1
|
Kawashima-Sonoyama Y, Wada K, Yamamoto K, Fujimoto M, Namba N, Taketani T. Clinical characteristics of and growth hormone treatment effects on short stature with type 1 insulin-like growth factor receptor (IGF1R) gene alteration. Endocr J 2024; 71:687-694. [PMID: 38710621 DOI: 10.1507/endocrj.ej23-0680] [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] [Indexed: 05/08/2024] Open
Abstract
Short stature with IGF-1 receptor (IGF1R) gene alteration is known as small-for-gestational-age (SGA) short stature with elevated serum IGF1 levels. Its prevalence and clinical characteristics remain unclear. No adapted treatment is available for short stature related to IGF1R gene alteration in Japan, and genetic testing is not yet widely accessible. We investigated short stature with IGF1R gene alterations and analyzed the clinical data of 13 patients using the results of questionnaires issued to the Japanese Society for Pediatric Endocrinology. Four cases were caused by a deletion of chromosome 15q26.3, and eight were caused by heterozygous pathogenic variants in the IGF1R gene. Cases with deletions showed a more severe degree of growth impairment (-4.5 ± 0.43 SD) than those caused by pathological variants (-2.71 ± 0.15 SD) and were accompanied by neurodevelopmental delay. However, cases caused by pathological variants lacked distinctive features. Only three of the 12 cases demonstrated serum IGF1 values exceeding +2 SD, and the other three had values below 0 SD. Four patients did not meet the criteria for SGA at birth. Six patients received GH therapy for SGA short stature and showed improvement in growth rate without any side effects or elevated serum IGF1 levels during treatment. Elevated IGF1 levels (over +2 SD) after GH treatment should be considered a suspicious finding. Owing to the lack of distinctive features, there was a possibility of undiagnosed cases of this condition. Promoting genetic testing and clinical trials on GH administration for this condition is recommended.
Collapse
Affiliation(s)
| | - Keisuke Wada
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane 693-8501, Japan
| | - Kei Yamamoto
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane 693-8501, Japan
| | - Masanobu Fujimoto
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Tottori 683-8504, Japan
| | - Noriyuki Namba
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Tottori 683-8504, Japan
| | - Takeshi Taketani
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane 693-8501, Japan
| |
Collapse
|
2
|
Liu L, Wen Y, Ni Q, Chen L, Wang H. Prenatal ethanol exposure and changes in fetal neuroendocrine metabolic programming. Biol Res 2023; 56:61. [PMID: 37978540 PMCID: PMC10656939 DOI: 10.1186/s40659-023-00473-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: 06/07/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Prenatal ethanol exposure (PEE) (mainly through maternal alcohol consumption) has become widespread. However, studies suggest that it can cause intrauterine growth retardation (IUGR) and multi-organ developmental toxicity in offspring, and susceptibility to various chronic diseases (such as neuropsychiatric diseases, metabolic syndrome, and related diseases) in adults. Through ethanol's direct effects and its indirect effects mediated by maternal-derived glucocorticoids, PEE alters epigenetic modifications and organ developmental programming during fetal development, which damages the offspring health and increases susceptibility to various chronic diseases after birth. Ethanol directly leads to the developmental toxicity of multiple tissues and organs in many ways. Regarding maternal-derived glucocorticoid-mediated IUGR, developmental programming, and susceptibility to multiple conditions after birth, ethanol induces programmed changes in the neuroendocrine axes of offspring, such as the hypothalamus-pituitary-adrenal (HPA) and glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axes. In addition, the differences in ethanol metabolic enzymes, placental glucocorticoid barrier function, and the sensitivity to glucocorticoids in various tissues and organs mediate the severity and sex differences in the developmental toxicity of ethanol exposure during pregnancy. Offspring exposed to ethanol during pregnancy have a "thrifty phenotype" in the fetal period, and show "catch-up growth" in the case of abundant nutrition after birth; when encountering adverse environments, these offspring are more likely to develop diseases. Here, we review the developmental toxicity, functional alterations in multiple organs, and neuroendocrine metabolic programming mechanisms induced by PEE based on our research and that of other investigators. This should provide new perspectives for the effective prevention and treatment of ethanol developmental toxicity and the early prevention of related fetal-originated diseases.
Collapse
Affiliation(s)
- Liang Liu
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yinxian Wen
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Qubo Ni
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Joint Disease Research Center of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| |
Collapse
|
3
|
Wannes S, El Ahmer I, Rjiba K, Jemmali N, Abdallah HH, Haj RB, Achour A, Bouzidi H, Saad A, Mougou S, Mahjoub B. Response to growth hormone therapy in ring chromosome 15: Review and evidence from a new case on possible beneficial effect in neurodevelopment. Growth Horm IGF Res 2023; 71:101550. [PMID: 37531800 DOI: 10.1016/j.ghir.2023.101550] [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: 05/27/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Type 1 Insulin-like Growth Factor Receptor(IGF1R) plays a fundamental role in normal growth and development. Its disruption is usually characterized by severe intrauterine and postnatal growth retardation, microcephaly and neurodevelopmental delay.The efficacy of recombinant human growth hormone treatment remains a challenge for children with IGF1 resistance and pathogenic mutations of IGF1R, with limited data in patients carrying the most severe form of IGF1R defect, the ring chromosome 15. SUBJECT AND METHOD We tested a high dose of rhGH in a new patient with ring chromosome 15, as confirmed by karyotype and CGH array. We performed a systematic review, and all published r(15) syndrome cases treated by growth hormone(GH) up to April 2023 were searched, and their response to GH therapy was recorded and summarized. RESULTS Twelve patients with ring chromosome 15 received GH therapy according to a literature review. We expand the spectrum by the 13th case treated by GH, and we report an impressive improvement in intellectual performance and progressive catch-up growth after 5 and 20 months of follow-up. By introducing our new case in the analysis, the sex ratio was 3:10, and GH therapy was started at the age of 5.5 (3/9.4) (years) for an age of diagnosis of 4.75 (1.3/9.5) (years). The height before GH therapy was -5.1(-5.9/-4.1) SDS. The median duration of treatment was 1.7(0.9/2) (years), with a median height gain of 1(0.3/1.8) SDS and an improvement in growth velocity of 4.1(2.8/5.3) (cm/year). CONCLUSION GH seems to be effective for r(15) syndrome patients with short stature.
Collapse
Affiliation(s)
- Selmen Wannes
- Department of Pediatrics, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia; Faculty of Medicine of Monastir, University of Monastir, 5019 Monastir, Tunisia; Department of Pediatrics, Mouwasat Hospital, Imam Al Termithy Street, Uhud, 32263 Dammam, Saudi Arabia; Applied Epidemiology in Maternal and child Health Research Laboratory (LR 12 SP 17), 5100, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia.
| | - Ikram El Ahmer
- Department of Pediatrics, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia; Department of Medical Biology, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia; Faculty of Pharmacy of Monastir, University of Monastir, 5019 Monastir, Tunisia
| | - Khouloud Rjiba
- Department of Cytogenetic and Reproductive Biology, Farhat Hached University Hospital, 4000 Sousse, Tunisia
| | - Nessrine Jemmali
- Department of Pediatrics, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia; Faculty of Medicine of Monastir, University of Monastir, 5019 Monastir, Tunisia
| | - Hamza Haj Abdallah
- Department of Cytogenetic and Reproductive Biology, Farhat Hached University Hospital, 4000 Sousse, Tunisia
| | - Rania Bel Haj
- Department of Psychiatric, Fattouma Bourguiba University Hospital, 5019 Monastir, Tunisia
| | - Asma Achour
- Department of Radiology, Fattouma Bourguiba University Hospital, 5019 Monastir, Tunisia
| | - Hassan Bouzidi
- Department of Medical Biology, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia; Faculty of Pharmacy of Monastir, University of Monastir, 5019 Monastir, Tunisia
| | - Ali Saad
- Department of Cytogenetic and Reproductive Biology, Farhat Hached University Hospital, 4000 Sousse, Tunisia; Faculty of Medicine of Sousse, University of Sousse, 4000 Sousse, Tunisia
| | - Soumaya Mougou
- Department of Cytogenetic and Reproductive Biology, Farhat Hached University Hospital, 4000 Sousse, Tunisia; Faculty of Medicine of Sousse, University of Sousse, 4000 Sousse, Tunisia
| | - Bahri Mahjoub
- Department of Pediatrics, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia; Faculty of Medicine of Monastir, University of Monastir, 5019 Monastir, Tunisia; Applied Epidemiology in Maternal and child Health Research Laboratory (LR 12 SP 17), 5100, Tahar Sfar University Hospital, 5100 Mahdia, Tunisia
| |
Collapse
|
4
|
Diagnosis of Chromosome 15q-Terminal Deletion Syndrome through Elevated Fasting Serum Growth Hormone Levels. ENDOCRINES 2022. [DOI: 10.3390/endocrines3010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Chromosome 15q26-qter deletion syndrome is a rare disease that causes prenatal and postnatal growth retardation, microcephaly, developmental delay, and congenital heart diseases, mainly due to haploinsufficiency of IGF1R. In addition, patients with pathogenic variants of the IGF1R show similar symptoms. We report the case of a 5-month-old girl with prenatal and postnatal growth retardation, microcephaly, and congenital heart disease. At 5 months of age, her length was 54.7 cm (−4.3 SD), her weight was 4.4 kg (−3.1 SD), and her head circumference was 37.4 cm (−2.8 SD), thus presenting severe growth retardation. Repeated pre-feeding serum GH levels were abnormally high (26.1–85.5 ng/mL), and IGF-1 levels (+0.16 to +1.2 SD) were relatively high. The 15q sub-telomere fluorescence in situ hybridization analysis revealed a heterozygous deletion in the 15q terminal region. Whole-genome single nucleotide polymorphism microarray analysis showed a terminal deletion of 6.4 Mb on 15q26.2q26.3. This is the first report showing that fasting GH levels are high in early infancy in patients with IGF1R abnormalities. In addition to relatively high IGF-1 levels, elevated fasting GH levels in early infancy may contribute to the diagnosis of IGF1R abnormalities.
Collapse
|
5
|
Kawashima-Sonoyama Y, Hotsubo T, Hamajima T, Hamajima N, Fujimoto M, Namba N, Kanzaki S. Various phenotypes of short stature with heterozygous IGF-1 receptor ( IGF1R) mutations. Clin Pediatr Endocrinol 2022; 31:59-67. [PMID: 35431446 PMCID: PMC8981046 DOI: 10.1297/cpe.2021-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/17/2022] [Indexed: 11/04/2022] Open
Affiliation(s)
- Yuki Kawashima-Sonoyama
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| | | | - Takashi Hamajima
- Department of Pediatric Endocrinology and Metabolism, Aichi Children’s Health and Medical Center, Obu, Japan
| | - Naoki Hamajima
- Department of Pediatrics, Nagoya City West Medical Center, Nagoya, Japan
| | - Masanobu Fujimoto
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Noriyuki Namba
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Susumu Kanzaki
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| |
Collapse
|
6
|
Hwa V, Fujimoto M, Zhu G, Gao W, Foley C, Kumbaji M, Rosenfeld RG. Genetic causes of growth hormone insensitivity beyond GHR. Rev Endocr Metab Disord 2021; 22:43-58. [PMID: 33029712 PMCID: PMC7979432 DOI: 10.1007/s11154-020-09603-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
Growth hormone insensitivity (GHI) syndrome, first described in 1966, is classically associated with monogenic defects in the GH receptor (GHR) gene which result in severe post-natal growth failure as consequences of insulin-like growth factor I (IGF-I) deficiency. Over the years, recognition of other monogenic defects downstream of GHR has greatly expanded understanding of primary causes of GHI and growth retardation, with either IGF-I deficiency or IGF-I insensitivity as clinical outcomes. Mutations in IGF1 and signaling component STAT5B disrupt IGF-I production, while defects in IGFALS and PAPPA2, disrupt transport and release of circulating IGF-I, respectively, affecting bioavailability of the growth-promoting IGF-I. Defects in IGF1R, cognate cell-surface receptor for IGF-I, disrupt not only IGF-I actions, but actions of the related IGF-II peptides. The importance of IGF-II for normal developmental growth is emphasized with recent identification of defects in the maternally imprinted IGF2 gene. Current application of next-generation genomic sequencing has expedited the pace of identifying new molecular defects in known genes or in new genes, thereby expanding the spectrum of GH and IGF insensitivity. This review discusses insights gained and future directions from patient-based molecular and functional studies.
