of Obstetrics, Zhangzhou Municipal Hospital Affiliated to Fujian Medical University, Zhangzhou, China
| |
Collapse
|
20
|
Shoji T, Yamauchi I, Kawasaki H, Iwanaga K, Hakata T, Tanaka D, Fujikura J, Masui T, Suzuki H, Yamada M, Kosaki K, Kasai Y, Hatano E, Inaba A, Wada T, Kosugi S, Ueda Y, Fujii T, Taura D, Inagaki N. Case report: Duplication of the GCK gene is a novel cause of nesidioblastosis: evidence from a case with Silver-Russell syndrome-like phenotype related to chromosome 7. Front Endocrinol (Lausanne) 2024; 15:1431547. [PMID: 39720245 PMCID: PMC11666348 DOI: 10.3389/fendo.2024.1431547] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 11/13/2024] [Indexed: 12/26/2024] Open
Abstract
Silver-Russell syndrome (SRS) is a syndrome characterized by prenatal and postnatal growth retardation, facial features, and body asymmetry. SRS is often complicated with hypoglycemia, whose etiology is unclear. We describe the clinical course of 25-year-old man with hypoglycemia. We diagnosed him with hyperinsulinemic hypoglycemia (HH) and treated him with laparoscopic distal pancreatectomy. Histological examination led to a diagnosis of nesidioblastosis. The juvenile onset of his nesidioblastosis and its slowly progressive course suggested a genetic etiology. Whole-exome sequencing (WES) identified the heterozygous NR0B2 Ala195Ser variant, which alone was unlikely to cause nesidioblastosis because this variant is sometimes detected in the Japanese population. Copy number analysis using WES data suggested duplication in chromosome 7, and subsequent G-banding chromosome analysis confirmed mos dup(7)(p11.2p14). We determined that the patient had SRS-like phenotype based on his clinical features and this duplication. Furthermore, we found that the duplicated region contained the GCK gene, whose gain-of function variants could cause HH. Taken together, the patient's HH may have been caused by duplication of the GCK gene, which could be a novel cause of nesidioblastosis.
Collapse
Affiliation(s)
- Takashi Shoji
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ichiro Yamauchi
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidenori Kawasaki
- Department of Genomic Medicine, Kyoto University School of Public Health, Kyoto, Japan
| | - Kogoro Iwanaga
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takuro Hakata
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Tanaka
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junji Fujikura
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshihiko Masui
- Department of Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Hisato Suzuki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Mamiko Yamada
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Yosuke Kasai
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Etsuro Hatano
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akira Inaba
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Takahito Wada
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Shinji Kosugi
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Yohei Ueda
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshihito Fujii
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Taura
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobuya Inagaki
- Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-kofukai, Osaka, Japan
| |
Collapse
|
21
|
Yordanova N, Iotova V, Mackay DJG, Temple IK, Stoyanova S, Hachmeriyan M. Long-term Follow-up of a Late Diagnosed Patient with Temple Syndrome. J Clin Res Pediatr Endocrinol 2024; 16:475-480. [PMID: 36728278 PMCID: PMC11629717 DOI: 10.4274/jcrpe.galenos.2022.2022-9-19] [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: 10/14/2022] [Accepted: 12/13/2022] [Indexed: 02/03/2023] Open
Abstract
Temple syndrome is a rare imprinting disorder, caused by alterations in the critical imprinted region 14q32 of chromosome 14. It is characterized by pre- and postnatal growth retardation, truncal hypotonia and facial dysmorphism in the neonatal period. We report an 18-year-old girl with a late diagnosis of Temple syndrome presenting with all typical signs and symptoms including small for gestational age at birth, feeding difficulties, muscle hypotonia and delayed developmental milestones, central precocious puberty, truncal obesity and reduced growth. The patient is the second reported in the literature with signs of clinical and biochemical hyperandrogenism and the first treated with Dehydrocortisone®, with a good response. The clinical diagnosis of this patient was made after long-term follow up at a single center for rare endocrine diseases, and a molecular genetics diagnosis of complete hypomethylation of 14q32 chromosome imprinting center (DLK/GTL2) was recently established. Growth hormone treatment was not given and although precocious puberty was treated in line with standard protocols, her final height remained below the target range. Increased awareness of Temple syndrome and timely molecular diagnosis enables improvement of clinical care of these patients as well as prevention of inherent metabolic consequences.
Collapse
Affiliation(s)
| | - Violeta Iotova
- Medical University-Varna, Department of Pediatrics, Varna, Bulgaria
| | - Deborah J. G. Mackay
- Wessex Regional Genetics Laboratory, Salisbury Foundation NHS Trust, Salisbury, United Kingdom
- University of Southampton Faculty of Medicine, Department of Medical Genetics, Southampton, United Kingdom
| | - I. Karen Temple
- University of Southampton Faculty of Medicine, Department of Medical Genetics, Southampton, United Kingdom
| | - Sara Stoyanova
- Medical University-Varna, Department of Pediatrics, Varna, Bulgaria
| | - Mari Hachmeriyan
- Medical University, Department of Medical Genetics, Varna, Bulgaria
| |
Collapse
|
22
|
Hara S, Matsuhisa F, Kitajima S, Yatsuki H, Kubiura-Ichimaru M, Higashimoto K, Soejima H. Identification of responsible sequences which mutations cause maternal H19-ICR hypermethylation with Beckwith-Wiedemann syndrome-like overgrowth. Commun Biol 2024; 7:1605. [PMID: 39623082 PMCID: PMC11612015 DOI: 10.1038/s42003-024-07323-x] [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: 08/07/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
Beckwith-Wiedemann syndrome (BWS) is caused by a gain of methylation (GOM) at the imprinting control region within the Igf2-H19 domain on the maternal allele (H19-ICR GOM). Mutations in the binding sites of several transcription factors are involved in H19-ICR GOM and BWS. However, the responsible sequence(s) for H19-ICR GOM with BWS-like overgrowth has not been identified in mice. Here, we report that a mutation in the SOX-OCT binding site (SOBS) causes partial H19-ICR GOM, which does not extend beyond CTCF binding site 3 (CTS3). Moreover, simultaneously mutating both SOBS and CTS3 causes complete GOM of the entire H19-ICR, leading to the misexpression of the imprinted genes, and frequent BWS-like overgrowth. In addition, CTS3 is critical for CTCF/cohesin-mediated chromatin conformation. These results indicate that SOBS and CTS3 are the sequences in which mutations cause H19-ICR GOM leading to BWS-like overgrowth and are essential for maintaining the unmethylated state of maternal H19-ICR.
Collapse
Affiliation(s)
- Satoshi Hara
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, 849-8501, Japan.
| | - Fumikazu Matsuhisa
- Division of Biological Resources and Development, Analytical Research Center for Experimental Sciences, Saga University, Saga, 849-8501, Japan
| | - Shuji Kitajima
- Division of Biological Resources and Development, Analytical Research Center for Experimental Sciences, Saga University, Saga, 849-8501, Japan
| | - Hitomi Yatsuki
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Musashi Kubiura-Ichimaru
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Ken Higashimoto
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Hidenobu Soejima
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, 849-8501, Japan.
| |
Collapse
|
23
|
Xi L, Cheng R, He Y, Li X, Ni J, Wu J, Xu Z, Luo F. Factors affecting growth hormone treatment in short stature children born small for gestational age in China: a single-centre, real-world study. Endocrine 2024; 86:1121-1130. [PMID: 39210232 DOI: 10.1007/s12020-024-04009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE The study aimed to evaluate the factors influencing recombinant human growth hormone (rhGH) treatment in Chinese children with short stature born small for gestational age (SGA). METHODS A single-centre, real-world retrospective study was conducted in short stature children born SGA in China. Outcomes were observed at 6, 12, 18, 24, 30, and 36 months. Outcome measures included height standard deviation score (HTSDS), height, growth velocity (GV), and change of HTSDS (ΔHTSDS). The study used the generalized estimating equation (GEE) to identify potential influencing factors, such as rhGH treatment duration, age at rhGH initiation, sex, 11p15 hypomethylation, GH secretion, and birth weight. A subgroup analysis was conducted to investigate the impact of 11p15 hypomethylation related to SGA or impaired GH secretion. RESULTS Of all 101 SGA patients included in the screening, 41 were eligible for inclusion in the study. The mean age at rhGH initiation was 5.6 ± 2.4 years. The results of the GEE analysis showed a significant association between time after rhGH initiation and HTSDS, height, GV, and ΔHTSDS. GV increased after treatment, with the highest increase observed in the first six months. Additionally, the study found negative correlations between 11p15 hypomethylation and GV, as well as between birth weight and both GV and ΔHTSDS. The study found a positive correlation between impairment in GH secretion and both GV and ΔHTSDS. No statistically significant difference was observed in the comparison of GV or ΔHTSDS between the initiation age of GH treatment and 11p15 hypomethylation. After 24 and 30 months of rhGH treatment, patients with impaired GH secretion had significantly higher ΔHTSDS scores. CONCLUSIONS In short stature Chinese children born SGA, those without SGA-related 11p15 hypomethylation or with impaired GH secretion showed better response to rhGH treatment. These findings highlight the importance of pre-treatment evaluation, including genetic and endocrine assessments.
Collapse
Affiliation(s)
- Li Xi
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Ruoqian Cheng
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Yingkai He
- Medical Affairs, GeneScience Pharmaceuticals Co., Ltd. (GenSci), Shanghai, China
| | - Xiaojing Li
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Jinwen Ni
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Jing Wu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Zhenran Xu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Feihong Luo
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Shi Y, Qu F, Zeng S, Wang X, Liu Y, Zhang Q, Yuan D, Yuan C. Targeting long non-coding RNA H19 as a therapeutic strategy for liver disease. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 194:1-9. [PMID: 39357625 DOI: 10.1016/j.pbiomolbio.2024.09.005] [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: 05/16/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
The liver has the function of regulating metabolic equilibrium in the human body, and the majority of liver disorders are chronic conditions that can significantly impair health. Recent research has highlighted the critical role of long noncoding RNAs (lncRNAs) in liver disease pathogenesis. LncRNA H19, an endogenous noncoding single-stranded RNA, exerts its influence through epigenetic modifications and affects various biological processes. This review focuses on elucidating the key molecular mechanisms underlying the regulation of H19 during the progression and advancement of liver diseases, aiming to highlight H19 as a potential therapeutic target and provide profound insights into the molecular underpinnings of liver pathologies.
Collapse
Affiliation(s)
- Yulan Shi
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Medicine and Health Science, China Three Gorges University Yichang, 443002, China
| | - Fenghua Qu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Medicine and Health Science, China Three Gorges University Yichang, 443002, China
| | - Shiyun Zeng
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University Yichang, 443002, China
| | - Xinchen Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Medicine and Health Science, China Three Gorges University Yichang, 443002, China
| | - Yuting Liu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Medicine and Health Science, China Three Gorges University Yichang, 443002, China
| | - Qirui Zhang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University Yichang, 443002, China
| | - Ding Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Medicine and Health Science, China Three Gorges University Yichang, 443002, China
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University Yichang, 443002, China.
| |
Collapse
|
25
|
Vieira DKR, Lima IBF, Rosenberg C, da Fonseca CR, Gomes LHF, Guida LDC, Mazzonetto PC, Llerena J, Bastos EF. Clinical and Cytogenetic Impact of Maternal Balanced Double Translocation: A Familial Case of 15q11.2 Microduplication and Microdeletion Syndromes with Genetic Counselling Implications. Genes (Basel) 2024; 15:1546. [PMID: 39766813 PMCID: PMC11728287 DOI: 10.3390/genes15121546] [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: 10/01/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Balanced chromosomal translocations occur in approximately 0.16 to 0.20% of live births. While most carriers are phenotypically normal, they are at risk of generating unbalanced gametes during meiosis, leading to genetic anomalies such as aneuploidies, deletions, duplications, and gene disruptions. These anomalies can result in spontaneous abortions or congenital anomalies, including neurodevelopmental disorders. Complex chromosomal rearrangements (CCRs) involving more than two chromosomes are rare but further increase the probability of producing unbalanced gametes. Neurodevelopmental disorders such as Angelman syndrome (AS) and duplication 15q11q13 syndrome (Dup15q) are associated with such chromosomal abnormalities. METHODS This study describes a family with a de novo maternal balanced double translocation involving chromosomes 13, 19, and 15, resulting in two offspring with unbalanced chromosomal abnormalities. Cytogenetic evaluations were performed using GTG banding, fluorescence in situ hybridization (FISH), and low-pass whole-genome sequencing (LP-WGS). Methylation analysis was conducted using methylation-sensitive high-resolution melting (MS-HRM) to diagnose Angelman syndrome. RESULTS The cytogenetic and molecular analyses identified an 8.9 Mb duplication in 15q11.2q13.3 in one child, and an 8.9 Mb deletion in the same region in the second child. Both abnormalities affected critical neurodevelopmental genes, such as SNRPN. FISH and MS-HRM confirmed the chromosomal imbalances and the diagnosis of Angelman syndrome in the second child. The maternal balanced translocation was found to be cryptic, contributing to the complex inheritance pattern. CONCLUSION This case highlights the importance of using multiple genetic platforms to uncover complex chromosomal rearrangements and their impact on neurodevelopmental disorders. The findings underscore the need for thorough genetic counseling, especially in families with such rare chromosomal alterations, to manage reproductive outcomes and neurodevelopmental risks.
Collapse
Affiliation(s)
- Daniela Koeller R. Vieira
- Centro de Genética Médica, Instituto Nacional da Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira–Fundação Oswaldo Cruz, Rio de Janeiro 22250-020, Brazil;
- Secretaria Municipal de Saúde de Angra dos Reis, Angra dos Reis 23906-010, Brazil
| | - Ingrid Bendas Feres Lima
- Laboratório de Citogenética Clínica, Centro de Genética Médica, Instituto Nacional da Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira–Fundação Oswaldo Cruz, Rio de Janeiro 22250-020, Brazil; (I.B.F.L.); (C.R.d.F.)
| | - Carla Rosenberg
- The Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo 05508-220, Brazil; (C.R.); (P.C.M.)
- Diagnósticos da América S.A., DASA, São Paulo 06455-010, Brazil
| | - Carlos Roberto da Fonseca
- Laboratório de Citogenética Clínica, Centro de Genética Médica, Instituto Nacional da Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira–Fundação Oswaldo Cruz, Rio de Janeiro 22250-020, Brazil; (I.B.F.L.); (C.R.d.F.)
| | - Leonardo Henrique Ferreira Gomes
- Departamento de Pesquisa Clínica, Instituto Nacional da Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira–Fundação Oswaldo Cruz, Rio de Janeiro 22250-020, Brazil; (L.H.F.G.); (L.d.C.G.)
| | - Letícia da Cunha Guida
- Departamento de Pesquisa Clínica, Instituto Nacional da Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira–Fundação Oswaldo Cruz, Rio de Janeiro 22250-020, Brazil; (L.H.F.G.); (L.d.C.G.)
| | - Patrícia Camacho Mazzonetto
- The Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo 05508-220, Brazil; (C.R.); (P.C.M.)
- Diagnósticos da América S.A., DASA, São Paulo 06455-010, Brazil
| | - Juan Llerena
- Centro de Genética Médica, Instituto Nacional da Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira–Fundação Oswaldo Cruz, Rio de Janeiro 22250-020, Brazil;
| | - Elenice Ferreira Bastos
- Laboratório de Citogenética Clínica, Centro de Genética Médica, Instituto Nacional da Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira–Fundação Oswaldo Cruz, Rio de Janeiro 22250-020, Brazil; (I.B.F.L.); (C.R.d.F.)
| |
Collapse
|
26
|
Jurca AD, Petchesi CD, Jurca S, Severin E, Jurca AA, Jurca CM. Clinical Challenges in Diagnosing Primordial Dwarfism: Insights from a MOPD II Case Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1906. [PMID: 39597091 PMCID: PMC11596399 DOI: 10.3390/medicina60111906] [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: 10/22/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives.Primordial dwarfism (PD) is a rare group of genetic conditions where individuals experience severe growth restriction, both in the womb and after birth. From as early as the fetal stage, those affected are significantly smaller than their peers. What makes PD distinct is its slow but steady growth pattern, resulting in proportionate dwarfism, where all parts of the body are equally shortened. Diagnosing and managing PD presents significant challenges due to its rarity and the wide range of clinical and genetic variability. The main conditions in this group include Seckel syndrome, Microcephalic Osteodysplastic Primordial Dwarfism (MOPD) types I/III, MOPD type II, Meier-Gorlin syndrome, and Silver-Russell syndrome (SRS). The first four-Seckel syndrome, MOPD types I/III, MOPD type II, and Meier-Gorlin syndrome-are associated with microcephaly, and together they are known as microcephalic PD. Given how uncommon PD is, establishing its exact incidence is difficult. It is estimated that about 4 million infants die within the first month of life, with 99% of these deaths occurring in the neonatal period. Materials and Methods. Accurately diagnosing PD requires meticulous evaluation, as it can be easily confused with other genetic disorders that also cause dwarfism. In this article, we present the case of a 10-year-old patient diagnosed with MOPD II, the most common and well-documented form of microcephalic PD. Results. Genetic analysis revealed a pathogenic variant in the PCNT (pericentrin) gene ((c.1550dup, p.Gln518Alafs*7), alongside a deletion of exons 37-41. Conclusions. This case sheds light on the clinical and genetic complexities of primordial dwarfism, underscoring the importance of timely and accurate diagnosis for effective patient care.
Collapse
Affiliation(s)
- Alexandru Daniel Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
| | - Codruța Diana Petchesi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN-ITHACA), 410469 Oradea, Romania
| | - Sânziana Jurca
- Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania;
| | - Emilia Severin
- Department of Genetics, ”Carol Davila”University of Medicine and Pharmacy–Bucharest, Dionisie Lupu Street, Number 37, District 2, 020021 Bucharest, Romania
| | | | - Claudia Maria Jurca
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410081 Oradea, Romania; (A.D.J.); (C.D.P.); (C.M.J.)
- Regional Center of Medical Genetics Bihor, County Emergency Clinical Hospital Oradea (Part of ERN-ITHACA), 410469 Oradea, Romania
| |
Collapse
|
27
|
Yadav S, Madhumita RC, Gupta N, Chauhan S, Kusmakar S, Balakrishnan P, Jana M, Puri RD, Phadke SR, Kabra M. Isolated Lateralized Overgrowth - Phenotypic Spectrum and Molecular Alterations. Indian J Pediatr 2024:10.1007/s12098-024-05273-0. [PMID: 39425824 DOI: 10.1007/s12098-024-05273-0] [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: 02/21/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES To evaluate the molecular aberrations at 11p15.5 locus in thirty-two patients with isolated lateralized overgrowth (ILO). METHODS Among selected 32 cases of ILO, methylation-sensitive multiplex ligation-dependent probe amplification (MS-MLPA) was performed initially followed by short tandem repeats (STR) marker analysis to confirm uniparental disomy (UPD). In those patients with normal MLPA reports, cyclin dependent kinase inhibitor 1C (CDKN1C) gene and whole exome sequencing was performed. RESULTS Molecular analysis by MS-MLPA showed methylation aberrations in 28% (9/32) of patients. Gain of methylation at IC1 imprinting center (H4, H7) and loss of methylation at IC2 (H6, H9) was observed in 2 patients each. Uniparental disomy was observed in 9% cases. Except one, all patients with methylation aberration had more than one limb hypertrophy. Two patients (H22/H29) also had loss of methylation at IC1. Though this molecular alteration is specifically associated with Silver Russel syndrome (SRS), but the affected children did not completely fulfill the diagnostic criteria for SRS. In a recent study, a discrepancy was reported between the diagnosis of Beckwith-Wiedemann syndrome (BWS)/SRS and the molecular findings in the patients. Many times, it is very difficult to differentiate between hemi hypertrophy/hemi hypotrophy. Patients, in whom no aberrations were detected on MS-MLPA, whole exome sequencing (WES) was performed and no pathogenic variant was identified. CONCLUSIONS Thus, ILO may be considered as a mild presentation on the extreme edge of BWS spectrum with methylation aberration and UPD in one third of cases which has implications in follow up.