Collapse
Affiliation(s)
- Vivian Hwa
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| | - Masanobu Fujimoto
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Pediatrics and Perinatology, Faculty of Medicine, Tottori University, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Gaohui Zhu
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, Chongqing, 40014, China
| | - Wen Gao
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Corinne Foley
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Meenasri Kumbaji
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Ron G Rosenfeld
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
7
|
Gonc EN, Ozon ZA, Oguz S, Kabacam S, Taskiran EZ, Kiper POS, Utine GE, Alikasifoglu A, Kandemir N, Boduroglu OK, Alikasifoglu M. Genetic IGF1R defects: new cases expand the spectrum of clinical features. J Endocrinol Invest 2020; 43:1739-1748. [PMID: 32356191 DOI: 10.1007/s40618-020-01264-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE We aimed to identify the phenotypic variability of IGF1R defects in a cohort of short children with normal GH secretion gathered through the last decade. PATIENTS AND METHODS Fifty children (25 girls) with short stature and a basal/stimulated growth hormone (GH) over 10 ng/ml having either a low birth weight or microcephaly were enrolled. MLPA and then Sanger sequence analysis were performed to detect IGF1R defects. The auxological and metabolic evaluation were carried out in index cases and their first degree family members whenever available. RESULTS A total of seven (14%) IGF1R defects were detected. Two IGF1R deletions and five heterozygous variants (one frameshift, four missense) were identified. Three (likely) pathogenic, one VUS and one likely benign were classified by using ACMG. All children with IGF1R defects had a height < - 2.5SDS, birth weight < - 1.4SDS, and head circumference < - 1.36SDS. IGF-1 ranged from - 2.44 to 2.13 SDS. One child with a 15q terminal deletion had a normal phenotype and intelligence, whereas low IQ is a finding in a case with missense variant. Two parents who carried IGF1R mutations had diabetes mellitus, hypertension and hyperlipidemia, one of whom also had hypergonadotropic hypogonadism. CONCLUSION We found a deletion or variant in IGF1R in 14% of short children. Birth weight, head circumference, intelligence, dysmorphic features, IGF-1 levels and even height are not consistent among patients. Additionally, metabolic and gonadal complications may appear during adulthood, suggesting that patients should be followed into adulthood to monitor for these late complications.
Collapse
Affiliation(s)
- E N Gonc
- Department of Pediatric Endocrinology, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey.
| | - Z A Ozon
- Department of Pediatric Endocrinology, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey
| | - S Oguz
- Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - S Kabacam
- Department of Pediatric Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - E Z Taskiran
- Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - P O S Kiper
- Department of Pediatric Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - G E Utine
- Department of Pediatric Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - A Alikasifoglu
- Department of Pediatric Endocrinology, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey
| | - N Kandemir
- Department of Pediatric Endocrinology, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey
| | - O K Boduroglu
- Department of Pediatric Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - M Alikasifoglu
- Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
8
|
Shapiro MR, Foster TP, Perry DJ, Rosenfeld RG, Dauber A, McNichols JA, Muir A, Hwa V, Brusko TM, Jacobsen LM. A Novel Mutation in Insulin-Like Growth Factor 1 Receptor (c.641-2A>G) Is Associated with Impaired Growth, Hypoglycemia, and Modified Immune Phenotypes. Horm Res Paediatr 2020; 93:322-334. [PMID: 33113547 PMCID: PMC7726096 DOI: 10.1159/000510764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/10/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Insulin-like growth factor 1 receptor (IGF1R) mutations lead to systemic disturbances in growth and glucose homeostasis due to widespread IGF1R expression throughout the body. IGF1R is expressed by innate and adaptive immune cells, facilitating their development and exerting immunomodulatory roles in the periphery. CASE PRESENTATION We report on a family presenting with a novel heterozygous IGF1R mutation with characterization of the mutation, IGF1R expression, and immune phenotyping. Twin probands presented clinically with short stature and hypoglycemia. Variable phenotypic expression was seen in 2 other family members carrying the IGF1R mutation. The probands were treated with exogenous growth hormone therapy and dietary cornstarch, improving linear growth and reducing hypoglycemic events. IGF1R c.641-2A>G caused abnormal mRNA splicing and premature protein termination. Flow cytometric immunophenotyping demonstrated lower IGF1R on peripheral blood mononuclear cells from IGF1R c.641-2A>G subjects. This alteration was associated with reduced levels of T-helper 17 cells and a higher percentage of T-helper 1 cells compared to controls, suggesting decreased IGF1R expression may affect CD4+ Th-cell lineage commitment. DISCUSSION Collectively, these data suggest a novel loss-of-function mutation (c.641-2A>G) leads to aberrant mRNA splicing and IGF1R expression resulting in hypoglycemia, growth restriction, and altered immune phenotypes.
Collapse
Affiliation(s)
- Melanie R Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Timothy P Foster
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Daniel J Perry
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Ron G Rosenfeld
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, USA
| | - Andrew Dauber
- Division of Endocrinology, Children's National Hospital, Washington, District of Columbia, USA
| | - James A McNichols
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Andrew Muir
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Vivian Hwa
- Division of Endocrinology, Department of Pediatrics, Cincinnati Center for Growth Disorders, Cincinnati Children's Hospital Medical Center, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Laura M Jacobsen
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA,
| |
Collapse
|
9
|
Göpel E, Rockstroh D, Pfäffle H, Schlicke M, Pozza SBD, Gannagé-Yared MH, Gucev Z, Mohn A, Harmel EM, Volkmann J, Weihrauch-Blüher S, Gausche R, Bogatsch H, Beger C, Klammt J, Pfäffle R. A Comprehensive Cohort Analysis Comparing Growth and GH Therapy Response in IGF1R Mutation Carriers and SGA Children. J Clin Endocrinol Metab 2020; 105:5611332. [PMID: 31680140 DOI: 10.1210/clinem/dgz165] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/03/2019] [Indexed: 01/21/2023]
Abstract
CONTEXT IGF1 receptor mutations (IGF1RM) are rare; however, patients exhibit pronounced growth retardation without catch-up. Although several case reports exist, a comprehensive statistical analysis investigating growth profile and benefit of recombinant human growth hormone (rhGH) treatment is still missing. OBJECTIVE AND METHODS Here, we compared IGF1RM carriers (n = 23) retrospectively regarding birth parameters, growth response to rhGH therapy, near final height, and glucose/insulin homeostasis to treated children born small for gestational age (SGA) (n = 34). Additionally, health profiles of adult IGF1RM carriers were surveyed by a questionnaire. RESULTS IGF1RM carriers were significantly smaller at rhGH initiation and had a diminished first-year response compared to SGA children (Δ height standard deviation score: 0.29 vs. 0.65), resulting in a lower growth response under therapy. Interestingly, the number of poor therapy responders was three times higher for IGF1RM carriers than for SGA patients (53 % vs. 17 %). However, most IGF1RM good responders showed catch-up growth to the levels of SGA patients. Moreover, we observed no differences in homeostasis model assessment of insulin resistance before treatment, but during treatment insulin resistance was significantly increased in IGF1RM carriers compared to SGA children. Analyses in adult mutation carriers indicated no increased occurrence of comorbidities later in life compared to SGA controls. CONCLUSION In summary, IGF1RM carriers showed a more pronounced growth retardation and lower response to rhGH therapy compared to non-mutation carriers, with high individual variability. Therefore, a critical reevaluation of success should be performed periodically. In adulthood, we could not observe a significant influence of IGF1RM on metabolism and health of carriers.
Collapse
Affiliation(s)
- Eric Göpel
- Integrated Research and Treatment Center (IFB) Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| | - Denise Rockstroh
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| | - Heike Pfäffle
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| | - Marina Schlicke
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| | | | | | - Zoran Gucev
- University Clinic of Child Diseases, Faculty of Medicine, Ss. Cyril and Methodius University of Skopje, Skopje, Republic of North Macedonia
| | - Angelika Mohn
- Department of Pediatrics Center of Excellence on Aging, "G. D'Annunzio" University Foundation, Chieti, Italy
| | - Eva-Maria Harmel
- Medical Center for Internal Medicine, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Julia Volkmann
- Pediatric Cardiology, Leipzig Heart Center, Leipzig, Germany
| | - Susann Weihrauch-Blüher
- Integrated Research and Treatment Center (IFB) Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Ruth Gausche
- Growth Network CrescNet, University of Leipzig, Leipzig, Germany
| | | | - Christoph Beger
- Growth Network CrescNet, University of Leipzig, Leipzig, Germany
| | - Jürgen Klammt
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
- MVZ Labor Dr. Reising-Ackermann und Kollegen GbR, Leipzig, Germany
| | - Roland Pfäffle
- Center for Pediatric Research Leipzig, University Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| |
Collapse
|
10
|
Schwiebert C, Kühnen P, Becker NP, Welsink T, Keller T, Minich WB, Wiegand S, Schomburg L. Antagonistic Autoantibodies to Insulin-Like Growth Factor-1 Receptor Associate with Poor Physical Strength. Int J Mol Sci 2020; 21:ijms21020463. [PMID: 31940750 PMCID: PMC7013472 DOI: 10.3390/ijms21020463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 01/04/2023] Open
Abstract
Natural autoantibodies to the IGF1 receptor (IGF1R-aAb) have been described in relation to Graves' ophthalmopathy. Other physiological roles of natural IGF1R-aAb are not known. We hypothesized that IGF1R-aAb may be related to muscle development. Serum samples (n = 408) from young overweight subjects (n = 143) were collected during a lifestyle intervention study. Anthropometric parameters, along with leptin, IGF1 and IGF1R-aAb concentrations, were analyzed, and the subjects were categorized into positive or negative for IGF1R-aAb. Eleven out of 143 subjects (7.7%) were positive for IGF1R-aAb. Identified IGF1R-aAb were molecularly characterized and showed antagonistic activity in vitro impairing IGF1-mediated IGF1R activation. Mean body weight, height or age were similar between IGF1R-aAb-positive and -negative subjects, but IGF1 concentrations differed. Jumping ability, as well as right and left handgrip strengths, were lower in the IGF1R-aAb-positive as compared to the IGF1R-aAb-negative subjects. We conclude that natural IGF1R-aAb are detectable in apparently healthy subjects and are capable of antagonizing IGF1-dependent IGF1R activation. Moreover, the presence of IGF1R-aAb is associated with poor physical strength. Although the causality of this association is unclear, the data imply a potential influence of IGF1R autoimmunity on muscle development.
Collapse
Affiliation(s)
- Christian Schwiebert
- Institute for Experimental Endocrinology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; (C.S.); (N.-P.B.); (T.W.); (W.B.M.)
| | - Peter Kühnen
- Department of Paediatric Endocrinology and Diabetology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; (P.K.); (S.W.)
| | - Niels-Peter Becker
- Institute for Experimental Endocrinology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; (C.S.); (N.-P.B.); (T.W.); (W.B.M.)
| | - Tim Welsink
- Institute for Experimental Endocrinology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; (C.S.); (N.-P.B.); (T.W.); (W.B.M.)
| | - Theresa Keller
- Institute of Biometry and Clinical Epidemiology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany;
| | - Waldemar B. Minich
- Institute for Experimental Endocrinology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; (C.S.); (N.-P.B.); (T.W.); (W.B.M.)
| | - Susanna Wiegand
- Department of Paediatric Endocrinology and Diabetology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; (P.K.); (S.W.)
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; (C.S.); (N.-P.B.); (T.W.); (W.B.M.)
- Correspondence: ; Tel.: +49-30-450-524289
| |
Collapse
|
11
|
Giabicani E, Willems M, Steunou V, Chantot-Bastaraud S, Thibaud N, Abi Habib W, Azzi S, Lam B, Bérard L, Bony-Trifunovic H, Brachet C, Brischoux-Boucher E, Caldagues E, Coutant R, Cuvelier ML, Gelwane G, Guemas I, Houang M, Isidor B, Jeandel C, Lespinasse J, Naud-Saudreau C, Jesuran-Perelroizen M, Perrin L, Piard J, Sechter C, Souchon PF, Storey C, Thomas D, Le Bouc Y, Rossignol S, Netchine I, Brioude F. Increasing knowledge in IGF1R defects: lessons from 35 new patients. J Med Genet 2019; 57:160-168. [PMID: 31586944 DOI: 10.1136/jmedgenet-2019-106328] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/06/2019] [Accepted: 08/24/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The type 1 insulin-like growth factor receptor (IGF1R) is a keystone of fetal growth regulation by mediating the effects of IGF-I and IGF-II. Recently, a cohort of patients carrying an IGF1R defect was described, from which a clinical score was established for diagnosis. We assessed this score in a large cohort of patients with identified IGF1R defects, as no external validation was available. Furthermore, we aimed to develop a functional test to allow the classification of variants of unknown significance (VUS) in vitro. METHODS DNA was tested for either deletions or single nucleotide variant (SNV) and the phosphorylation of downstream pathways studied after stimulation with IGF-I by western blot analysis of fibroblast of nine patients. RESULTS We detected 21 IGF1R defects in 35 patients, including 8 deletions and 10 heterozygous, 1 homozygous and 1 compound-heterozygous SNVs. The main clinical characteristics of these patients were being born small for gestational age (90.9%), short stature (88.2%) and microcephaly (74.1%). Feeding difficulties and varying degrees of developmental delay were highly prevalent (54.5%). There were no differences in phenotypes between patients with deletions and SNVs of IGF1R. Functional studies showed that the SNVs tested were associated with decreased AKT phosphorylation. CONCLUSION We report eight new pathogenic variants of IGF1R and an original case with a homozygous SNV. We found the recently proposed clinical score to be accurate for the diagnosis of IGF1R defects with a sensitivity of 95.2%. We developed an efficient functional test to assess the pathogenicity of SNVs, which is useful, especially for VUS.