Collapse
Affiliation(s)
- Sakshi Yadav
- Faith Diagnostic and Fetal Centre, Mohali, India
| | - R C Madhumita
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Neerja Gupta
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeepa Chauhan
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Shweta Kusmakar
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | | | - Manisha Jana
- Department of Radiodiagnosis, All India Institute of Medical Sciences, New Delhi, India
| | - Ratna D Puri
- Institute of Genetics & Genomics, Sir Gangaram Hospital, New Delhi, India
| | - Shubha R Phadke
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Madhulika Kabra
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
28
|
Lokulo-Sodipe O, Inskip HM, Byrne CD, Child J, Wakeling EL, Mackay DJG, Temple IK, Davies JH. Body Composition and Metabolism in Adults With Molecularly Confirmed Silver-Russell Syndrome. J Clin Endocrinol Metab 2024; 109:e2001-e2008. [PMID: 38330234 PMCID: PMC11479702 DOI: 10.1210/clinem/dgae074] [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: 11/04/2023] [Revised: 01/23/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
CONTEXT Low birth weight, as seen in Silver-Russell syndrome (SRS), is associated with later cardiometabolic disease. Data on long-term outcomes and adult body composition in SRS are limited. OBJECTIVE To evaluate body composition and metabolic health in adults with SRS. METHODS This was an observational study of 25 individuals with molecularly confirmed SRS, aged ≥ 18 years, from research facilities across the UK. Body composition and metabolic health were assessed at a single appointment. Individuals with SRS were compared with unaffected men and women (from the Southampton Women's Survey [SWS]). Fat mass, lean mass, bone mineral density (BMD), blood pressure, lipids, and blood glucose were measured. RESULTS Twenty-five adults with SRS were included (52% female). The median age was 32.9 years (range, 22.0 to 69.7). Fat percentage was greater in the SRS group than the SWS cohort (44.1% vs 30.3%, P < .001). Fat mass index was similar (9.6 vs 7.8, P = .3). Lean mass percentage (51.8% vs 66.2%, P < .001) and lean mass index (13.5 kg/m2 vs 17.3 kg/m2, P < .001) were lower in the SRS group than the SWS cohort. BMD was lower in the SRS group than the SWS cohort (1.08 vs 1.24, P < .001; all median values). Total cholesterol was ≥ 5 mmol/L in 52.0%. Triglyceride levels were ≥ 1.7 mmol/L in 20.8%. Fasting blood glucose levels were ≥ 6.1 mmol/L in 25.0%. Hypertension was present in 33.3%. CONCLUSION Adults with SRS have an unfavorable body composition and predisposition to cardiometabolic disease. These results support the need for a health surveillance strategy to mitigate adverse outcomes.
Collapse
Affiliation(s)
- Oluwakemi Lokulo-Sodipe
- Department of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Regional Paediatric Endocrinology Service, University Southampton Hospitals NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Hazel M Inskip
- Department of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- MRC Epidemiology Unit, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Christopher D Byrne
- Department of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Jenny Child
- Child Growth Foundation, c/o Kinnair Associates Limited, Aston House, Newcastle, NE5 1NB, UK (affiliation at the time of this work)
| | - Emma L Wakeling
- North East Thames Regional Genetic Service, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Deborah J G Mackay
- Department of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Wessex Regional Genetics Laboratory, Salisbury District Hospital, Salisbury, Wiltshire, SP2 8BJ, UK
| | - I Karen Temple
- Department of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- The Wessex Clinical Genetics Service, University Hospitals Southampton NHS Foundation Trust, Princess Anne Hospital, Coxford Road, Southampton, SO16 5YA, UK
| | - Justin H Davies
- Department of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Regional Paediatric Endocrinology Service, University Southampton Hospitals NHS Foundation Trust, Southampton, SO16 6YD, UK
| |
Collapse
|
29
|
Urakawa T, Soejima H, Yamoto K, Hara-Isono K, Nakamura A, Kawashima S, Narusawa H, Kosaki R, Nishimura Y, Yamazawa K, Hattori T, Muramatsu Y, Inoue T, Matsubara K, Fukami M, Saitoh S, Ogata T, Kagami M. Comprehensive molecular and clinical findings in 29 patients with multi-locus imprinting disturbance. Clin Epigenetics 2024; 16:138. [PMID: 39369220 PMCID: PMC11452994 DOI: 10.1186/s13148-024-01744-5] [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: 05/13/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Multi-locus imprinting disturbance (MLID) with methylation defects in various differentially methylated regions (DMRs) has recently been identified in approximately 150 cases with imprinting disorders (IDs), and deleterious variants have been found in genes related to methylation maintenance of DMRs, such as those encoding proteins constructing the subcortical maternal complex (SCMC), in a small fraction of patients and/or their mothers. However, integrated methylation analysis for DMRs and sequence analysis for MLID-causative genes in MLID cases and their mothers have been performed only in a single study focusing on Beckwith-Wiedemann syndrome (BWS) and Silver-Russell syndrome (SRS) phenotypes. RESULTS Of 783 patients with various IDs we have identified to date, we examined a total of 386 patients with confirmed epimutation and 71 patients with epimutation or uniparental disomy. Consequently, we identified MLID in 29 patients with epimutation confirmed by methylation analysis for multiple ID-associated DMRs using pyrosequencing and/or methylation-specific multiple ligation-dependent probe amplification. MLID was detected in approximately 12% of patients with BWS phenotype and approximately 5% of patients with SRS phenotype, but not in patients with Kagami-Ogata syndrome, Prader-Willi syndrome, or Angelman syndrome phenotypes. We next conducted array-based methylation analysis for 78 DMRs and whole-exome sequencing in the 29 patients, revealing hypomethylation-dominant aberrant methylation patterns in various DMRs of all the patients, eight probably deleterious variants in genes for SCMC in the mothers of patients, and one homozygous deleterious variant in ZNF445 in one patient. These variants did not show gene-specific methylation disturbance patterns. Clinically, neurodevelopmental delay and/or intellectual developmental disorder (ND/IDD) was observed in about half of the MLID patients, with no association with the identified methylation disturbance patterns and genetic variants. Notably, seven patients with BWS phenotype were conceived by assisted reproductive technology (ART). CONCLUSIONS The frequency of MLID was 7.5% (29/386) in IDs caused by confirmed epimutation. Furthermore, we revealed diverse patterns of hypomethylation-dominant methylation defects, nine deleterious variants, ND/IDD complications in about half of the MLID patients, and a high frequency of MLID in ART-conceived patients.
Collapse
Affiliation(s)
- Tatsuki Urakawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
- Department of Pediatrics, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8102, Japan
| | - Hidenobu Soejima
- Division of Molecular Genetics and Epigenetics, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-0937, Japan
| | - Kaori Yamoto
- Department of Biochemistry, Hamamatsu University School of Medicine, 1‑20‑1 Handayama, Higashi‑ku, Hamamatsu, 431‑3192, Japan
| | - Kaori Hara-Isono
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Akie Nakamura
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Sayaka Kawashima
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Hiromune Narusawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Rika Kosaki
- Department of Medical Genetics, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Yutaka Nishimura
- Department of Neonatology, Hiroshima City Hiroshima Citizens Hospital, 7-33 Motomachi, Naka-Ku, Hiroshima, 730-8518, Japan
| | - Kazuki Yamazawa
- Medical Genetics Center, NHO Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-Ku, Tokyo, 152-8902, Japan
| | - Tetsuo Hattori
- Department of Pediatrics, Anjo Kosei Hospital, 28 Higashihirokute, Anjo, 446-8602, Japan
| | - Yukako Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Shouwa‑ku, Nagoya, 466‑8560, Japan
| | - Takanobu Inoue
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta, Midori-Ku, Chiba, 266-0007, Japan
| | - Keiko Matsubara
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
- Division of Diversity Research, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Tsutomu Ogata
- Department of Biochemistry, Hamamatsu University School of Medicine, 1‑20‑1 Handayama, Higashi‑ku, Hamamatsu, 431‑3192, Japan
- Department of Pediatrics, Hamamatsu Medical Center, 328 Tomizuka-Cho, Chuo-Ku, Hamamatsu, 432-8580, Japan
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-Ku, Tokyo, 157-8535, Japan.
| |
Collapse
|
30
|
Sun H, Zhang G, Li N, Bu X. Molecular diagnosis of patients with syndromic short stature identified by trio whole-exome sequencing. Front Genet 2024; 15:1399186. [PMID: 39415983 PMCID: PMC11479978 DOI: 10.3389/fgene.2024.1399186] [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: 03/11/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background Short stature is a complex disorder with phenotypic and genetic heterogeneity. This study aimed to investigate clinical phenotypes and molecular basis of a cohort of patients with short stature. Methods Trio whole-exome sequencing (Trio-WES) was performed to explore the genetic aetiology and obtain a molecular diagnosis in twenty Chinese probands with syndromic and isolated short stature. Results Of the twenty probands, six (6/20, 30%) patients with syndromic short stature obtained a molecular diagnosis. One novel COMP pathogenic variant c.1359delC, p.N453fs*62 and one LZTR1 likely pathogenic variant c.509G>A, p.R170Q were identified in a patient with short stature and skeletal dysplasia. One novel de novo NAA15 pathogenic variant c.63T>G, p.Y21X and one novel de novo KMT2A pathogenic variant c.3516T>A, p.N1172K was identified in two probands with short stature, intellectual disability and abnormal behaviours, respectively. One patient with short stature, cataract, and muscle weakness had a de novo POLG pathogenic variant c.2863 T>C, p.Y955H. One PHEX pathogenic variant c.1104G>A, p.W368X was identified in a patient with short stature and rickets. Maternal uniparental disomy 7 (mUPD7) was pathogenic in a patient with pre and postnatal growth retardation, wide forehead, triangular face, micrognathia and clinodactyly. Thirteen patients with isolated short stature had negative results. Conclusion Trio-WES is an important strategy for identifying genetic variants and UPD in patients with syndromic short stature, in which dual genetic variants are existent in some individuals. It is important to differentiate between syndromic and isolated short stature. Genetic testing has a high yield for syndromic patients but low for isolated patients.