Collapse
Affiliation(s)
- Eloïse Giabicani
- Sorbonne Université, UFR Médecine, Paris, France .,AP-HP, Hôpital Armand Trousseau-Explorations Fonctionnelles Endocriniennes, Paris, France.,INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | | | | | | | - Nathalie Thibaud
- AP-HP, Hôpital Armand Trousseau-Explorations Fonctionnelles Endocriniennes, Paris, France
| | | | - Salah Azzi
- INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | - Bich Lam
- INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | - Laurence Bérard
- AP-HP, Hôpital Armand Trousseau-Explorations Fonctionnelles Endocriniennes, Paris, France
| | | | - Cécile Brachet
- U.L.B., Pediatric Endocrinology, Reine Fabiola Children's Hospital, Brussels, Belgium
| | | | | | - Regis Coutant
- CHU Angers, Endocrinologie et Diabétologie Pédiatriques, Angers, France
| | | | - Georges Gelwane
- AP-HP, Hôpital Robert Debré, Endocrinologie et Diabétologie Pédiatriques, Paris, France.,Université Paris Diderot, Hôpital Robert Debré, Paris, France
| | | | - Muriel Houang
- Sorbonne Université, UFR Médecine, Paris, France.,AP-HP, Hôpital Armand Trousseau-Explorations Fonctionnelles Endocriniennes, Paris, France.,INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | | | - Claire Jeandel
- CHRU Montpellier Pôle Mère et enfant, Pédiatrie Spécialisée Endocrinologie Gynécologie de l'Enfant et de l'Adolescent, Montpellier, France
| | | | | | - Monique Jesuran-Perelroizen
- Endocrinologie-pédiatrique, Cabinet libéral, Toulouse, France.,AFPEL, Association Française des Pédiatres Endocrinologues Libéraux, Lille, France
| | - Laurence Perrin
- Université Paris Diderot, Hôpital Robert Debré, Paris, France.,AP-HP, Hôpital Robert Debré, Génétique Clinique, Paris, France
| | - Juliette Piard
- Université de Franche-Comté, CHU Besançon, Centre de Génétique Humaine, Besançon, France
| | - Claire Sechter
- Université de Franche-Comté, CHU Jean Minjoz, Unité d'Endocrinologie et Diabétologie Pédiatriques, Besançon, France
| | - Pierre-François Souchon
- American Memorial Hospital, Diabétologie et Endocrinologie Pédiatriques, CHU Reims, Reims, France
| | - Caroline Storey
- AP-HP, Hôpital Robert Debré, Endocrinologie et Diabétologie Pédiatriques, Paris, France.,Université Paris Diderot, Hôpital Robert Debré, Paris, France
| | | | - Yves Le Bouc
- Sorbonne Université, UFR Médecine, Paris, France.,INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | - Sylvie Rossignol
- Hopitaux universitaires de Strasbourg, Service de Pédiatrie 1, Strasbourg, France.,INSERM U1112, Institut de Génétique Médicale d'Alsace, Laboratoire de Génétique Médicale, Strasbourg, France
| | - Irène Netchine
- Sorbonne Université, UFR Médecine, Paris, France.,AP-HP, Hôpital Armand Trousseau-Explorations Fonctionnelles Endocriniennes, Paris, France.,INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | - Frédéric Brioude
- Sorbonne Université, UFR Médecine, Paris, France.,AP-HP, Hôpital Armand Trousseau-Explorations Fonctionnelles Endocriniennes, Paris, France.,INSERM, Centre de Recherche Saint-Antoine, Paris, France
| |
Collapse
|
12
|
Ocaranza P, Losekoot M, Walenkamp MJE, De Bruin C, Wit JM, Mericq V. Intrauterine Twin Discordancy and Partial Postnatal Catch-up Growth in a Girl with a Pathogenic IGF1R Mutation. J Clin Res Pediatr Endocrinol 2019; 11:293-300. [PMID: 30859796 PMCID: PMC6745462 DOI: 10.4274/jcrpe.galenos.2019.2018.0236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Insulin like growth factors-1 (IGF-1) is essential for normal in utero and postnatal human growth. It mediates its effects through the IGF-1 receptor (IGF1R), a widely expressed cell surface tyrosine kinase receptor. The aim of the study was to analyze pre- and post-natal growth, clinical features and laboratory findings in a small for gestational age (SGA) girl in whom discordant postnatal growth persisted and her appropriate for gestational age (AGA) brother. METHODS A girl born with a low weight and length [-2.3 and -2.4 standard deviation (SD) score (SDS), respectively] but borderline low head circumference (-1.6 SD) presented with a height of -1.7 SDS, in contrast to a normal height twin brother (0.0 SDS). IGF-1 resistance was suspected because of elevated serum IGF-1 levels. RESULTS Sequencing revealed the presence of a previously described pathogenic heterozygous mutation (p.Glu1050Lys) in the SGA girl which was not present in the parents nor in the AGA twin brother. CONCLUSION The pathogenic IGF1R mutation in this girl led to intrauterine growth retardation followed by partial postnatal catch-up growth. Height in mid-childhood was in the lower half of the reference range, but still 1.7 SD shorter than her twin brother.
Collapse
Affiliation(s)
- Paula Ocaranza
- University of Chile Faculty of Medicine, Institute of Maternal and Child Research, Santiago, Chile
| | - Monique Losekoot
- Leiden University Medical Center, Department of Clinical Genetics, Leiden, The Netherlands
| | - Marie J. E. Walenkamp
- Emma Children’s Hospital, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Department of Pediatric Endocrinology, Amsterdam, The Netherlands
| | - Christiaan De Bruin
- Leiden University Medical Center, Department of Pediatrics, Leiden, The Netherlands
| | - Jan M. Wit
- Leiden University Medical Center, Department of Pediatrics, Leiden, The Netherlands
| | - Veronica Mericq
- University of Chile Faculty of Medicine, Institute of Maternal and Child Research, Santiago, Chile,* Address for Correspondence: University of Chile Faculty of Medicine, Institute of Maternal and Child Research, Santiago, Chile E-mail:
| |
Collapse
|
13
|
Walenkamp MJE, Robers JML, Wit JM, Zandwijken GRJ, van Duyvenvoorde HA, Oostdijk W, Hokken-Koelega ACS, Kant SG, Losekoot M. Phenotypic Features and Response to GH Treatment of Patients With a Molecular Defect of the IGF-1 Receptor. J Clin Endocrinol Metab 2019; 104:3157-3171. [PMID: 30848790 DOI: 10.1210/jc.2018-02065] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
CONTEXT The phenotype and response to GH treatment of children with an IGF1R defect is insufficiently known. OBJECTIVE To develop a clinical score for selecting children with short stature for genetic testing and evaluate the efficacy of treatment. DESIGN AND SETTING Case series with an IGF1R defect identified in a university genetic laboratory. PATIENTS AND INTERVENTIONS Of all patients with sufficient clinical data, 18 had (likely) pathogenic mutations (group 1) and 7 had 15q deletions including IGF1R (group 2); 19 patients were treated with GH. MAIN OUTCOME MEASURES Phenotype and response to GH treatment. RESULTS In groups 1 and 2, mean (range) birth weight, length, and head circumference (HC) SD scores (SDSs) were -2.1 (-3.7 to -0.4), -2.7 (-5.0 to -1.0), and -1.6 (-3.0 to 0.0), respectively. At presentation, height, HC, and serum IGF-1 SDSs were -3.0 (-5.5 to -1.7), -2.5 (-4.2 to -0.5), and +1.2 (-1.3 to 3.2), respectively. Feeding problems were reported in 15 of 19 patients. A clinical score with 76% sensitivity is proposed. After 3 years of GH treatment [1.1 (0.2) mg/m2/d] height gain in groups 1 (n = 12) and 2 (n = 7) was 0.9 SDS and 1.3 SDS (at a mean IGF-1 of 3.5 SDS), less than reported for small for gestational age (1.8 SDS). CONCLUSION A clinical score encompassing birth weight and/or length, short stature, microcephaly, and IGF-1 is useful for selecting patients for IGF1R analysis. Feeding problems are common and the growth response to GH treatment is moderate.
Collapse
Affiliation(s)
- Marie J E Walenkamp
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jasmijn M L Robers
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Jan M Wit
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Wilma Oostdijk
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Anita C S Hokken-Koelega
- Dutch Growth Research Foundation, Rotterdam, Netherlands
- Department of Pediatrics, Subdivision of Endocrinology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sarina G Kant
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Monique Losekoot
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
14
|
Bodle EE, Gupta R, Cherry AM, Muffly L, Manning MA. Acute leukemia in a patient with 15q overgrowth syndrome. Am J Med Genet A 2019; 179:1025-1029. [PMID: 30861314 DOI: 10.1002/ajmg.a.61115] [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: 06/28/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 11/10/2022]
Abstract
Overgrowth syndromes are rare genetic conditions which present as global or segmental hyperplasia and are sometimes associated with increased risk of malignancy. Trisomy of the terminal portion of 15q which includes the IGFR1 gene, produces a rare overgrowth phenotype that has been termed 15q overgrowth syndrome (15q OGS). Upregulation of IGF1R has long been implicated in oncogenesis of multiple cancer types, including acute leukemias, and has been shown to render cells more susceptible to other transforming events. To date, too few cases of 15q OGS have been reported to identify any cancer predisposition. We present a case of a 34-year-old female with intellectual disability, macrocephaly, and subtle dysmorphic features who was diagnosed with mixed phenotype acute leukemia (lymphoid and myeloid). Prior to initiation of therapy she was referred to medical genetics for further evaluation and was identified as having a chromosomal translocation resulting in a partial trisomy of chromosome 15q, consistent with 15q OGS. A review of the literature for cases of malignancy in individuals with increased copy number of 15q revealed only one other reported patient. Given the small number of reported individuals, we cannot rule out an increased risk of cancer associated with this chromosomal overgrowth syndrome. Although concerns have been raised regarding treatment feasibility in the setting of chromosomal disorders, the reported patient underwent successful treatment with allogeneic hematopoietic stem-cell transplant.
Collapse
Affiliation(s)
- Ethan E Bodle
- Division of Medical Genetics, Department of Pediatrics, Stanford University, Stanford, California
| | - Ridhi Gupta
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | - Athena M Cherry
- Department of Pathology, Stanford University, Stanford, California
| | - Lori Muffly
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University, Stanford, California
| | - Melanie A Manning
- Division of Medical Genetics, Department of Pediatrics, Stanford University, Stanford, California.,Department of Pathology, Stanford University, Stanford, California
| |
Collapse
|
15
|
Serum concentrations of IGF-I/IGF-II as biomarkers of alcohol damage during foetal development and diagnostic markers of Foetal Alcohol Syndrome. Sci Rep 2019; 9:1562. [PMID: 30733584 PMCID: PMC6367511 DOI: 10.1038/s41598-018-38041-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/30/2018] [Indexed: 12/23/2022] Open
Abstract
Foetal Alcohol Syndrome (FAS) is the most deleterious health effect derived from alcohol consumption during pregnancy and is placed at the end of the Foetal Alcohol Spectrum Disorders (FASD). Few studies have proposed potential molecular biomarkers of physical and neurological damage associated with prenatal alcohol exposure. We prospectively recruited 55 children from 8 to 12 years old, with a prenatal assessment for ethanol exposure using meconium analysis of fatty acid ethyl esters (FAEE). The control group was established for FAEE < 2 nmol/g (n = 31) and a Prenatal Ethanol Exposure (PEE) group for FAEEs > 2 nmol/g (n = 33). Moreover, 98 children adopted from Eastern European Countries (EEC) were also recruited to evaluate FASD diagnosis comprising 31 cases with complete FAS, 42 with partial FAS, 6 with ARBD and 5 with ARND. Serum values of IGF-I and IGF-II for all children recruited were determined by immunoassay. Anthropometric and neurocognitive evaluation showed severe impairments in FAS children, moderate effects in PEE and no harmful effects in the control group with no prenatal exposure to alcohol. Analysis of IGF-I and IGF-II serum concentrations revealed that FASD from EEC as well as PEE children showed significantly lower concentrations of both IGF-I and IFG-II than the control group and reference values. Moreover, Spearman correlations showed a significant effect of IGF-I on anthropometric measurements in girls, whereas IGF-II affected the neuropsychological variables in both genders. These findings validate the use of growth factors IGF-I and IGF-II as surrogate biomarkers of damage induced by prenatal exposure to ethanol and could be used in the diagnosis of foetal alcohol spectrum disorders.
Collapse
|
16
|
Cabrera-Salcedo C, Hawkes CP, Tyzinski L, Andrew M, Labilloy G, Campos D, Feld A, Deodati A, Hwa V, Hirschhorn JN, Grimberg A, Dauber A. Targeted Searches of the Electronic Health Record and Genomics Identify an Etiology in Three Patients with Short Stature and High IGF-I Levels. Horm Res Paediatr 2019; 92:186-195. [PMID: 31865343 PMCID: PMC7173346 DOI: 10.1159/000504884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Short stature is one of the most common reasons for referral to a pediatric endocrinologist and can result from many etiologies. However, many patients with short stature do not receive a definitive diagnosis. OBJECTIVE To ascertain whether integrating targeted bioinformatics searches of electronic health records (EHRs) combined with genomic studies could identify patients with previously undiagnosed rare genetic etiologies of short stature. We focused on a specific rare phenotypic subgroup: patients with short stature and elevated IGF-I levels. METHODS We performed a cross-sectional cohort study at three large academic pediatric healthcare networks. Eligible subjects included children with heights below -2 SD, IGF-I levels >90th percentile, and no known etiology for short stature. We performed a search of the EHRs to identify eligible patients. Patients were then recruited for phenotyping followed by exome sequencing and in vitro assays of IGF1R function. RESULTS A total of 234 patients were identified by the bioinformatics algorithm with 39 deemed eligible after manual review (17%). Of those, 9 were successfully recruited. A genetic etiology was identified in 3 of the 9 patients including 2 novel variants in IGF1R and a de novo variant in CHD2. In vitro studies supported the pathogenicity of the IGF1R variants. CONCLUSIONS This study provides proof of principle that patients with rare phenotypic subgroups can be identified based on discrete data elements in the EHRs. Although limitations exist to fully automating this approach, these searches may help find patients with previously unidentified rare genetic disorders.
Collapse
Affiliation(s)
- Catalina Cabrera-Salcedo
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, Division of Endocrinology, University of Louisville, Louisville, Kentucky, USA
| | - Colin P. Hawkes
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leah Tyzinski
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Melissa Andrew
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Division of Endocrinology and Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Guillaume Labilloy
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Diego Campos
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Amalia Feld
- Division of Endocrinology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Annalisa Deodati
- Division of Endocrinology and Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA,Dipartimento Pediatrico Universitario Ospedaliero “Bambino Gesù” Children’s Hospital-Tor Vergata University, Rome, Italy
| | | | - Vivian Hwa
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Joel N. Hirschhorn
- Division of Endocrinology, Boston Children’s Hospital, Boston, Massachusetts, USA,Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Adda Grimberg
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrew Dauber
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA, .,Division of Endocrinology and Center for Genetic Medicine Research, Children's National Hospital, Washington, District of Columbia, USA, .,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA,
| |
Collapse
|
17
|
Finken MJJ, van der Steen M, Smeets CCJ, Walenkamp MJE, de Bruin C, Hokken-Koelega ACS, Wit JM. Children Born Small for Gestational Age: Differential Diagnosis, Molecular Genetic Evaluation, and Implications. Endocr Rev 2018; 39:851-894. [PMID: 29982551 DOI: 10.1210/er.2018-00083] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/21/2018] [Indexed: 12/25/2022]
Abstract
Children born small for gestational age (SGA), defined as a birth weight and/or length below -2 SD score (SDS), comprise a heterogeneous group. The causes of SGA are multifactorial and include maternal lifestyle and obstetric factors, placental dysfunction, and numerous fetal (epi)genetic abnormalities. Short-term consequences of SGA include increased risks of hypothermia, polycythemia, and hypoglycemia. Although most SGA infants show catch-up growth by 2 years of age, ∼10% remain short. Short children born SGA are amenable to GH treatment, which increases their adult height by on average 1.25 SD. Add-on treatment with a gonadotropin-releasing hormone agonist may be considered in early pubertal children with an expected adult height below -2.5 SDS. A small birth size increases the risk of later neurodevelopmental problems and cardiometabolic diseases. GH treatment does not pose an additional risk.
Collapse
Affiliation(s)
- Martijn J J Finken
- Department of Pediatrics, VU University Medical Center, MB Amsterdam, Netherlands
| | - Manouk van der Steen
- Department of Pediatrics, Erasmus University Medical Center/Sophia Children's Hospital, CN Rotterdam, Netherlands
| | - Carolina C J Smeets
- Department of Pediatrics, Erasmus University Medical Center/Sophia Children's Hospital, CN Rotterdam, Netherlands
| | - Marie J E Walenkamp
- Department of Pediatrics, VU University Medical Center, MB Amsterdam, Netherlands
| | - Christiaan de Bruin
- Department of Pediatrics, Leiden University Medical Center, RC Leiden, Netherlands
| | - Anita C S Hokken-Koelega
- Department of Pediatrics, Erasmus University Medical Center/Sophia Children's Hospital, CN Rotterdam, Netherlands
| | - Jan M Wit
- Department of Pediatrics, Leiden University Medical Center, RC Leiden, Netherlands
| |
Collapse
|
18
|
Yu M, Cui X, Wang H, Liu J, Qin H, Liu S, Yan Q. FUT8 drives the proliferation and invasion of trophoblastic cells via IGF-1/IGF-1R signaling pathway. Placenta 2018; 75:45-53. [PMID: 30712666 DOI: 10.1016/j.placenta.2018.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 11/07/2018] [Accepted: 11/15/2018] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Trophoblast proliferation and invasion are essential for embryo implantation and placentation. Protein glycosylation is one of the most common and vital post-translational modifications, regulates protein physical and biochemical properties. FUT8 is the only known fucosyltransferase responsible for catalyzing α1,6-fucosylation in mammals, and α1,6-fucosylated glycoproteins are found to participate in various physiopathological processes. However, whether FUT8/α1,6-fucosylation modulates the functions of trophoblastic cells remains elusive. METHODS FUT8 in human placenta villi during 6-8 gestational weeks and trophoblastic cells were detected by Western blot and immunofluorescent staining. α1,6-fucosylation in tissues or cells were measured by Lectin LCA (Lens culinaris) fluorescent staining and Lectin blot. FUT8 expression was down-regulated by siRNA transfection in JAR and JEG-3 cells, and cell viability, motility and invasiveness ability were detected by the functional experiments. α1,6-fucosylation of insulin-like growth factor receptor (IGF-1R) was examined by immunoprecipitation, and the amount of phosphorylated IGF-1R was detected in FUT8 down-regulated JAR cells. RESULTS Human placenta villi and trophoblastic cells expressed FUT8/α1,6-fucosylation. Knockdown FUT8 by siRNA transfection suppressed the proliferation, epithelial-mesenchymal transition, migration and invasion of JAR and JEG-3 cells. Furthermore, we found that FUT8 modified the α1,6-fucosylation of IGF-1R, and regulated IGF-1 dependent activation of IGF-1R, MAPK and PI3K/Akt signaling pathways in JAR cells. CONCLUSIONS Our results implicate a critical role for FUT8 in maintaining the normal functions of trophoblastic cells, suggesting manipulating FUT8 may be an effective approach in pregnancy.
Collapse
Affiliation(s)
- Ming Yu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People's Republic of China
| | - Xinyuan Cui
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People's Republic of China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People's Republic of China
| | - Jianwei Liu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People's Republic of China
| | - Huamin Qin
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Shuai Liu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People's Republic of China.
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People's Republic of China.
| |
Collapse
|
19
|
Janchevska A, Krstevska-Konstantinova M, Pfäffle H, Schlicke M, Laban N, Tasic V, Gucev Z, Mironska K, Dimovski A, Kratzsch J, Klammt J, Pfäffle R. IGF1R Gene Alterations in Children Born Small for Gestitional Age (SGA). Open Access Maced J Med Sci 2018; 6:2040-2044. [PMID: 30559857 PMCID: PMC6290431 DOI: 10.3889/oamjms.2018.416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND: Small for gestational age (SGA)-born children are a heterogeneous group with few genetic causes reported. Genetic alterations in the IGF1 receptor (IGF1R) are found in some SGA children. AIM: To investigate whether alterations in IGF1R gene are present in SGA born children. PATIENTS AND METHODS: We analysed 64 children born SGA who stayed short (mean -3.25 ± 0.9 SDS) within the first 4 years of age, and 36 SGA children who caught up growth (0.20 ± 1.1 SDS). PCR products of all coding IGF1R exons were screened by dHPLC followed by direct sequencing of conspicuous fragments to identify small nucleotide variants. The presence of IGF1R gene copy number alterations was determined by Multiplex Ligation-dependent Probe Amplification (MLPA). RESULTS: The cohort of short SGA born children revealed a heterozygous, synonymous variant c.3453C > T in one patient and a novel heterozygous 3 bp in-frame deletion (c.3234_3236delCAT) resulting in one amino acid deletion (p.Ile1078del) in another patient. The first patient had normal serum levels of IGF1. The second patient had unusually low IGF1 serum concentrations (-1.57 SD), which contrasts previously published data where IGF1 levels rarely are found below the age-adjusted mean. CONCLUSIONS: IGF1R gene alterations were present in 2 of 64 short SGA children. The patients did not have any dysmorphic features or developmental delay. It is remarkable that one of them had significantly decreased serum concentrations of IGF1. Growth response to GH treatment in one of the patients was favourable, while the second one discontinued the treatment, but with catch-up growth.
Collapse
Affiliation(s)
- Aleksandra Janchevska
- Medical Faculty, Ss. Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | | | | | | | - Nevenka Laban
- Medical Faculty, Ss. Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Velibor Tasic
- Medical Faculty, Ss. Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Zoran Gucev
- Medical Faculty, Ss. Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Kristina Mironska
- Medical Faculty, Ss. Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | | | | | | | | |
Collapse
|
20
|
West RC, Bouma GJ, Winger QA. Shifting perspectives from "oncogenic" to oncofetal proteins; how these factors drive placental development. Reprod Biol Endocrinol 2018; 16:101. [PMID: 30340501 PMCID: PMC6195737 DOI: 10.1186/s12958-018-0421-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/09/2018] [Indexed: 12/23/2022] Open
Abstract
Early human placental development strongly resembles carcinogenesis in otherwise healthy tissues. The progenitor cells of the placenta, the cytotrophoblast, rapidly proliferate to produce a sufficient number of cells to form an organ that will contribute to fetal development as early as the first trimester. The cytotrophoblast cells begin to differentiate, some towards the fused cells of the syncytiotrophoblast and some towards the highly invasive and migratory extravillous trophoblast. Invasion and migration of extravillous trophoblast cells mimics tumor metastasis. One key difference between cancer progression and placental development is the tight regulation of these oncogenes and oncogenic processes. Often, tumor suppressors and oncogenes work synergistically to regulate cell proliferation, differentiation, and invasion in a restrained manner compared to the uncontrollable growth in cancer. This review will compare and contrast the mechanisms that drive both cancer progression and placental development. Specifically, this review will focus on the molecular mechanisms that promote cell proliferation, evasion of apoptosis, cell invasion, and angiogenesis.