Collapse
Affiliation(s)
- Huihui Sun
- Department of Paediatrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Geng Zhang
- Beijing Chigene Translational Medical Research Center Company, Beijing, China
| | - Na Li
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Xiangfang Bu
- Department of Paediatrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Perge K, Capel E, Senée V, Julier C, Vigouroux C, Nicolino M. Ciliopathies are responsible for short stature and insulin resistance: A systematic review of this clinical association regarding SOFT syndrome. Rev Endocr Metab Disord 2024; 25:827-838. [PMID: 39017987 PMCID: PMC11470920 DOI: 10.1007/s11154-024-09894-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
SOFT syndrome (Short stature-Onychodysplasia-Facial dysmorphism-hypoTrichosis) is a rare primordial dwarfism syndrome caused by biallelic variants in POC1A encoding a centriolar protein. To refine the phenotypic spectrum of SOFT syndrome, recently shown to include metabolic features, we conducted a systematic review of all published cases (19 studies, including 42 patients). The SOFT tetrad affected only 24 patients (57%), while all cases presented with short stature from birth (median height: -5.5SDS([-8.5]-[-2.8])/adult height: 132.5 cm(103.5-148)), which was most often disproportionate (90.5%), with relative macrocephaly. Bone involvement resulted in short hands and feet (100%), brachydactyly (92.5%), metaphyseal (92%) or epiphyseal (84%) anomalies, and/or sacrum/pelvis hypoplasia (58%). Serum IGF-I was increased (median IGF-I level: + 2 SDS ([-0.5]-[+ 3])). Recombinant human growth hormone (rhGH) therapy was stopped for absence/poor growth response (7/9 patients, 78%) and/or hyperglycemia (4/9 patients, 45%). Among 11 patients evaluated, 10 (91%) presented with central distribution of fat (73%), clinical (64%) and/or biological insulin resistance (IR) (100%, median HOMA-IR: 18), dyslipidemia (80%), and hepatic steatosis (100%). Glucose tolerance abnormalities affected 58% of patients aged over 10 years. Patients harbored biallelic missense (52.4%) or truncating (45.2%) POC1A variants. Biallelic null variants, affecting 36% of patients, were less frequently associated with the SOFT tetrad (33% vs 70% respectively, p = 0.027) as compared to other variants, without difference in the prevalence of metabolic abnormalities. POC1A should be sequenced in children with short stature, altered glucose/insulin homeostasis and/or centripetal fat distribution. In patients with SOFT syndrome, rhGH treatment is not indicated, and IR-related complications should be regularly screened and monitored.PROSPERO registration: CRD42023460876.
Collapse
Affiliation(s)
- Kevin Perge
- Pediatric Endocrinology, Diabetology and Metabolism Department, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Bron, France.
- Claude Bernard University, Lyon 1, Lyon, France.
- Paris University, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France.
| | - Emilie Capel
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Valérie Senée
- Paris University, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France
| | - Cécile Julier
- Paris University, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France
| | - Corinne Vigouroux
- Sorbonne University, Inserm U938, Saint-Antoine Research Centre, Institute of Cardiometabolism and Nutrition, Paris, France
- Department of Endocrinology, Diabetology and Reproductive Endocrinology, Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, National Reference Center for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
- Department of Molecular Biology and Genetics, Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, Paris, France
| | - Marc Nicolino
- Pediatric Endocrinology, Diabetology and Metabolism Department, Femme Mère Enfant Hospital, Hospices Civils de Lyon, Bron, France
- Claude Bernard University, Lyon 1, Lyon, France
| |
Collapse
|
32
|
Gaudioso F, Meossi C, Pezzani L, Grilli F, Silipigni R, Russo S, Masciadri M, Vimercati A, Marchisio PG, Bedeschi MF, Milani D. A long way to syndromic short stature. Ital J Pediatr 2024; 50:192. [PMID: 39334216 PMCID: PMC11437795 DOI: 10.1186/s13052-024-01737-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Silver-Russell Syndrome (SRS, MIM #180860) is a clinically and genetically heterogeneous disorder characterized by intrauterine and postnatal growth retardation; SRS is also accompanied by dysmorphic features such as triangular facial appearance, broad forehead, body asymmetry and significant feeding difficulties. The incidence is unknown but estimated at 1:30,000-100,000 live births. The diagnosis of SRS is guided by specific criteria described in the Netchine-Harbison clinical scoring system (NH-CSS). CASE PRESENTATION Hereby we describe four patients with syndromic short stature in whom, despite fitting the criteria for SRS genetic analysis (and one on them even meeting the clinical criteria for SRS), molecular analysis actually diagnosed a different syndrome. Some additional features such as hypotonia, microcephaly, developmental delay and/or intellectual disability, and family history of growth failure, were actually discordant with SRS in our cohort. CONCLUSIONS The clinical resemblance of other short stature syndromes with SRS poses a risk of diagnostic failure, in particular when clinical SRS only criteria are met, allowing SRS diagnosis in the absence of a positive result of a genetic test. The presence of additional features atypical for SRS diagnosis becomes a red flag for a more extensive and thorough analysis. The signs relevant to the differential diagnosis should be valued as much as possible since a correct diagnosis of these patients is the only way to provide the appropriate care pathway, a thorough genetic counselling, prognosis definition, follow up setting, appropriate monitoring and care of possible medical problems.
Collapse
Affiliation(s)
- Federica Gaudioso
- Clinical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, 20122, Italy
| | - Camilla Meossi
- Clinical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, 20122, Italy.
- Unità di Genetica medica, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Via della Commenda, 9, Milano, 20122, Italy.
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy.
| | - Lidia Pezzani
- Clinical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, 20122, Italy
- Paediatric Unit, ASST Papa Giovanni XXIII, Bergamo, 24127, Italy
| | - Federico Grilli
- Clinical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, 20122, Italy
| | - Rosamaria Silipigni
- Laboratory of Medical Genetics, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Silvia Russo
- Experimental Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, Via Ariosto 13, Milano, 20145, Italy
| | - Maura Masciadri
- Experimental Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, Via Ariosto 13, Milano, 20145, Italy
| | - Alessandro Vimercati
- Experimental Research Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, Via Ariosto 13, Milano, 20145, Italy
| | - Paola Giovanna Marchisio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, 20122, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
| | - Maria Francesca Bedeschi
- Clinical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, 20122, Italy
| | - Donatella Milani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, 20122, Italy
| |
Collapse
|
33
|
Kucharska A, Witkowska-Sędek E, Erazmus M, Artemniak-Wojtowicz D, Krajewska M, Pyrżak B. The Effects of Growth Hormone Treatment Beyond Growth Promotion in Patients with Genetic Syndromes: A Systematic Review of the Literature. Int J Mol Sci 2024; 25:10169. [PMID: 39337654 PMCID: PMC11432634 DOI: 10.3390/ijms251810169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Recombinant human growth hormone therapy (rhGH) has been widely accepted as the safe treatment for short stature in children with such genetic syndromes as Prader-Willi syndrome and Turner or Noonan syndrome. Some patients with short stature and rare genetic syndromes are treated with rhGH as growth hormone-deficient individuals or as children born small for their gestational age. After years of experience with this therapy in syndromic short stature, it has been proved that there are some aspects of long-term rhGH treatment beyond growth promotion, which can justify rhGH use in these individuals. This paper summarizes the data of a literature review of the effects of rhGH treatment beyond growth promotion in selected genetic syndromes. We chose three of the most common syndromes, Prader-Willi, Turner, and Noonan, in which rhGH treatment is indicated, and three rarer syndromes, Silver-Russel, Kabuki, and Duchenne muscular dystrophy, in which rhGH treatment is not widely indicated. Many studies have shown a significant impact of rhGH therapy on body composition, resting energy expenditure, insulin sensitivity, muscle tonus, motor function, and mental and behavioral development. Growth promotion is undoubtedly the primary benefit of rhGH therapy; nevertheless, especially with genetic syndromes, the additional effects should also be considered as important indications for this treatment.
Collapse
Affiliation(s)
- Anna Kucharska
- Department of Pediatrics and Endocrinology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.E.); (D.A.-W.); (M.K.); (B.P.)
| | - Ewelina Witkowska-Sędek
- Department of Pediatrics and Endocrinology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.E.); (D.A.-W.); (M.K.); (B.P.)
| | | | | | | | | |
Collapse
|
34
|
Kurup U, Lim DBN, Palau H, Maharaj AV, Ishida M, Davies JH, Storr HL. Approach to the Patient With Suspected Silver-Russell Syndrome. J Clin Endocrinol Metab 2024; 109:e1889-e1901. [PMID: 38888172 PMCID: PMC11403326 DOI: 10.1210/clinem/dgae423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Silver-Russell syndrome (SRS) is a clinical diagnosis requiring the fulfillment of ≥ 4/6 Netchine-Harbison Clinical Scoring System (NH-CSS) criteria. A score of ≥ 4/6 NH-CSS (or ≥ 3/6 with strong clinical suspicion) warrants (epi)genetic confirmation, identifiable in ∼60% patients. The approach to the investigation and diagnosis of SRS is detailed in the only international consensus guidance, published in 2016. In the intervening years, the clinical, biochemical, and (epi)genetic characteristics of SRS have rapidly expanded, largely attributable to advancing molecular genetic techniques and a greater awareness of related disorders. The most common etiologies of SRS remain loss of methylation of chromosome 11p15 (11p15LOM) and maternal uniparental disomy of chromosome 7 (upd(7)mat). Rarer causes of SRS include monogenic pathogenic variants in imprinted (CDKN1C and IGF2) and non-imprinted (PLAG1 and HMGA2) genes. Although the age-specific NH-CSS can identify more common molecular causes of SRS, its use in identifying monogenic causes is unclear. Preliminary data suggest that NH-CSS is poor at identifying many of these cases. Additionally, there has been increased recognition of conditions with phenotypes overlapping with SRS that may fulfill NH-CSS criteria but have distinct genetic etiologies and disease trajectories. This group of conditions is frequently overlooked and under-investigated, leading to no or delayed diagnosis. Like SRS, these conditions are multisystemic disorders requiring multidisciplinary care and tailored management strategies. Early identification is crucial to improve outcomes and reduce the major burden of the diagnostic odyssey for patients and families. This article aims to enable clinicians to identify key features of rarer causes of SRS and conditions with overlapping phenotypes, show a logical approach to the molecular investigation, and highlight the differences in clinical management strategies.