Collapse
Affiliation(s)
- Rachel C. West
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, 10290 Ridgegate Circle, Lone Tree, Fort Collins, CO 80124 USA
| | - Gerrit J. Bouma
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, 10290 Ridgegate Circle, Lone Tree, Fort Collins, CO 80124 USA
| | - Quinton A. Winger
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, 10290 Ridgegate Circle, Lone Tree, Fort Collins, CO 80124 USA
| |
Collapse
|
21
|
Janchevska A, Dimovski A, Mironska K, Tasic V, Gucev Z. IGF1R Gene Alterations in Small for Gestational Age (SGA) Children. Open Access Maced J Med Sci 2018; 6:790-793. [PMID: 29896311 PMCID: PMC5985885 DOI: 10.3889/oamjms.2018.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Small for gestational age children (SGA) is born on term with BW and or BL of -2.0 standard deviation score (SDS). SGA children have an increased risk of being short, developing DM, and cardiovascular and cerebrovascular disease. Often defects of IGF1R are the cause of SGA. Most frequently affected part of the IGF1R gene is the exon 2. AIM To investigate whether the exon 2 of the IGF1R gene is affected in the SGA children. PATIENTS AND METHODS A cohort of 100 SGA children born in term was evaluated for alterations in IG1R gene. Their anthropometric parameters, IGF1 serum concentrations and IGF1 SDS values were analysed. The molecular analysis of IGF1R gene was performed by PCR restriction-site analysis and followed by direct sequencing of conspicuous fragments. RESULTS Within our cohort, 64 SGA children were with short stature (height SDS -3.25 ± 0.90 SDS), and 36 were with normal height for their age and sex, (H SDS was 0.20 ± 1.1 SDS). None of these children had microcephaly (occipitofrontal circumference -0.70 ± 1.01 SDS vs 0.06 ± 0.56 SDS in SGA children with normal height) or dysmorphic features. The IGF1 serum concentrations and IGF1 SDS values of all children were within normal range. Only one child had lower normal serum IGF1 concentration. No alterations in exon 2 of IGF1R gene were detected. CONCLUSIONS The genetic analysis of the exon 2 of the IGF1R gene did not detect any gene defects in the analysed patients. The putative genetic defect in those children affects other parts of the IGF1R gene or another gene (s), or yet unidentified factors.
Collapse
Affiliation(s)
- Aleksandra Janchevska
- University Children’s Hospital, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | | | - Kristina Mironska
- University Children’s Hospital, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Velibor Tasic
- University Children’s Hospital, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Zoran Gucev
- University Children’s Hospital, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
22
|
Mainieri A, Haig D. Lost in translation: The 3'-UTR of IGF1R as an ancient long noncoding RNA. EVOLUTION MEDICINE AND PUBLIC HEALTH 2018; 2018:82-91. [PMID: 29644076 PMCID: PMC5887972 DOI: 10.1093/emph/eoy008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 02/21/2018] [Indexed: 12/20/2022]
Abstract
Background and objectives The insulin-like growth factor (IGF) signaling system is a major arena of intragenomic conflict over embryonic growth between imprinted genes of maternal and paternal origin and the IGF type 1 receptor (IGF1R) promotes proliferation of many human cancers. The 3'-untranslated region (3'-UTR) of the mouse Igf1r mRNA is targeted by miR-675-3p derived from the imprinted H19 long noncoding RNA. We undertook a comparative sequence analysis of vertebrate IGF1R 3'-UTRs to determine the evolutionary history of miR-675 target sequences and to identify conserved features that are likely to be involved in post-transcriptional regulation of IGF1R translation. Methodology Sequences of IGF1R 3'-UTRs were obtained from public databases and analyzed using publicly available algorithms. Results A very long 3'-UTR is a conserved feature of vertebrate IGF1R mRNAs. We found that some ancient microRNAs, such as let-7 and mir-182, have predicted binding sites that are conserved between cartilaginous fish and mammals. One very conserved region is targeted by multiple, maternally expressed imprinted microRNAs that appear to have evolved more recently than the targeted sequences. Conclusions and implications The conserved structures we identify in the IGF1R 3'-UTR are strong candidates for regulating cell proliferation during development and carcinogenesis. These conserved structures are now targeted by multiple imprinted microRNAs. These observations emphasize the central importance of IGF signaling pathways in the mediation of intragenomic conflicts over embryonic growth and identify possible targets for therapeutic interventions in cancer.
Collapse
Affiliation(s)
- Avantika Mainieri
- Department of Organismic and Evolutionary Biology, Harvard University, 26 Oxford Street, Cambridge, MA 02138, USA
| | - David Haig
- Department of Organismic and Evolutionary Biology, Harvard University, 26 Oxford Street, Cambridge, MA 02138, USA
| |
Collapse
|
23
|
Mahmoud R, Naidu A, Risheg H, Kimonis V. Response to Growth Hormone Treatment in a Patient with Insulin-Like Growth Factor 1 Receptor Deletion. J Clin Res Pediatr Endocrinol 2017; 9:380-386. [PMID: 28720553 PMCID: PMC5785648 DOI: 10.4274/jcrpe.4456] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
We report a six-year-old boy who presented with short stature, microcephaly, dysmorphic features, and developmental delay and who was identified with a terminal deletion of 15q26.2q26.3 containing the insulin-like growth factor receptor (IGF1R) gene in addition to a terminal duplication of the 4q35.1q35.2 region. We compare our case with other reports of deletions and mutations affecting the IGF1R gene associated with pre-and postnatal growth restriction. We report the dramatic response to growth hormone therapy in this patient which highlights the importance of identifying patients with IGF1R deletion and treating them early.
Collapse
Affiliation(s)
- Ranim Mahmoud
- Laboratory Corporation of America/Dynacare, Department of Cytogenetics, Seattle, Washington, USA
| | - Ajanta Naidu
- University of California, Department of Pediatrics, Division of Endocrinology, Irvine, California, USA
| | - Hiba Risheg
- Laboratory Corporation of America/Dynacare, Department of Cytogenetics, Seattle, Washington, USA
| | - Virginia Kimonis
- University of California, Department of Pediatrics, Division of Genetics and Genomic Medicine, Irvine, California, USA
,* Address for Correspondence: University of California, Irvine Medical Center, Department of Pediatrics, Division of Genetics and Genomic Medicine, Irvine, California, USA Phone: +714 456-5791 E-mail:
| |
Collapse
|
24
|
Solomon-Zemler R, Basel-Vanagaite L, Steier D, Yakar S, Mel E, Phillip M, Bazak L, Bercovich D, Werner H, de Vries L. A novel heterozygous IGF-1 receptor mutation associated with hypoglycemia. Endocr Connect 2017; 6. [PMID: 28649085 PMCID: PMC5551424 DOI: 10.1530/ec-17-0038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mutation in the insulin-like growth factor-1 receptor (IGF1R) gene is a rare cause for intrauterine and postnatal growth disorders. Patients identified with IGF1R mutations present with either normal or impaired glucose tolerance. None of the cases described so far showed hypoglycemia. We aimed to identify the genetic basis for small for gestational age, short stature and hypoglycemia over three generations in one family. The proband, a 9-year-old male, presented in infancy with recurrent hypoglycemic episodes, symmetric intrauterine growth retardation and postnatal growth retardation. Blood DNA samples from the patient, his parents, a maternal sister and maternal grandmother underwent Sanger sequencing of the IGF1R gene. Primary skin fibroblast cultures of the patient, his mother and age- and sex-matched control donors were used for gene expression and receptor functional analyses. We found a novel heterozygous mutation (c.94 + 1g > a, D1105E) affecting the splicing site of the IGF1R mRNA in the patient, his mother and his grandmother. Primary fibroblast cultures derived from the patient and his mother showed reduced proliferation and impaired activation of the IGF1R, evident by reduced IGF1R and AKT phosphorylation upon ligand binding. In conclusion, the newly identified heterozygous missense mutation in exon 1 of IGF1R (D1105E) results in impaired IGF1R function and is associated with small for gestational age, microcephaly and abnormal glucose metabolism. Further studies are required to understand the mechanisms by which this mutation leads to hypoglycemia.
Collapse
Affiliation(s)
- R Solomon-Zemler
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
| | - L Basel-Vanagaite
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetic InstituteRabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
- Felsenstein Medical Research CenterPetach Tikva, Israel
- Pediatric GeneticsSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - D Steier
- Day Hospitalization DepartmentSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - S Yakar
- David B. Kriser Dental CenterDepartment of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA
| | - E Mel
- Jesse Z. and Sara Lea Shafer Institute for Endocrinology and DiabetesSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - M Phillip
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Jesse Z. and Sara Lea Shafer Institute for Endocrinology and DiabetesSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - L Bazak
- Raphael Recanati Genetic InstituteRabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | | | - H Werner
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Shalom and VardaYoran Institute for Human Genome ResearchTel Aviv University, Tel Aviv, Israel
| | - L de Vries
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Jesse Z. and Sara Lea Shafer Institute for Endocrinology and DiabetesSchneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
25
|
Işık E, Haliloglu B, van Doorn J, Demirbilek H, Scheltinga SA, Losekoot M, Wit JM. Clinical and biochemical characteristics and bone mineral density of homozygous, compound heterozygous and heterozygous carriers of three novel IGFALS mutations. Eur J Endocrinol 2017; 176:657-667. [PMID: 28249955 DOI: 10.1530/eje-16-0999] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/13/2017] [Accepted: 03/01/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Acid-labile subunit (ALS) deficiency (ACLSD), caused by homozygous or compound heterozygous IGFALS mutations, is associated with moderate short stature, delayed puberty, low serum IGF-I and ALS and extremely low serum IGFBP-3. Its effect on birth weight, head circumference, bone mineral density (BMD), serum IGF-II and IGFBP-2 is uncertain, as well as the phenotype of heterozygous carriers of IGFALS mutations (partial ACLSD). DESIGN From all available members of five Turkish families, carrying three mutations in exon 2 of IGFALS (c.1462G > A, p.Asp488Asn (families A, B, E); c.251A > G, p.Asn84Ser (families C and E) and c.1477del, p.Arg493fs (family D)), clinical, laboratory and BMD data were collected. METHODS Auxological and biochemical findings were expressed as SDS for age and gender. Ternary complex formation in serum was investigated by size-exclusion chromatography. BMD using DXA bone densitometry was adjusted for height and age (Ha-BMD z-score). RESULTS In ACLSD (n = 24), mean ± s.d. height SDS (-2.7 ± 1.2), head circumference SDS (-2.3 ± 0.5) and body mass index (BMI) (-0.6 ± 1.0 SDS) were lower than those in partial ACLSD (n = 26, P ≤ 0.01) and birth weight SDS (n = 7) tended to be lower (-2.2 ± 1.1 vs -0.6 ± 0.3 in partial ACLSD (P = 0.07)). Serum IGF-I was -3.7 ± 1.4 vs -1.0 ± 1.0, IGF-II: -5.6 ± 0.7 vs -1.3 ± 0.7, ALS: <-4.4 ± 1.2 vs -2.1 ± 0.9 and IGFBP-3: -9.0 ± 1.9 vs -1.6 ± 0.8 SDS respectively (P < 0.001). Ha-BMD z-score was similar and normal in both groups. CONCLUSIONS To the known phenotype of ACLSD (i.e. short stature, reduced serum levels of IGF-I and ALS, extremely low serum IGFBP-3 and disturbed ternary complex formation), we add reduced birth weight, head circumference and serum IGF-II.
Collapse
Affiliation(s)
- Emregül Işık
- Department of Pediatric EndocrinologyGaziantep Children's Hospital, Gaziantep, Turkey
| | - Belma Haliloglu
- Department of Pediatric EndocrinologyYeditepe University School of Medicine, İstanbul, Turkey
| | - Jaap van Doorn
- Department of GeneticsUniversity Medical Center Utrecht, The Netherlands
| | - Hüseyin Demirbilek
- Department of Pediatric EndocrinologyHacettepe University Faculty of Medicine, Ankara, Turkey
| | | | | | - Jan M Wit
- Departments of PediatricsLeiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
26
|
Thakur S, Garg N, Zhang N, Hussey SE, Musi N, Adamo ML. IGF-1 receptor haploinsufficiency leads to age-dependent development of metabolic syndrome. Biochem Biophys Res Commun 2017; 486:937-944. [DOI: 10.1016/j.bbrc.2017.03.129] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 03/23/2017] [Indexed: 01/02/2023]
|
27
|
Pelosi P, Lapi E, Cavalli L, Verrotti A, Pantaleo M, de Martino M, Stagi S. Bone Status in a Patient with Insulin-Like Growth Factor-1 Receptor Deletion Syndrome: Bone Quality and Structure Evaluation Using Dual-Energy X-Ray Absorptiometry, Peripheral Quantitative Computed Tomography, and Quantitative Ultrasonography. Front Endocrinol (Lausanne) 2017; 8:227. [PMID: 28936199 PMCID: PMC5595156 DOI: 10.3389/fendo.2017.00227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 08/21/2017] [Indexed: 01/02/2023] Open
Abstract
Haploinsufficiency of the insulin-like growth factor (IGF)-1 receptor (IGF1R) gene is a rare, probably under-diagnosed, cause of short stature. However, the effects of IGF1R haploinsufficiency on glucose metabolism, bone status, and metabolism have rarely been investigated. We report the case of a patient referred to our center at the age of 18 months for short stature, failure to thrive, and Silver-Russell-like phenotype. Genetic analysis did not show hypomethylation of the 11p15.5 region or uniparental disomy of chromosome 7. Growth hormone (GH) stimulation tests revealed GH deficiency, whereas IGF-1 was 248 ng/mL. r-hGH treatment showed only a slight improvement (from -4.4 to -3.5 SDS). At 10 years of age, the child was re-evaluated: CGH-array identified a heterozygous de novo 4.92 Mb deletion in 15q26.2, including the IGF1R gene. Dual-energy X-ray absorptiometry showed a normal bone mineral density z-score, while peripheral quantitative computed tomography revealed reduced cortical and increased trabecular elements. A phalangeal bone quantitative ultrasonography showed significantly reduced amplitude-dependent speed of sound and bone transmission time values. The changes in bone architecture, quality, and metabolism in heterozygous IGF1R deletion patients, support the hypothesis that IGF-1 can be a key factor in bone modeling and accrual.
Collapse
Affiliation(s)
- Paola Pelosi
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Elisabetta Lapi
- Genetics and Molecular Medicine Unit, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Loredana Cavalli
- Department of Neuroscience, Neurorehabilitation Section, University of Pisa, Pisa, Italy
| | - Alberto Verrotti
- Department of Paediatrics, University of L’Aquila, L’Aquila, Italy
| | - Marilena Pantaleo
- Genetics and Molecular Medicine Unit, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Maurizio de Martino
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Stefano Stagi
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
- *Correspondence: Stefano Stagi,
| |
Collapse
|
28
|
Abstract
The retina is part of the central nervous system and both the retina as well as the brain can suffer from severe damage after very preterm birth. Retinopathy of prematurity is one of the major causes of blindness in these children and brain neuronal impairments including cognitive defects, cerebral palsy and intraventricular hemorrhage (IVH) are also complications of very preterm birth. Insulin-like growth factor 1 (IGF-1) acts to promote proliferation, maturation, growth and survival of neural cells. Low levels of circulating IGF-1 are associated with ROP and defects in the IGF-1 gene are associated with CNS disorders including learning deficits and brain growth restriction. Treatment of preterm infants with recombinant IGF-1 may potentially prevent ROP and CNS disorders. This review compares the role of IGF-1 in ROP and CNS disorders. A recent phase 2 study showed a positive effect of IGF-1 on the severity of IVH but no effect on ROP. A phase 3 trial is planned.
Collapse
Affiliation(s)
- Raffael Liegl
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chatarina Löfqvist
- Department of Ophthalmology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ann Hellström
- Department of Ophthalmology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Hellström A, Ley D, Hansen‐Pupp I, Hallberg B, Löfqvist C, Marter L, Weissenbruch M, Ramenghi LA, Beardsall K, Dunger D, Hård A, Smith LEH. Insulin-like growth factor 1 has multisystem effects on foetal and preterm infant development. Acta Paediatr 2016; 105:576-86. [PMID: 26833743 PMCID: PMC5069563 DOI: 10.1111/apa.13350] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/19/2015] [Accepted: 01/27/2016] [Indexed: 01/28/2023]
Abstract
Poor postnatal growth after preterm birth does not match the normal rapid growth in utero and is associated with preterm morbidities. Insulin‐like growth factor 1 (IGF‐1) axis is the major hormonal mediator of growth in utero, and levels of IGF‐1 are often very low after preterm birth. We reviewed the role of IGF‐1 in foetal development and the corresponding preterm perinatal period to highlight the potential clinical importance of IGF‐1 deficiency in preterm morbidities. Conclusion There is a rationale for clinical trials to evaluate the potential benefits of IGF‐1 replacement in very preterm infants.
Collapse
Affiliation(s)
- Ann Hellström
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - David Ley
- Department of Pediatrics Institute of Clinical Sciences Lund Skane University Hospital Lund University Lund Sweden
| | - Ingrid Hansen‐Pupp
- Department of Pediatrics Institute of Clinical Sciences Lund Skane University Hospital Lund University Lund Sweden
| | - Boubou Hallberg
- Department of Neonatology University Hospital Karolinska Institute Stockholm Sweden
| | - Chatarina Löfqvist
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Linda Marter
- Brigham and Women's Hospital Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Mirjam Weissenbruch
- Department of Neonatology VU University Medical Center Amsterdam The Netherlands
| | - Luca A. Ramenghi
- Genova Neonatal Intensive Care Unit Instituto Pediatrico Giannina Gaslini Genova Italy
| | | | - David Dunger
- Faculty of Academy of Medical Sciences Department of Paediatrics Institute of Metabolic Science University of Cambridge Cambridge UK
| | - Anna‐Lena Hård
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Lois E. H. Smith
- Department of Ophthalmology Boston Children's Hospital Harvard Medical School Boston MA USA
| |
Collapse
|
30
|
Fujimoto M, Kawashima Sonoyama Y, Hamajima N, Hamajima T, Kumura Y, Miyahara N, Nishimura R, Adachi K, Nanba E, Hanaki K, Kanzaki S. Heterozygous nonsense mutations near the C-terminal region of IGF1R in two patients with small-for-gestational-age-related short stature. Clin Endocrinol (Oxf) 2015; 83:834-41. [PMID: 25866162 DOI: 10.1111/cen.12791] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/14/2015] [Accepted: 04/04/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The type I insulin-like growth factor I receptor (IGF1R) plays an important role in growth. We aimed to evaluate the detailed mechanism underlying the effect of IGF1R on human growth. PATIENTS AND METHODS We have performed sequence analysis of IGF1R in 55 patients with SGA short stature in Japan, since 2004, and identified novel heterozygous nonsense mutations in 2 patients: an 8-year-old Japanese boy (case 1), with a birthweight of 2228 g (-3·3 SDS) and height of 46 cm (-2·1 SDS), and a 3-year-old Japanese girl (case 2), with a birthweight of 2110 g (-3·0 SDS) and height of 44·3 cm (-2·8 SDS). Both patients had a short stature (-3·2 SDS, -3·1 SDS). We determined the protein expression of mutated IGF1R, assessed the effect of the endoplasmic reticulum-associated degradation (ERAD) pathway on mutated IGF1R, assessed the dominant-negative effect of IGF1R and performed quantitative RT-PCR analysis of IGF1R mRNA expression in whole blood cells. RESULTS Two novel heterozygous nonsense mutations (case 1: p.Q1250X and case 2: p.W1249X) were identified. Although these mutations did not affect blood IGF1R mRNA levels, they significantly decreased the expression of IGF1R protein in transiently transfected cells. Treatment with the proteasome inhibitor MG132 showed significantly increased IGF1R protein. CONCLUSIONS Heterozygous nonsense mutations affecting the C-terminal region (p.Q1250X, p.W1249X) of IGF1R decreased the expression of IGF1R through the ERAD pathway. Our study revealed the importance of the C-terminal region and the dosage of this receptor for growth.
Collapse
Affiliation(s)
- Masanobu Fujimoto
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Yuki Kawashima Sonoyama
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Naoki Hamajima
- Department of Pediatrics, Nagoya City West Medical Center, Nagoya, Japan
| | - Takashi Hamajima
- Department of Pediatric Endocrinology and Metabolism, Aichi Children's Health and Medical Center, Obu, Japan
| | - Yumiko Kumura
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, Yonago, Japan
| | - Naoki Miyahara
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Rei Nishimura
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kaori Adachi
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, Yonago, Japan
| | - Eiji Nanba
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, Yonago, Japan
| | - Keiichi Hanaki
- Department of Women's & Children's Family Nursing, Tottori University Faculty of Medicine, Yonago, Japan
| | - Susumu Kanzaki
- Division of Pediatrics & Perinatology, Tottori University Faculty of Medicine, Yonago, Japan
| |
Collapse
|
31
|
Juanes M, Guercio G, Marino R, Berensztein E, Warman DM, Ciaccio M, Gil S, Bailez M, Rivarola MA, Belgorosky A. Three novel IGF1R mutations in microcephalic patients with prenatal and postnatal growth impairment. Clin Endocrinol (Oxf) 2015; 82:704-11. [PMID: 25040157 DOI: 10.1111/cen.12555] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/09/2014] [Accepted: 07/11/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND IGF1R gene mutations have been associated with varying degrees of intrauterine and postnatal growth retardation, and microcephaly. OBJECTIVE To identify and characterize IGF1R gene variations in a cohort of 28 Argentinean children suspected of having IGF-1 insensitivity, who were selected on the basis of the association of pre/postnatal growth failure and microcephaly. METHODS The coding sequence and flanking intronic regions of IGF1R gene were amplified and directly sequenced. Functional characterization was performed by two in vitro assays: 1) [Methyl-(3) H] thymidine incorporation into DNA in fibroblast cell primary cultures from patients and controls treated with IGF-1 for 16-24 h. 2) PI3K/Akt pathway was evaluated with phospho-Akt (Ser473) STAR ELISA Kit (Millipore) in fibroblast cultures from patients and controls stimulated with IGF-1 for 10 min. Prepubertal clinical and GH-IGF-1 axis evaluation was followed up. RESULTS We identified three novel heterozygous missense mutations in three unrelated patients, de novo p.Arg1256Ser, de novo p.Asn359Tyr and p.Tyr865Cys. In control cells, proliferation assay showed that IGF-1 significantly induced DNA synthesis at 20 h and Akt phosphorylation assay that it significantly stimulated phosphorylation after 10 min (P < 0·05 by anova and Bonferroni Tests). However, no significant increase was observed in any of the three patient fibroblasts in both functional studies. GH therapy growth response in two patients was inconsistent. CONCLUSION These variations led to failure of the IGF1R function causing pre- and postnatal growth retardation and microcephaly. Microcephaly should be considered in the evaluation of SGA patients, because it seems to favour the frequency of detection of IGF1R mutations.
Collapse
Affiliation(s)
- Matias Juanes
- Endocrinology Service, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Burkhardt S, Gesing J, Kapellen TM, Kovacs P, Kratzsch J, Schlicke M, Stobbe H, Tönjes A, Klammt J, Pfäffle R. Novel heterozygous IGF1R mutation in two brothers with developing impaired glucose tolerance. J Pediatr Endocrinol Metab 2015; 28:217-25. [PMID: 25153223 DOI: 10.1515/jpem-2014-0132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 06/27/2014] [Indexed: 11/15/2022]
Abstract
Infants born small for gestational age (SGA) are at risk to develop metabolic complications. Insulin-like growth factor 1 (IGF-1) resistance due to IGF-1 receptor (IGF1R) mutations is a rare genetic condition that causes proportionate growth retardation. The contribution of an impaired IGF1R function to the development of comorbidities such as disturbed glucose homeostasis is not well understood. Genetic analysis and detailed auxological, endocrine and psychological investigations in two male SGA siblings were performed. The two patients and their father bear a novel heterozygous mutation (p.Cys1248Tyr) in the IGF1R gene. Both brothers displayed very similar growth pattern before and during recombinant human growth hormone treatment, whereas oral glucose tolerance tests showed variable manifestations of progressive impaired glucose tolerance. The father had already developed type 2 diabetes mellitus. Growth retardation in our patients is likely caused by the IGF1R mutation that might predispose to disturbances of carbohydrate homeostasis. Therefore, a close metabolic monitoring of affected patients is indicated, particularly if growth hormone therapy is commenced.
Collapse
|
33
|
Forbes K, Shah VK, Siddals K, Gibson JM, Aplin JD, Westwood M. Statins inhibit insulin-like growth factor action in first trimester placenta by altering insulin-like growth factor 1 receptor glycosylation. Mol Hum Reprod 2014; 21:105-14. [PMID: 25304981 PMCID: PMC4275043 DOI: 10.1093/molehr/gau093] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The rapid rise in obesity, metabolic syndrome and type 2 diabetes is one of the major healthcare problems of the Western world. Affected individuals are often treated with statins (3-hydroxy-3-methylglutaryl co-enzyme A [HMG CoA] reductase inhibitors) to reduce circulating cholesterol levels and the risk of developing cardiovascular disease; given the evolving demographic profile of these conditions, such drugs are increasingly prescribed to women of reproductive age. We have previously shown that exposure of placental tissue to statins inhibits the action of insulin-like growth factors (IGF)-I and -II which are key regulators of trophoblast proliferation and placental development. N-linked glycans in the IGF receptor, IGF1R, influence its presentation at the cell surface. This study aimed to determine whether statins, which are known to affect N-glycosylation, modulate IGF1R function in placenta. Treatment of first trimester villous tissue explants with statins (pravastatin or cerivastatin) or inhibitors of N-glycosylation (tunicamycin, deoxymannojirimycin or castanospermine) altered receptor distribution in trophoblast and attenuated proliferation induced by IGF-I or IGF-II (Ki67; P < 0.05, n = 5). Decreased binding of Phaseolus vulgaris lectin and phytohaemagglutinin to IGF1R immunoprecipitated from treated explants demonstrated reduced levels of complex N-linked glycans. Co-incubation of tissue explants with statins and farnesyl pyrophosphate (which increases the supply of dolichol intermediates), prevented statin-mediated disruption of IGF1R localization and reversed the negative effect on IGF-mediated trophoblast proliferation. These data suggest that statins attenuate IGF actions in the placenta by inhibiting N-linked glycosylation and subsequent expression of mature IGF1R at the placental cell surface.