Collapse
Affiliation(s)
- Uttara Kurup
- Centre for Endocrinology, William Harvey Research Institute (WHRI), Charterhouse Square, Barts and the London School of Medicine, London EC1M 6BQ, UK
| | - David B N Lim
- Paediatric Endocrinology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Helena Palau
- Centre for Endocrinology, William Harvey Research Institute (WHRI), Charterhouse Square, Barts and the London School of Medicine, London EC1M 6BQ, UK
| | - Avinaash V Maharaj
- Centre for Endocrinology, William Harvey Research Institute (WHRI), Charterhouse Square, Barts and the London School of Medicine, London EC1M 6BQ, UK
| | - Miho Ishida
- Centre for Endocrinology, William Harvey Research Institute (WHRI), Charterhouse Square, Barts and the London School of Medicine, London EC1M 6BQ, UK
| | - Justin H Davies
- Paediatric Endocrinology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Helen L Storr
- Centre for Endocrinology, William Harvey Research Institute (WHRI), Charterhouse Square, Barts and the London School of Medicine, London EC1M 6BQ, UK
| |
Collapse
|
35
|
Mackay DJG, Gazdagh G, Monk D, Brioude F, Giabicani E, Krzyzewska IM, Kalish JM, Maas SM, Kagami M, Beygo J, Kahre T, Tenorio-Castano J, Ambrozaitytė L, Burnytė B, Cerrato F, Davies JH, Ferrero GB, Fjodorova O, Manero-Azua A, Pereda A, Russo S, Tannorella P, Temple KI, Õunap K, Riccio A, de Nanclares GP, Maher ER, Lapunzina P, Netchine I, Eggermann T, Bliek J, Tümer Z. Multi-locus imprinting disturbance (MLID): interim joint statement for clinical and molecular diagnosis. Clin Epigenetics 2024; 16:99. [PMID: 39090763 PMCID: PMC11295890 DOI: 10.1186/s13148-024-01713-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: 04/25/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Imprinting disorders are rare diseases resulting from altered expression of imprinted genes, which exhibit parent-of-origin-specific expression patterns regulated through differential DNA methylation. A subgroup of patients with imprinting disorders have DNA methylation changes at multiple imprinted loci, a condition referred to as multi-locus imprinting disturbance (MLID). MLID is recognised in most but not all imprinting disorders and is also found in individuals with atypical clinical features; the presence of MLID often alters the management or prognosis of the affected person. Some cases of MLID are caused by trans-acting genetic variants, frequently not in the patients but their mothers, which have counselling implications. There is currently no consensus on the definition of MLID, clinical indications prompting testing, molecular procedures and methods for epigenetic and genetic diagnosis, recommendations for laboratory reporting, considerations for counselling, and implications for prognosis and management. The purpose of this study is thus to cover this unmet need. METHODS A comprehensive literature search was conducted resulting in identification of more than 100 articles which formed the basis of discussions by two working groups focusing on clinical diagnosis (n = 12 members) and molecular testing (n = 19 members). Following eight months of preparations and regular online discussions, the experts from 11 countries compiled the preliminary documentation and determined the questions to be addressed during a face-to-face meeting which was held with the attendance of the experts together with four representatives of patient advocacy organisations. RESULTS In light of available evidence and expert consensus, we formulated 16 propositions and 8 recommendations as interim guidance for the clinical and molecular diagnosis of MLID. CONCLUSIONS MLID is a molecular designation, and for patients with MLID and atypical phenotypes, we propose the alternative term multi-locus imprinting syndrome. Due to the intrinsic variability of MLID, the guidelines underscore the importance of involving experts from various fields to ensure a confident approach to diagnosis, counselling, and care. The authors advocate for global, collaborative efforts in both basic and translational research to tackle numerous crucial questions that currently lack answers, and suggest reconvening within the next 3-5 years to evaluate the research advancements and update this guidance as needed.
Collapse
Affiliation(s)
| | - Gabriella Gazdagh
- Faculty of Medicine, University of Southampton, Southampton, UK
- Wessex Clinical Genetics Service, Princess Anne Hospital, University Hospital Southampton NHS Trust, Southampton, UK
| | - David Monk
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Frederic Brioude
- Centre de Recherche Saint Antoine, Endocrinologie Moléculaire et Pathologies d'empreinte, INSERMSorbonne Université, Hôpital Armand TrousseauAPHP, 75012, Paris, France
| | - Eloise Giabicani
- Centre de Recherche Saint Antoine, Endocrinologie Moléculaire et Pathologies d'empreinte, INSERMSorbonne Université, Hôpital Armand TrousseauAPHP, 75012, Paris, France
| | - Izabela M Krzyzewska
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jennifer M Kalish
- Division of Human Genetics and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Departments of Pediatrics and Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Saskia M Maas
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Jasmin Beygo
- Institut Für Humangenetik, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Tiina Kahre
- Department of Laboratory Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Jair Tenorio-Castano
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- Institute of Medical and Molecular Genetics, INGEMM-Idipaz, Madrid, Spain
| | - Laima Ambrozaitytė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Birutė Burnytė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Flavia Cerrato
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università Degli Studi Della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Justin H Davies
- Faculty of Medicine, University of Southampton, Southampton, UK
- Regional Centre for Paediatric Endocrinology, Faculty of Medicine, Southampton Children's Hospital, University of Southampton, Southampton, UK
| | - Giovanni Battista Ferrero
- Department of Clinical and Biological Science, School of Medicine, Centre for Hemoglobinopathies, AOU San Luigi Gonzaga, University of Turin, Turin, Italy
| | - Olga Fjodorova
- Department of Laboratory Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Africa Manero-Azua
- Rare Diseases Research Group, Molecular (Epi)Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Araba, Spain
| | - Arrate Pereda
- Rare Diseases Research Group, Molecular (Epi)Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Araba, Spain
| | - Silvia Russo
- IRCCS Research Laboratory of Medical Cytogenetics and Molecular Genetics, Istituto Auxologico Italiano, Milan, Italy
| | - Pierpaola Tannorella
- IRCCS Research Laboratory of Medical Cytogenetics and Molecular Genetics, Istituto Auxologico Italiano, Milan, Italy
| | - Karen I Temple
- Faculty of Medicine, University of Southampton, Southampton, UK
- Wessex Clinical Genetics Service, Princess Anne Hospital, University Hospital Southampton NHS Trust, Southampton, UK
| | - Katrin Õunap
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Department of Clinical Genetics, Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Andrea Riccio
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università Degli Studi Della Campania "Luigi Vanvitelli", Caserta, Italy
- Institute of Genetics and Biophysics (IGB),"Adriano Buzzati-Traverso", Consiglio Nazionale Delle Ricerche (CNR), Naples, Italy
| | - Guiomar Perez de Nanclares
- Rare Diseases Research Group, Molecular (Epi)Genetics Laboratory, Bioaraba Health Research Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Araba, Spain
| | - Eamonn R Maher
- Aston Medical School, Aston University, Birmingham, UK
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Pablo Lapunzina
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- Institute of Medical and Molecular Genetics, INGEMM-Idipaz, Madrid, Spain
| | - Irène Netchine
- Centre de Recherche Saint Antoine, Endocrinologie Moléculaire et Pathologies d'empreinte, INSERMSorbonne Université, Hôpital Armand TrousseauAPHP, 75012, Paris, France
| | - Thomas Eggermann
- Institute for Human Genetics and Genome Medicine. Faculty of Medicine, RWTH University Aachen, Aachen, Germany
| | - Jet Bliek
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Zeynep Tümer
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Urzua A, Catena S, Morales P, Lay-Son G. Silver-Russell syndrome-like features in a child with recombinant chromosome 11 derived from maternal pericentric inversion. Clin Dysmorphol 2024; 33:105-109. [PMID: 38818816 DOI: 10.1097/mcd.0000000000000483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Silver-Russell syndrome (SRS) is a well-known syndrome but with heterogeneous etiologies. We present the case of a child with severe SRS-like features resulting from a complex rearrangement of chromosome 11 inherited from his mother. We studied the index case with karyotyping, MS-MLPA and molecular karyotyping. The mother was studied with karyotyping and subtelomeric FISH. We found a child with marked developmental delay and fatal outcome due to failure to thrive, carrying an 11p15 duplication and an 11q25 deletion of maternal origin. We discovered that the mother was a carrier of a pericentric inversion of chromosome 11, with a history of recurrence in other family members who had severe growth retardation and early death. To our knowledge, no similar SRS-like cases have been described in the literature. This report supports the importance of identification the causative genetic mechanism in SRS-like individuals with duplication in 11p15 region due to high risk of recurrence and to provide an appropriate genetic counseling to the family.
Collapse
Affiliation(s)
- Abraham Urzua
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo
- Current affiliation: Laboratorio de Biología Molecular y Citogenética, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sofía Catena
- Jackson Memorial Hospital, University of Miami, USA
| | - Paulina Morales
- Laboratorio de Citogenética y Genética Molecular, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile
| | - Guillermo Lay-Son
- Hospital Padre Hurtado
- Unidad de Genética y Enfermedades Metabólicas, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
37
|
Punt LD, van der Kaay DCM, van Setten PA, de Groote K, Kruijsen AR, Bocca G, de Munnik SA, Renes JS, de Bruin C, Losekoot M, van Duyvenvoorde HA, Wit JM, Joustra SD. IGF1 Haploinsufficiency: Phenotype and Response to Growth Hormone Treatment in 9 Patients. Horm Res Paediatr 2024:1-11. [PMID: 38952118 DOI: 10.1159/000540053] [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: 04/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
INTRODUCTION The clinical features of bi-allelic IGF1 defects are well established, i.e., severe growth failure and microcephaly, delayed psychomotor development, and sensorineural deafness. However, information on clinical and endocrine consequences of heterozygous IGF1 variants and treatment options is scarce. We aimed at extending the knowledge base of the clinical presentation and growth response to recombinant human growth hormone (rhGH) of patients carrying such variants. METHODS Retrospective case series of patients with pathogenic heterozygous IGF1 variants. RESULTS Nine patients from six families were included, harbouring five whole or partial gene deletions and one frameshift variant resulting in a premature stop codon (three de novo, one unknown inheritance). In the other two families, variants segregated with short stature. Mean (SD) birth length was -1.9 (1.3) SDS (n = 7), height -3.8 (0.6) SDS, head circumference -2.5 (0.6) SDS, serum IGF-I -1.9 (0.7) SDS, serum IGFBP-3 1.1 (0.4) SDS (n = 7), and GH peak range 5-31 μg/L (n = 4). Five patients showed feeding problems in infancy. Average height increased after 1 and 2 years of rhGH treatment by 0.8 SDS (range 0.3-1.3 SDS) and 1.3 SDS (range 0.5-2.0 SDS), respectively. Adult height in 2 patients was -2.8 and -1.3 SDS, which was, respectively, 1.3 and 2.9 SDS taller than predicted before start of treatment. CONCLUSION Haploinsufficiency of IGF1 causes a variable phenotype of prenatal and postnatal growth failure, microcephaly, feeding difficulties, low/low-normal serum IGF-I values in contrast to serum IGFBP-3 in the upper-normal range. Treatment with rhGH increased growth in the first 2 years of treatment, and in 2 patients adult height after treatment was higher than predicted at treatment initiation.