Collapse
Affiliation(s)
- Karen Forbes
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9WL, UK Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Vinit K Shah
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9WL, UK Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Kirk Siddals
- Centre for Imaging Sciences, Institute of Population Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PY, UK
| | - J Martin Gibson
- Centre for Imaging Sciences, Institute of Population Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9PY, UK
| | - John D Aplin
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9WL, UK Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Melissa Westwood
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9WL, UK Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| |
Collapse
|
34
|
Agrogiannis GD, Sifakis S, Patsouris ES, Konstantinidou AE. Insulin-like growth factors in embryonic and fetal growth and skeletal development (Review). Mol Med Rep 2014; 10:579-84. [PMID: 24859417 PMCID: PMC4094767 DOI: 10.3892/mmr.2014.2258] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/16/2014] [Indexed: 11/06/2022] Open
Abstract
The insulin-like growth factors (IGF)-I and -II have a predominant role in fetal growth and development. IGFs are involved in the proliferation, differentiation and apoptosis of fetal cells in vitro and the IGF serum concentration has been shown to be closely correlated with fetal growth and length. IGF transcripts and peptides have been detected in almost every fetal tissue from as early in development as pre‑implantation to the final maturation stage. Furthermore, IGFs have been demonstrated to be involved in limb morphogenesis. However, although ablation of Igf genes in mice resulted in growth retardation and delay in skeletal maturation, no impact on outgrowth and patterning of embryonic limbs was observed. Additionally, various molecular defects in the Igf1 and Igf1r genes in humans have been associated with severe intrauterine growth retardation and impaired skeletal maturation, but not with truncated limbs or severe skeletal dysplasia. The conflicting data between in vitro and in vivo observations with regard to bone morphogenesis suggests that IGFs may not be the sole trophic factors involved in fetal skeletal growth and that redundant mechanisms may exist in chondro- and osteogenesis. Further investigation is required in order to elucidate the functions of IGFs in skeletal development.
Collapse
Affiliation(s)
| | - Stavros Sifakis
- Department of Obstetrics and Gynecology, University Hospital of Heraklion, Crete, Greece
| | | | | |
Collapse
|
35
|
Copy number variants in patients with short stature. Eur J Hum Genet 2013; 22:602-9. [PMID: 24065112 DOI: 10.1038/ejhg.2013.203] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 07/30/2013] [Accepted: 08/07/2013] [Indexed: 12/20/2022] Open
Abstract
Height is a highly heritable and classic polygenic trait. Recent genome-wide association studies (GWAS) have revealed that at least 180 genetic variants influence adult height. However, these variants explain only about 10% of the phenotypic variation in height. Genetic analysis of short individuals can lead to the discovery of novel rare gene defects with a large effect on growth. In an effort to identify novel genes associated with short stature, genome-wide analysis for copy number variants (CNVs), using single-nucleotide polymorphism arrays, in 162 patients (149 families) with short stature was performed. Segregation analysis was performed if possible, and genes in CNVs were compared with information from GWAS, gene expression in rodents' growth plates and published information. CNVs were detected in 40 families. In six families, a known cause of short stature was found (SHOX deletion or duplication, IGF1R deletion), in two combined with a de novo potentially pathogenic CNV. Thirty-three families had one or more potentially pathogenic CNVs (n=40). In 24 of these families, segregation analysis could be performed, identifying three de novo CNVs and nine CNVs segregating with short stature. Four were located near loci associated with height in GWAS (ADAMTS17, TULP4, PRKG2/BMP3 and PAPPA). Besides six CNVs known to be causative for short stature, 40 CNVs with possible pathogenicity were identified. Segregation studies and bioinformatics analysis suggested various potential candidate genes.
Collapse
|
36
|
El-Magd MA, Abbas HE, El-kattawy AM, Mokhbatly A. Novel polymorphisms of the IGF1R gene and their association with average daily gain in Egyptian buffalo (Bubalus bubalis). Domest Anim Endocrinol 2013; 45:105-10. [PMID: 23820243 DOI: 10.1016/j.domaniend.2013.06.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 06/14/2013] [Accepted: 06/15/2013] [Indexed: 11/26/2022]
Abstract
The objective of this study was to detect insulin-like growth factor 1 receptor (IGF1R) polymorphisms, their allele, and genotype frequencies and to determine associations between these polymorphisms and growth traits in Egyptian water buffalo. Three loci of the IGF1R coding region were amplified by RT-PCR and, subsequently, subjected to sequence analysis, followed by single-strand conformation polymorphism to identify different allelic patterns. A total of 11 novel polymorphisms were detected; 6 SNPs among Egyptian water buffaloes and 5 polymorphisms compared with Indian buffalo (Y12700). Three of those polymorphisms; GAG Indel polymorphism, C261G, and G263C SNPs, were nonsynonymous mutations. The GAG Indel polymorphism led to deletion of E (glutamic) amino acid (aa) in the IGF1R of Egyptian water buffaloes compared with Indian buffalo. However, C261G SNP, which replaced A (alanine) by G (glycine) aa, and G263C SNP, which changed A (alanine) to P (proline) aa, were detected among Egyptian water buffaloes. Three different single-strand conformation polymorphism patterns were observed in exon 21: CC/CC, GG/GG, and CG/GC with frequencies of 0.291, 0.253, and 0.556, respectively. The heterozygous animals (CG/GC) had a higher ADG than homozygous animals (CC/CC and GG/GG) from birth to 6 mo of age. We conclude that the heterozygous haplotype, C261G/G263C, in exon 21 of the IGF1R gene is associated with the ADG during the early stages of life (from birth to 6 mo of age) and could be used as a genetic marker for selection of growth traits in Egyptian buffalo.
Collapse
Affiliation(s)
- M A El-Magd
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, El-Geish Street, Kafrelsheikh, Post Box 33516, Egypt.
| | | | | | | |
Collapse
|
37
|
Wang SR, Carmichael H, Andrew SF, Miller TC, Moon JE, Derr MA, Hwa V, Hirschhorn JN, Dauber A. Large-scale pooled next-generation sequencing of 1077 genes to identify genetic causes of short stature. J Clin Endocrinol Metab 2013; 98:E1428-37. [PMID: 23771920 PMCID: PMC3733853 DOI: 10.1210/jc.2013-1534] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
CONTEXT The majority of patients presenting with short stature do not receive a definitive diagnosis. Advances in genetic sequencing allow for large-scale screening of candidate genes, potentially leading to genetic diagnoses. OBJECTIVES The purpose of this study was to discover genetic variants that contribute to short stature in a cohort of children with no known genetic etiology. DESIGN This was a prospective cohort study of subjects with short stature. SETTING The setting was a pediatric endocrinology and genetics clinics at an academic center. PATIENTS A total of 192 children with short stature with no defined genetic etiology and 192 individuals of normal stature from the Framingham Heart Study were studied. INTERVENTION Pooled targeted sequencing using next-generation DNA sequencing technology of the exons of 1077 candidate genes was performed. MAIN OUTCOME MEASURES The numbers of rare nonsynonymous genetic variants found in case patients but not in control subjects, known pathogenic variants in case patients, and potentially pathogenic variants in IGF1R were determined. RESULTS We identified 4928 genetic variants in 1077 genes that were present in case patients but not in control subjects. Of those, 1349 variants were novel (898 nonsynonymous). False-positive rates from pooled sequencing were 4% to 5%, and the false-negative rate was 0.1% in regions covered well by sequencing. We identified 3 individuals with known pathogenic variants in PTPN11 causing undiagnosed Noonan syndrome. There were 9 rare potentially nonsynonymous variants in IGF1R, one of which is a novel, probably pathogenic, frameshift mutation. A previously reported pathogenic variant in IGF1R was present in a control subject. CONCLUSIONS Large-scale sequencing efforts have the potential to rapidly identify genetic etiologies of short stature, but data interpretation is complex. Noonan syndrome may be an underdiagnosed cause of short stature.
Collapse
Affiliation(s)
- Sophie R Wang
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Appleman SS, Kalkwarf HJ, Dwivedi A, Heubi JE. Bone deficits in parenteral nutrition-dependent infants and children with intestinal failure are attenuated when accounting for slower growth. J Pediatr Gastroenterol Nutr 2013; 57:124-30. [PMID: 23518489 PMCID: PMC4303576 DOI: 10.1097/mpg.0b013e318291fec5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The aim of the present study was to determine whether bone mineral content (BMC) and density (BMD) of infants and children with parenteral nutrition (PN)-dependent intestinal failure (IF) is lower than healthy controls, and investigate potential causes of lower BMC and BMD. METHODS We performed a cross-sectional study comparing infants and children with PN-dependent IF with duos of age-, sex-, and race-matched controls. Lumbar spine BMC and BMD were measured by dual-energy x-ray absorptiometry, and serum cytokines, aluminum, insulin-like growth factor-1 (IGF-1), IGF-binding protein 3 (IGF-BP3), parathyroid hormone, 25-hydroxy vitamin D, and 1,25-dihydroxy vitamin D were measured. Generalized estimating equation models accounting for matching were used for comparisons. RESULTS BMC was 15% and BMD was 12% lower in IF participants than in controls (P ≤ 0.004). Group differences were attenuated to 3% and 7% and were not statistically significant (P = 0.40 and P = 0.07) when adjusted for length and weight; length- and weight-for-age were lower in IF than in control participants (12.5% vs 63%; 29.5% vs 54%, P ≤ 0.03). IF participants had higher serum aluminum (23 vs 7 μg/L, P < 0.0001), IGF-1 (97 vs 64 ng/mL, P = 0.04), and 25-hydroxy vitamin D concentrations (40 vs 30 ng/mL, P = 0.0005), and lower IGF-BP3 (1418 vs 1812 ng/mL, P < 0.0001) and parathyroid hormone concentrations (51 vs 98 pg/mL, P = 0.0002) than controls. There was no difference in serum cytokine concentrations (P ≥ 0.09). CONCLUSIONS Growth retardation is a significant problem for patients with PN-dependent IF. Additional investigation is needed to elucidate the cause and its effect on bone mass and density, especially the role of IGF-1 resistance and aluminum toxicity.
Collapse
Affiliation(s)
- Stephanie S. Appleman
- Division of Gastroenterology, Hepatology, and Nutrition Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Heidi J. Kalkwarf
- General and Community Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Alok Dwivedi
- Department of Environmental Health, University of Cincinnati, Cincinnati, OH
| | - James E. Heubi
- Division of Gastroenterology, Hepatology, and Nutrition Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
39
|
Migliano AB, Romero IG, Metspalu M, Leavesley M, Pagani L, Antao T, Huang DW, Sherman BT, Siddle K, Scholes C, Hudjashov G, Kaitokai E, Babalu A, Belatti M, Cagan A, Hopkinshaw B, Shaw C, Nelis M, Metspalu E, Mägi R, Lempicki RA, Villems R, Lahr MM, Kivisild T. Evolution of the Pygmy Phenotype: Evidence of Positive Selection from Genome-wide Scans in African, Asian, and Melanesian Pygmies. Hum Biol 2013; 85:251-84. [DOI: 10.3378/027.085.0313] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2013] [Indexed: 11/05/2022]
|
40
|
Leal AC, Montenegro LR, Saito RF, Ribeiro TC, Coutinho DC, Mendonca BB, Arnhold IJP, Jorge AAL. Analysis of the insulin-like growth factor 1 receptor gene in children born small for gestational age: in vitro characterization of a novel mutation (p.Arg511Trp). Clin Endocrinol (Oxf) 2013; 78:558-63. [PMID: 22998174 DOI: 10.1111/cen.12048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/11/2012] [Accepted: 08/31/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Insulin-like growth factor 1 insensitivity caused by IGF1R mutations has been previously identified as one of the causes of growth impairment in children born small for gestational age (SGA). OBJECTIVE To analyse the IGF1R in children born SGA. SUBJECTS From an initial cohort of 54 sequential children born SGA, without catch-up growth, 25 children were selected for this IGF1R study due to the presence of serum IGF-1 values above the mean for their age and sex. METHODS The proximal IGF1R promoter region, the entire coding region and the exon-intron boundaries were directly sequenced, and multiplex ligation-dependent probe amplification analysis was performed. Fibroblast cultures were developed from one patient with a mutation for the in vitro characterization of IGF-1 insensitivity. RESULTS The copy number variation analysis did not identify deletions involving the IGF1R gene. We identified two children carrying heterozygous nucleotide substitutions in IGF1R: c.16G>A/p.Gly6Arg and c.1531C>T/p.Arg511Trp. The first variant (p.Gly6Arg) was identified in control subjects (0·3%) and in a relative with normal growth; thus, it was considered to be a rare benign allelic variation. The second variant (p.Arg511Trp) was not found in 306 alleles from control subjects, and it segregated with the growth impairment phenotype in the patient's family. Fibroblasts obtained from this patient had a significantly reduced proliferative response and AKT phosphorylation after IGF-1 stimulation compared with control fibroblasts. CONCLUSION The identification of an inactivating IGF1R mutation in the present cohort should encourage further studies of larger series to establish the precise frequency of this molecular defect in children with growth impairment of a prenatal onset.