Collapse
Affiliation(s)
- Lauren D Punt
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Daniëlle C M van der Kaay
- Division of Paediatric Endocrinology, Department of Paediatrics, Erasmus University Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Petra A van Setten
- Department of Paediatrics, Amalia Childrens Hospital, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Kirsten de Groote
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anne R Kruijsen
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gianni Bocca
- Division of Paediatric Endocrinology, Department of Paediatrics, Beatrix Children's Hospital, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Sonja A de Munnik
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Judith S Renes
- Dutch Growth Research Foundation, Rotterdam, The Netherlands
- Department of Paediatrics, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Christiaan de Bruin
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Monique Losekoot
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Jan M Wit
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sjoerd D Joustra
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
38
|
Yamoto K, Saitsu H, Ohkubo Y, Kagami M, Ogata T. Pathogenic sequence variant and microdeletion affecting HMGA2 in Silver-Russell syndrome: case reports and literature review. Clin Epigenetics 2024; 16:73. [PMID: 38840187 PMCID: PMC11155105 DOI: 10.1186/s13148-024-01688-w] [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: 04/02/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024] Open
Abstract
Silver-Russell syndrome (SRS) is a representative imprinting disorder characterized by pre- and postnatal growth failure. We encountered two Japanese SRS cases with a de novo pathogenic frameshift variant of HMGA2 (NM_003483.6:c.138_141delinsCT, p.(Lys46Asnfs*16)) and a de novo ~ 3.4 Mb microdeletion at 12q14.2-q15 involving HMGA2, respectively. Furthermore, we compared clinical features in previously reported patients with various genetic conditions leading to compromised IGF2 expression, i.e., HMGA2 aberrations, PLAG1 aberrations, IGF2 aberrations, and H19/IGF2:IG-DMR epimutations (hypomethylations). The results provide further support for HMGA2 being involved in the development of SRS and imply some characteristic features in patients with HMGA2 aberrations.
Collapse
Affiliation(s)
- Kaori Yamoto
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1, Handayama, Chuo-ku, Hamamatsu, 431-3192, Japan
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1, Handayama, Chuo-ku, Hamamatsu, 431-3192, Japan
| | - Yumiko Ohkubo
- Department of Pediatrics, Shizuoka Saiseikai Hospital, Shizuoka, Japan
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Tsutomu Ogata
- Department of Biochemistry, Hamamatsu University School of Medicine, 1-20-1, Handayama, Chuo-ku, Hamamatsu, 431-3192, Japan.
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan.
- Department of Pediatrics, Hamamatsu Medical Center, Hamamatsu, Japan.
| |
Collapse
|
39
|
Wu K, Zhu Y, Zhu Q. Prenatal diagnosis of Silver-Russell syndrome with 8q12 deletion including the PLAG1 gene: a case report and review. Front Genet 2024; 15:1387649. [PMID: 38826801 PMCID: PMC11140101 DOI: 10.3389/fgene.2024.1387649] [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: 02/18/2024] [Accepted: 04/30/2024] [Indexed: 06/04/2024] Open
Abstract
Silver-Russell syndrome (SRS) is a clinically and genetically heterogeneous disorder. A retrospective analysis predicted that the live birth prevalence of SRS in Estonia is 1:15,886 [Yakoreva et al., Eur J Hum Genet, 2019, 27(11), 1649-1658]. The most common causative genetic mechanism in the proband is loss of paternal methylation in the imprinted control region 1 (ICR1) at 11p15.5 chromosome. A few studies suggested that inherited or de novo loss-of-function alterations of the PLAG1 gene, including the whole-gene deletion and intragenic pathogenic variants, could cause a rare type of SRS. To date, less than 20 unrelated PLAG1-related SRS cases have been reported, and the clinical information about these cases is limited. We report the first prenatal case of SRS with 8q12 deletion (including the PLAG1 gene). The fetus presented with intrauterine growth retardation, small for gestational age, relative macrocephaly at birth, and a protruding forehead. Unlike classical SRS cases, the fetus had micrognathia and did not show body asymmetry. We hope that the literature review in this study provides new insights into genotype-phenotype relationships of PLAG1-related SRS.
Collapse
Affiliation(s)
- Ke Wu
- Laboratory of Prenatal Diagnosis Center, Quzhou Maternal and Child Health Care Hospital, Quzhou, Zhejiang, China
| | - Yuying Zhu
- Prenatal Diagnosis Center, Quzhou Maternal and Child Health Care Hospital, Quzhou, Zhejiang, China
| | - Qiumin Zhu
- Obstetrics Department, Quzhou Maternal and Child Health Care Hospital, Quzhou, Zhejiang, China
| |
Collapse
|
40
|
Ciancia S, Madeo SF, Calabrese O, Iughetti L. The Approach to a Child with Dysmorphic Features: What the Pediatrician Should Know. CHILDREN (BASEL, SWITZERLAND) 2024; 11:578. [PMID: 38790573 PMCID: PMC11120268 DOI: 10.3390/children11050578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
The advancement of genetic knowledge and the discovery of an increasing number of genetic disorders has made the role of the geneticist progressively more complex and fundamental. However, most genetic disorders present during childhood; thus, their early recognition is a challenge for the pediatrician, who will be also involved in the follow-up of these children, often establishing a close relationship with them and their families and becoming a referral figure. In this review, we aim to provide the pediatrician with a general knowledge of the approach to treating a child with a genetic syndrome associated with dysmorphic features. We will discuss the red flags, the most common manifestations, the analytic collection of the family and personal medical history, and the signs that should alert the pediatrician during the physical examination. We will offer an overview of the physical malformations most commonly associated with genetic defects and the way to describe dysmorphic facial features. We will provide hints about some tools that can support the pediatrician in clinical practice and that also represent a useful educational resource, either online or through apps downloaded on a smartphone. Eventually, we will offer an overview of genetic testing, the ethical considerations, the consequences of incidental findings, and the main indications and limitations of the principal technologies.
Collapse
Affiliation(s)
- Silvia Ciancia
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Largo del Pozzo 71, 41124 Modena, Italy
| | - Simona Filomena Madeo
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Largo del Pozzo 71, 41124 Modena, Italy
| | - Olga Calabrese
- Medical Genetics Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Lorenzo Iughetti
- Pediatric Unit, Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Largo del Pozzo 71, 41124 Modena, Italy
| |
Collapse
|
41
|
Cong X, Zhang T, Li Z, Luo X, Hu L, Liu W. Prenatal diagnosis of a trisomy 7 mosaic case: CMA, CNV-seq, karyotyping, interphase FISH, and MS-MLPA, which technique to choose? BMC Pregnancy Childbirth 2024; 24:338. [PMID: 38702634 PMCID: PMC11067092 DOI: 10.1186/s12884-024-06522-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: 12/07/2023] [Accepted: 04/15/2024] [Indexed: 05/06/2024] Open
Abstract
OBJECTIVE This study aims to perform a prenatal genetic diagnosis of a high-risk fetus with trisomy 7 identified by noninvasive prenatal testing (NIPT) and to evaluate the efficacy of different genetic testing techniques for prenatal diagnosis of trisomy mosaicism. METHODS For prenatal diagnosis of a pregnant woman with a high risk of trisomy 7 suggested by NIPT, karyotyping and chromosomal microarray analysis (CMA) were performed on an amniotic fluid sample. Low-depth whole-genome copy number variation sequencing (CNV-seq) and fluorescence in situ hybridization (FISH) were used to clarify the results further. In addition, methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) was performed to analyze the possibility of uniparental disomy(UPD). RESULTS Amniotic fluid karyotype analysis revealed a 46, XX result. Approximately 20% mosaic trisomy 7 was detected according to the CMA result. About 16% and 4% of mosaicism was detected by CNV-seq and FISH, respectively. MS-MLPA showed no methylation abnormalities. The fetal ultrasound did not show any detectable abnormalities except for mild intrauterine growth retardation seen at 39 weeks of gestation. After receiving genetic counseling, the expectant mother decided to continue the pregnancy, and follow-up within three months of delivery was normal. CONCLUSION In high-risk NIPT diagnosis, a combination of cytogenetic and molecular genetic techniques proves fruitful in detecting low-level mosaicism. Furthermore, the exclusion of UPD on chromosome 7 remains crucial when NIPT indicates a positive prenatal diagnosis of trisomy 7.
Collapse
Affiliation(s)
- Xiaoyi Cong
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China
- Longgang District Key Laboratory for Birth Defects Prevention, Shenzhen, 518172, China
| | - Tong Zhang
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China
- Longgang District Key Laboratory for Birth Defects Prevention, Shenzhen, 518172, China
| | - Zhenming Li
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China
- Longgang District Key Laboratory for Birth Defects Prevention, Shenzhen, 518172, China
| | - Xiaojin Luo
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China
- Longgang District Key Laboratory for Birth Defects Prevention, Shenzhen, 518172, China
| | - Liang Hu
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China
- Longgang District Key Laboratory for Birth Defects Prevention, Shenzhen, 518172, China
| | - Weiqiang Liu
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, China.
- Longgang District Key Laboratory for Birth Defects Prevention, Shenzhen, 518172, China.
| |
Collapse
|
42
|
Cavarzere P, Pietrobelli A, Gandini A, Munari S, Baffico AM, Maffei M, Gaudino R, Guzzo A, Arrigoni M, Coviello D, Piacentini G, Antoniazzi F. Role of genetic investigation in the diagnosis of short stature in a cohort of Italian children. J Endocrinol Invest 2024; 47:1237-1250. [PMID: 38087044 DOI: 10.1007/s40618-023-02243-9] [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] [Received: 07/14/2023] [Accepted: 11/04/2023] [Indexed: 04/23/2024]
Abstract
BACKGROUND Short stature (SS) is defined as height more than 2 standard deviations below the mean for age and sex. Hypothyroidism, celiac disease, growth hormone deficiency, hormonal abnormalities, and genetic conditions are among its causes. A wide range of conditions often due to largely unknown genetic variants can elude conventional diagnostic workup. AIM We used next-generation sequencing (NGS) to better understand the etiology of SS in a cohort of Italian children. PATIENTS AND METHODS The study sample was 125 children with SS of unknown origin referred to our Institute between 2015 and 2021. All had undergone complete auxological and hormonal investigations to exclude common causes of SS. Genetic analysis was performed using a NGS panel of 104 genes. Clinical data were reviewed to clarify the pathogenicity of the variants detected. RESULTS In this cohort, 43 potentially causing variants were identified in 38 children. A syndromic genetic condition was diagnosed in 7: Noonan syndrome in 3, Leri-Weill syndrome in 3, and hypochondroplasia in 1. Moreover, 8 benign variants and other 37 like benign variants were found. In 88 children, 179 variants of uncertain significance (VUS) were identified. No variant was found in 16 children. CONCLUSION Genetic analysis is a useful tool in the diagnostic workup of patients with SS, in adapting management and treatment, and in identifying syndromes with mild atypical clinical features. The role of VUS should not be underestimated, particularly when multiple VUS with possible mutual worsening effects are present in the same child.
Collapse
Affiliation(s)
- P Cavarzere
- Pediatric Division, Department of Pediatrics, University Hospital of Verona (Full Member of European Reference Network Endo-ERN), Verona, Italy.
- Department of Pediatrics, Child and Mother's Hospital, Piazzale Stefani 1, 37126, Verona, Italy.
| | - A Pietrobelli
- Pediatric Division, Department of Pediatrics, University Hospital of Verona (Full Member of European Reference Network Endo-ERN), Verona, Italy
- Department Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Clinic, University of Verona, Verona, Italy
| | - A Gandini
- Department Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Clinic, University of Verona, Verona, Italy
| | - S Munari
- Pediatric Division, Department of Pediatrics, University Hospital of Verona (Full Member of European Reference Network Endo-ERN), Verona, Italy
| | - A M Baffico
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - M Maffei
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - R Gaudino
- Pediatric Division, Department of Pediatrics, University Hospital of Verona (Full Member of European Reference Network Endo-ERN), Verona, Italy
- Department Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Clinic, University of Verona, Verona, Italy
| | - A Guzzo
- Laboratory Unit, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - M Arrigoni
- Pediatric Division, Department of Pediatrics, University Hospital of Verona (Full Member of European Reference Network Endo-ERN), Verona, Italy
| | - D Coviello
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - G Piacentini
- Pediatric Division, Department of Pediatrics, University Hospital of Verona (Full Member of European Reference Network Endo-ERN), Verona, Italy
- Department Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Clinic, University of Verona, Verona, Italy
| | - F Antoniazzi
- Pediatric Division, Department of Pediatrics, University Hospital of Verona (Full Member of European Reference Network Endo-ERN), Verona, Italy
- Department Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Clinic, University of Verona, Verona, Italy
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Regional Center for the Diagnosis and Treatment of Children and Adolescents with Rare Skeletal Disorders, Pediatric Clinic, University of Verona, Verona, Italy
| |
Collapse
|
43
|
Boro H, Patra S, Pasam KK, Dalvi M, Bundela V. Russell-Silver Syndrome With Growth Hormone Deficiency. Cureus 2024; 16:e60018. [PMID: 38854326 PMCID: PMC11162538 DOI: 10.7759/cureus.60018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
Russell-Silver syndrome (RSS) is a rare genetic disorder characterized by intrauterine growth restriction (IUGR), postnatal growth failure, and distinctive dysmorphic features. We present a case of a four-year-old male presenting with a slow growth velocity with a history of IUGR and surgical interventions, exhibiting classic RSS features. Laboratory investigations revealed low insulin-like growth factor 1 (IGF-1) and low growth hormone (GH) levels on stimulation tests. Clinical exome sequencing revealed a de novo mutation in the insulin-like growth factor 2 (IGF2) gene. Additionally, a variant of uncertain significance in the DHX37 gene was noted in the patient and the asymptomatic father. After genetic counseling, recombinant GH therapy was initiated. This case underscores the genetic complexity of RSS and highlights the importance of early diagnosis, genetic testing, and multidisciplinary management in optimizing outcomes for patients with RSS.
Collapse
Affiliation(s)
- Hiya Boro
- Endocrinology, Aadhar Health Institute, Hisar, IND
| | - Shinjan Patra
- Endocrinology, All India Institute of Medical Sciences, Nagpur, IND
| | | | - Mazhar Dalvi
- Endocrinology, Mediclinic Al Noor Hospital, Abu Dhabi, ARE
| | | |
Collapse
|
44
|
Prodani C, Irvine EE, Sardini A, Gleneadie HJ, Dimond A, Van de Pette M, John R, Kokkinou M, Howes O, Withers DJ, Ungless MA, Merkenschlager M, Fisher AG. Protein restriction during pregnancy alters Cdkn1c silencing, dopamine circuitry and offspring behaviour without changing expression of key neuronal marker genes. Sci Rep 2024; 14:8528. [PMID: 38609446 PMCID: PMC11014953 DOI: 10.1038/s41598-024-59083-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/07/2024] [Indexed: 04/14/2024] Open
Abstract
We tracked the consequences of in utero protein restriction in mice throughout their development and life course using a luciferase-based allelic reporter of imprinted Cdkn1c. Exposure to gestational low-protein diet (LPD) results in the inappropriate expression of paternally inherited Cdkn1c in the brains of embryonic and juvenile mice. These animals were characterised by a developmental delay in motor skills, and by behavioural alterations indicative of reduced anxiety. Exposure to LPD in utero resulted in significantly more tyrosine hydroxylase positive (dopaminergic) neurons in the midbrain of adult offspring as compared to age-matched, control-diet equivalents. Positron emission tomography (PET) imaging revealed an increase in striatal dopamine synthesis capacity in LPD-exposed offspring, where elevated levels of dopamine correlated with an enhanced sensitivity to cocaine. These data highlight a profound sensitivity of the developing epigenome to gestational protein restriction. Our data also suggest that loss of Cdkn1c imprinting and p57KIP2 upregulation alters the cellular composition of the developing midbrain, compromises dopamine circuitry, and thereby provokes behavioural abnormalities in early postnatal life. Molecular analyses showed that despite this phenotype, exposure to LPD solely during pregnancy did not significantly change the expression of key neuronal- or dopamine-associated marker genes in adult offspring.
Collapse
Affiliation(s)
- Chiara Prodani
- Epigenetic Memory Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Elaine E Irvine
- Metabolic Signalling Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Alessandro Sardini
- Whole Animal Physiology and Imaging, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Hannah J Gleneadie
- Epigenetic Memory Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Andrew Dimond
- Epigenetic Memory Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Mathew Van de Pette
- MRC Toxicology Unit, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QR, UK
| | - Rosalind John
- Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Michelle Kokkinou
- Psychiatric Imaging Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Dominic J Withers
- Metabolic Signalling Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Mark A Ungless
- MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Matthias Merkenschlager
- Lymphocyte Development Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Amanda G Fisher
- Epigenetic Memory Group, MRC LMS, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
| |
Collapse
|
45
|
Darneau D, Giabicani E, Netchine I, Pham A. Perinatal features of children with Silver-Russell syndrome due to 11p15 loss of methylation. Front Pediatr 2024; 12:1367433. [PMID: 38638586 PMCID: PMC11024461 DOI: 10.3389/fped.2024.1367433] [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: 01/08/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Background A diagnosis of Silver-Russell syndrome (SRS), a rare imprinting disorder responsible for foetal growth restriction, is considered for patients presenting at least four criteria of the Netchine-Harbison clinical scoring system (NH-CSS). Certain items of the NH-CSS are not assessable until the age of 2 years. The objective was to determine perinatal characteristics of children with SRS to allow an early diagnosis. Methods We retrospectively compared the perinatal characteristics of children with SRS (n = 17) with those of newborns small for gestational age (SGA) due to placental insufficiency (PI) (n = 21). Results Children with SRS showed earlier and more severely altered foetal biometry than SGA newborns due to PI. Twenty-three percent of patients with SRS showed uterine artery Doppler anomalies. SRS children were significantly smaller at birth (birth length <-3 SDS in 77% of cases in the SRS group vs. 15% in the PI group, p = 0.0001). Conclusion The diagnosis of SRS must be evoked in the neonatal period for SGA newborns with a growth delay present from the second trimester of pregnancy, a birth length <-3 SDS and a relative macrocephaly. Doppler anomalies, classically used to orient the cause of SGA towards PI, did not rule out the diagnosis of SRS.
Collapse
Affiliation(s)
- Diane Darneau
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, APHP, Hôpital Armand Trousseau, Endocrinologie Moléculaire et Pathologies d’Empreinte, Paris, France
| | - Eloïse Giabicani
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, APHP, Hôpital Armand Trousseau, Endocrinologie Moléculaire et Pathologies d’Empreinte, Paris, France
| | - Irène Netchine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, APHP, Hôpital Armand Trousseau, Endocrinologie Moléculaire et Pathologies d’Empreinte, Paris, France
| | - Aurélie Pham
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, APHP, Hôpital Armand Trousseau, Service de Néonatologie, Paris, France
| |
Collapse
|
46
|
Klein Haneveld MJ, Hieltjes IJ, Langendam MW, Cornel MC, Gaasterland CMW, van Eeghen AM. Improving care for rare genetic neurodevelopmental disorders: A systematic review and critical appraisal of clinical practice guidelines using AGREE II. Genet Med 2024; 26:101071. [PMID: 38224026 DOI: 10.1016/j.gim.2024.101071] [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: 09/15/2023] [Revised: 12/08/2023] [Accepted: 12/19/2023] [Indexed: 01/16/2024] Open
Abstract
PURPOSE Rare genetic neurodevelopmental disorders associated with intellectual disability require lifelong multidisciplinary care. Clinical practice guidelines may support healthcare professionals in their daily practice, but guideline development for rare conditions can be challenging. In this systematic review, the characteristics and methodological quality of internationally published recommendations for this population are described to provide an overview of current guidelines and inform future efforts of European Reference Network ITHACA (Intellectual disability, TeleHealth, Autism, and Congenital Anomalies). METHODS MEDLINE, Embase, and Orphanet were systematically searched to identify guidelines for conditions classified as "rare genetic intellectual disability" (ORPHA:183757). Methodological quality was assessed using the Appraisal of Guidelines, Research, and Evaluation II tool. RESULTS Seventy internationally published guidelines, addressing the diagnosis and/or management of 28 conditions, were included. The methodological rigor of development was highly variable with limited reporting of literature searches and consensus methods. Stakeholder involvement and editorial independence varied as well. Implementation was rarely addressed. CONCLUSION Comprehensive, high-quality guidelines are lacking for many rare genetic neurodevelopmental disorders. Use and transparent reporting of sound development methodologies, active involvement of affected individuals and families, robust conflict of interest procedures, and attention to implementation are vital for enhancing the impact of clinical practice recommendations.
Collapse
Affiliation(s)
- Mirthe J Klein Haneveld
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands; European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability ERN-ITHACA, Clinical Genetics Department, Robert Debré University Hospital, Paris, France; Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Iméze J Hieltjes
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands; Knowledge Institute of the Dutch Association of Medical Specialists, Utrecht, The Netherlands
| | - Miranda W Langendam
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Martina C Cornel
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Human Genetics, Amsterdam, The Netherlands
| | - Charlotte M W Gaasterland
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands; European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability ERN-ITHACA, Clinical Genetics Department, Robert Debré University Hospital, Paris, France; Knowledge Institute of the Dutch Association of Medical Specialists, Utrecht, The Netherlands
| | - Agnies M van Eeghen
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands; European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability ERN-ITHACA, Clinical Genetics Department, Robert Debré University Hospital, Paris, France; Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands; Advisium, 's Heeren Loo Zorggroep, Amersfoort, The Netherlands.
| |
Collapse
|
47
|
Ventresca S, Lepri FR, Criscuolo S, Bottaro G, Novelli A, Loche S, Cappa M. Case report: Long term response to growth hormone in a child with Silver-Russell syndrome-like phenotype due to a novel paternally inherited IGF2 variant. Front Endocrinol (Lausanne) 2024; 15:1364234. [PMID: 38596219 PMCID: PMC11002242 DOI: 10.3389/fendo.2024.1364234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/12/2024] [Indexed: 04/11/2024] Open
Abstract
Silver-Russell syndrome (SRS, OMIM, 180860) is a rare genetic disorder with a wide spectrum of symptoms. The most common features are intrauterine growth retardation (IUGR), poor postnatal development, macrocephaly, triangular face, prominent forehead, body asymmetry, and feeding problems. The diagnosis of SRS is based on a combination of clinical features. Up to 60% of SRS patients have chromosome 7 or 11 abnormalities, and <1% show abnormalities in IGF2 signaling pathway genes (IGF2, HMGA2, PLAG1 and CDKN1C). The underlying genetic cause remains unknown in about 40% of cases (idiopathic SRS). We report a novel IGF2 variant c.[-6-2A>G] (NM_000612) in a child with severe IUGR and clinical features of SRS and confirm the utility of targeted exome sequencing in patients with negative results to common genetic analyses. In addition, we report that long-term growth hormone treatment improves height SDS in this patient.
Collapse
Affiliation(s)
- Silvia Ventresca
- Pediatric Section, University Hospital Arcispedale Sant’Anna, University of Ferrara, Ferrara, Italy
- Endocrinology and Diabetology Unit, Pediatric University Department, Bambino Gesù Children’s Hospital, Rome, Italy
| | | | - Sabrina Criscuolo
- Endocrinology and Diabetology Unit, Pediatric University Department, Bambino Gesù Children’s Hospital, Rome, Italy
- Pediatric University Department, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Giorgia Bottaro
- Endocrinology and Diabetology Unit, Pediatric University Department, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Sandro Loche
- Research Area for Innovative Therapies in Endocrinopathies, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marco Cappa
- Research Area for Innovative Therapies in Endocrinopathies, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
48
|
张 余, 罗 飞. [Recent advances in the genetic etiology of central precocious puberty]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:302-307. [PMID: 38557384 PMCID: PMC10986386 DOI: 10.7499/j.issn.1008-8830.2309098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/29/2024] [Indexed: 04/04/2024]
Abstract
Central precocious puberty (CPP) is a developmental disorder caused by early activation of the hypothalamic-pituitary-gonadal axis. The incidence of CPP is rapidly increasing, but the underlying mechanisms are not fully understood. Previous studies have shown that gain-of-function mutations in the KISS1R and KISS1 genes and loss-of-function mutations in the MKRN3, LIN28, and DLK1 genes may lead to early initiation of pubertal development. Recent research has also revealed the significant role of epigenetic factors such as DNA methylation and microRNAs in the regulation of gonadotropin-releasing hormone neurons, as well as the modulating effect of gene networks involving multiple variant genes on pubertal initiation. This review summarizes the genetic etiology and pathogenic mechanisms underlying CPP.
Collapse
|
49
|
Amin AK, Krause J, Eggermann T. 11p13 microduplication: a differential diagnosis of Silver-Russell syndrome? Mol Cytogenet 2024; 17:5. [PMID: 38486332 PMCID: PMC10941370 DOI: 10.1186/s13039-024-00672-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/21/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Silver-Russel syndrome (SRS) is a congenital disorder which is mainly characterized by intrauterine and postnatal growth retardation, relative macrocephaly, and characteristic (facial) dysmorphisms. The majority of patients shows a hypomethylation of the imprinting center region 1 (IC1) in 11p15 and maternal uniparental disomy of chromosome 7 (upd(7)mat), but in addition a broad spectrum of copy number variations (CNVs) and monogenetic variants (SNVs) has been reported in this cohort. These heterogeneous findings reflect the clinical overlap of SRS with other congenital disorders, but some of the CNVs are recurrent and have therefore been suggested as SRS-associated loci. However, this molecular heterogeneity makes the decision on the diagnostic workup of patients with SRS features challenging. CASE PRESENTATION A girl with clinical features of SRS but negatively tested for the IC1 hypomethylation and upd(7)mat was analyzed by whole genome sequencing in order to address both CNVs and SNVs in the same run. We identified a 11p13 microduplication affecting a region overlapping with a variant reported in a previously published patient with clinical features of Silver-Russel syndrome. CONCLUSIONS The identification of a 11p13 microduplication in a patient with SRS features confirms the considerable contribution of CNVs to SRS-related phenotypes, and it strengthens the evidence for a 11p13 microduplication syndrome as a differential diagnosis SRS. Furthermore, we could confirm that WGS is a valuable diagnostic tool in patients with SRS and related disorders, as it allows CNVs and SNV detection in the same run, thereby avoiding a time-consuming diagnostic testing process.
Collapse
Affiliation(s)
- Asmaa K Amin
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Jeremias Krause
- Institute for Human Genetics and Genome Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Thomas Eggermann
- Institute for Human Genetics and Genome Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
50
|
Maharaj AV, Cottrell E, Thanasupawat T, Joustra SD, Triggs-Raine B, Fujimoto M, Kant SG, van der Kaay D, Clement-de Boers A, Brooks AS, Aguirre GA, Martín del Estal I, Castilla de Cortázar Larrea MI, Massoud A, van Duyvenvoorde HA, De Bruin C, Hwa V, Klonisch T, Hombach-Klonisch S, Storr HL. Characterization of HMGA2 variants expands the spectrum of Silver-Russell syndrome. JCI Insight 2024; 9:e169425. [PMID: 38516887 PMCID: PMC11063932 DOI: 10.1172/jci.insight.169425] [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: 02/16/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Silver-Russell syndrome (SRS) is a heterogeneous disorder characterized by intrauterine and postnatal growth retardation. HMGA2 variants are a rare cause of SRS and its functional role in human linear growth is unclear. Patients with suspected SRS negative for 11p15LOM/mUPD7 underwent whole-exome and/or targeted-genome sequencing. Mutant HMGA2 protein expression and nuclear localization were assessed. Two Hmga2-knockin mouse models were generated. Five clinical SRS patients harbored HMGA2 variants with differing functional impacts: 2 stop-gain nonsense variants (c.49G>T, c.52C>T), c.166A>G missense variant, and 2 frameshift variants (c.144delC, c.145delA) leading to an identical, extended-length protein. Phenotypic features were highly variable. Nuclear localization was reduced/absent for all variants except c.166A>G. Homozygous knockin mice recapitulating the c.166A>G variant (Hmga2K56E) exhibited a growth-restricted phenotype. An Hmga2Ter76-knockin mouse model lacked detectable full-length Hmga2 protein, similarly to patient 3 and 5 variants. These mice were infertile, with a pygmy phenotype. We report a heterogeneous group of individuals with SRS harboring variants in HMGA2 and describe the first Hmga2 missense knockin mouse model (Hmga2K56E) to our knowledge causing a growth-restricted phenotype. In patients with clinical features of SRS but negative genetic screening, HMGA2 should be included in next-generation sequencing testing approaches.
Collapse
Affiliation(s)
- Avinaash V. Maharaj
- Centre for Endocrinology, William Harvey Research Institute, QMUL, London, United Kingdom
| | - Emily Cottrell
- Centre for Endocrinology, William Harvey Research Institute, QMUL, London, United Kingdom
| | - Thatchawan Thanasupawat
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sjoerd D. Joustra
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Centre, Leiden, Netherlands
| | - Barbara Triggs-Raine
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Masanobu Fujimoto
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Sarina G. Kant
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Centre, Leiden, Netherlands
| | - Danielle van der Kaay
- Division of Paediatric Endocrinology, Department of Paediatrics, Erasmus University Medical Centre, Sophia Children’s Hospital, Rotterdam, Netherlands
| | - Agnes Clement-de Boers
- Department of Paediatrics, Juliana Children’s Hospital/Haga Teaching Hospital, The Hague, Netherlands
| | - Alice S. Brooks
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | | | | | | | - Ahmed Massoud
- Department of Paediatrics and Child Health, HCA Healthcare UK, London, United Kingdom
| | - Hermine A. van Duyvenvoorde
- Laboratory for Diagnostic Genome analysis (LDGA), Department of Clinical Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Christiaan De Bruin
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Centre, Leiden, Netherlands
| | - Vivian Hwa
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pathology, and
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pathology, and
| | - Helen L. Storr
- Centre for Endocrinology, William Harvey Research Institute, QMUL, London, United Kingdom
| |
Collapse
|