Collapse
Affiliation(s)
- Andrea C Leal
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM/25, Disciplina de Endocrinologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Labarta JI, Barrio E, Audí L, Fernández-Cancio M, Andaluz P, de Arriba A, Puga B, Calvo MT, Mayayo E, Carrascosa A, Ferrández-Longás A. Familial short stature and intrauterine growth retardation associated with a novel mutation in the IGF-I receptor (IGF1R) gene. Clin Endocrinol (Oxf) 2013; 78:255-62. [PMID: 22738321 DOI: 10.1111/j.1365-2265.2012.04481.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/15/2012] [Accepted: 06/21/2012] [Indexed: 11/27/2022]
Abstract
CONTEXT IGF-I is essential for normal human growth and mediates its effects through the IGF1R. IGF1R mutations have been associated with varying degrees of intrauterine and postnatal growth retardation. OBJECTIVE To identify IGF1R gene mutations in a short-statured family with intrauterine growth retardation and microcephaly. METHODS Direct DNA sequencing was used to identify IGF1R mutations. Multiplex ligation-dependent probe amplification analyses were performed for deletions and duplications of all IGF1R exons. Functional studies were conducted to assess mutation pathogenicity. RESULTS A novel heterozygous IGF1R missense mutation in exon 7 (c.A1549T, p.Y487F) was identified in a short-statured girl with severe prenatal growth retardation and microcephaly. The same mutation was also identified in her mother, who presented prenatal and postnatal growth failure, and her short-statured maternal grandmother, both of whom exhibited microcephaly. The index case showed a partial response to rhGH. Functional studies performed in dermal fibroblasts from the index case and her mother showed normal IGF-I binding; however, IGF-I activation of intracellular signalling measured as AKT and extracellular signal-regulated kinase phosphorylation was markedly reduced, with patients' values being lower than those of her mother. IGF-I stimulation of DNA synthesis was significantly reduced compared with controls. CONCLUSION Our results show a novel missense mutation in the IGF1R gene (c.A1549T, p.Y487F) associated with prenatal and postnatal growth failure and microcephaly in the context of familial short stature. The functional studies are in line with the inactivation of one copy of the IGF1R gene with variable expression within the same family.
Collapse
Affiliation(s)
- José I Labarta
- Endocrinology Unit, Department of Pediatrics, Hospital Infantil Universitario Miguel Servet, Zaragoza, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Fadiel A, Epperson B, Shaw MI, Hamza A, Petito J, Naftolin F. Bioinformatic analysis of benzo-α-pyrene-induced damage to the human placental insulin-like growth factor-1 gene. Reprod Sci 2013; 20:917-28. [PMID: 23344457 DOI: 10.1177/1933719112468946] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Intrauterine growth restriction (IUGR) has been associated with exposure to polyaromatic hydrocarbons (PAHs) which are released in the combustion of oil, fuel, gas, garbage, and tobacco. Pregnant women exposed to PAHs are at risk of the effects of these environmental toxins; for example, benzo-α-pyrene (BαP) is able to enter the blood stream and could contribute to IUGR or other developmental abnormalities via effects on the placental cells. Since IUGR has been associated with decreased cord blood concentrations of immunoreactive insulin-like growth factor 1 (ir-IGF-1) and IUGR has been associated with disordered development and fetal programming, we tested the effects of BαP on human placental trophoblast cells in culture. EXPERIMENTAL IGF-1 expression and activation was studied using an immortalized human placental trophoblast cell line (HTR-8). The cells were treated with vehicle control or 1 µmol/L BαP, or 5 µmol/L BαP for 12 hours. RNA was extracted and the exons of IGF-1 were amplified using reverse transcriptase-polymerase chain reaction (RT-PCR). The ir-IGF-1 expression levels were compared using gel electrophoresis. The PCR products were sequenced, and levels of mutation were measured with comparative sequence analysis. A computational protein analysis (computer simulation) was performed in order to assess the potential impact of BαP-associated mutation on IGF-1 protein function. RESULTS The IGF-1 expression decreased considerably in BαP-treated cells relative to untreated controls (P < .05), also in a dose-dependent manner. Comparative sequence analysis indicated that the level of BαP exposure correlated with the percentage of base pair mutations in IGF-1 nucleotide sequences for both treatment groups (P < .05). Shifts were observed in the open reading frame, indicating a possible change in the IGF-1 start codon. Protein folding simulation analysis indicated that the base pair changes induced by BαP weakened IGF-1-IGF binding protein (IGFBP) interaction. CONCLUSIONS In concordance with the previous findings, exposure of human placental trophoblast cells to BαP exposure results in reduction of IGF-1 expression and base pair mutations. The direct action of BαP on the placenta indicates that it may not be necessary for BαP to access other maternal tissues in order for gene abnormalities to occur. Given that PAHs are known to work through aryl hydrocarbon hydrolase (AHH), these results are likely due to the presence of AHH in HTR cells. Computational modeling of BαP actions on IGF1, substrate-ligand binding, supports the biological premise of this work and underlines the need to determine actual biological effects rather than equating immune to bioactivity of IGF1.
Collapse
Affiliation(s)
- A Fadiel
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Gannagé-Yared MH, Klammt J, Chouery E, Corbani S, Mégarbané H, Abou Ghoch J, Choucair N, Pfäffle R, Mégarbané A. Homozygous mutation of the IGF1 receptor gene in a patient with severe pre- and postnatal growth failure and congenital malformations. Eur J Endocrinol 2013; 168:K1-7. [PMID: 23045302 DOI: 10.1530/eje-12-0701] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Heterozygous mutations in the IGF1 receptor (IGF1R) gene lead to partial resistance to IGF1 and contribute to intrauterine growth retardation (IUGR) with postnatal growth failure. To date, homozygous mutations of this receptor have not been described. SUBJECT A 13.5-year-old girl born from healthy first-cousin parents presented with severe IUGR and persistent short stature. Mild intellectual impairment, dysmorphic features, acanthosis nigricans, and cardiac malformations were also present. METHODS Auxological and endocrinological profiles were measured. All coding regions of the IGF1R gene including intron boundaries were amplified and directly sequenced. Functional characterization was performed by immunoblotting using patient's fibroblasts. RESULTS IGF1 level was elevated at 950NG/ML (+7 S.D.). Fasting glucose level was normal associated with high insulin levels at baseline and during an oral glucose tolerance test. Fasting triglyceride levels were elevated. sequencing of the IGF1R gene led to the identification of a homozygous variation in exon 2: c.119G>T (p.Arg10Leu). As a consequence, IGF1-dependent receptor autophosphorylation and downstream signaling were reduced in patient's fibroblasts. Both parents were heterozygous for the mutation. CONCLUSION The homozygous mutation of the IGF1R is associated with severe IUGR, dysmorphic features, and insulin resistance, while both parents were asymptomatic heterozygous carriers of the same mutation.
Collapse
|
44
|
Kansra AR, Dolan LM, Martin LJ, Deka R, Chernausek SD. IGF receptor gene variants in normal adolescents: effect on stature. Eur J Endocrinol 2012; 167:777-81. [PMID: 22972910 DOI: 10.1530/eje-12-0565] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE IGF1 is essential for human growth and mediates its effects through the type 1 IGF receptor (IGF1R). Our objective was to determine the frequency of certain previously reported IGF1R gene variants in the normal population and their effect on stature. DESIGN A cross-sectional study was conducted in a population of 2500 children enrolled in public school grades 5 through 12 for whom DNA and anthropometric data were available. Subjects were genotyped at five previously reported loci that affect receptor abundance or function. METHODS The frequency of the following IGF1R variants Arg108Gln, Lys115Asn, Arg59stop, Arg481Gln, and Arg605His was measured by a PCR-based assay. Circulating concentrations of IGF1 or IGF binding protein-3 (IGFBP3) were measured by ELISA in those affected and matched controls. RESULTS A scan of 1300 subjects detected none with Arg108Gln, Lys115Asn, or Arg59stop mutations. In contrast, nucleotide changes leading to heterozygosity at codon 605 were identified in nine of 2500 subjects and six of 1800 subjects at codon 481. These individuals were, on average, 4 cm shorter than the others. There were no differences in circulating concentrations of IGF1 or IGFBP3 between those with the gene variants and controls matched for sex, ethnicity, age, and BMI. CONCLUSION Rare IGF1R variants exerting a moderate effect on stature are present in the general population, supporting the importance of IGF1R function in growth control and indicating that variation in height within healthy individuals may be explained, in some cases, by larger effects of a small subset of gene variants.
Collapse
Affiliation(s)
- Alvina R Kansra
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | | | |
Collapse
|
45
|
Epaud R, Aubey F, Xu J, Chaker Z, Clemessy M, Dautin A, Ahamed K, Bonora M, Hoyeau N, Fléjou JF, Mailleux A, Clement A, Henrion-Caude A, Holzenberger M. Knockout of insulin-like growth factor-1 receptor impairs distal lung morphogenesis. PLoS One 2012; 7:e48071. [PMID: 23139760 PMCID: PMC3491012 DOI: 10.1371/journal.pone.0048071] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 09/19/2012] [Indexed: 11/19/2022] Open
Abstract
Background Insulin-like growth factors (IGF-I and -II) are pleiotropic regulators of somatic growth and development in vertebrate species. Endocrine and paracrine effects of both hormones are mediated by a common IGF type 1 receptor (IGF-1R). Lethal respiratory failure in neonatal IGF-1R knockout mice suggested a particular role for this receptor in pulmonary development, and we therefore investigated the consequences of IGF-1R inactivation in lung tissue. Methods and Findings We first generated compound heterozygous mutant mice harboring a hypomorphic (Igf1rneo) and a null (Igf1r−) allele. These IGF-1Rneo/− mice express only 22% of normal IGF-1R levels and are viable. In adult IGF-1Rneo/− mice, we assessed lung morphology and respiratory physiology and found normal histomorphometric characteristics and normal breathing response to hypercapnia. We then generated homozygous IGF-1R knockout mutants (IGF-1R−/−) and analyzed their lung development during late gestation using histomorphometric and immunohistochemical methods. IGF-1R−/− embryos displayed severe lung hypoplasia and markedly underdeveloped diaphragms, leading to lethal neonatal respiratory distress. Importantly, IGF-1R−/− lungs from late gestation embryos were four times smaller than control lungs and showed markedly thickened intersaccular mesenchyme, indicating strongly delayed lung maturation. Cell proliferation and apoptosis were significantly increased in IGF-1R−/− lung tissue as compared with IGF-1R+/+ controls. Immunohistochemistry using pro-SP-C, NKX2-1, CD31 and vWF as markers revealed a delay in cell differentiation and arrest in the canalicular stage of prenatal respiratory organ development in IGF-1R−/− mutant mice. Conclusions/Significance We found that low levels of IGF-1R were sufficient to ensure normal lung development in mice. In contrast, complete absence of IGF-1R significantly delayed end-gestational lung maturation. Results indicate that IGF-1R plays essential roles in cell proliferation and timing of cell differentiation during fetal lung development.
Collapse
Affiliation(s)
- Ralph Epaud
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
- INSERM U955, Faculté de Médecine, Université Paris-Est, Créteil, France
| | - Flore Aubey
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
| | - Jie Xu
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
| | - Zayna Chaker
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
| | - Maud Clemessy
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
| | - Alexandre Dautin
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
| | - Karmène Ahamed
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
| | - Monique Bonora
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
| | - Nadia Hoyeau
- UPMC, Université Paris 6, Paris, France
- APHP, Hôpital Saint Antoine, Paris, France
| | - Jean-François Fléjou
- UPMC, Université Paris 6, Paris, France
- APHP, Hôpital Saint Antoine, Paris, France
| | | | - Annick Clement
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
- APHP, Hôpital Trousseau, Paris, France
| | - Alexandra Henrion-Caude
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
- INSERM UMRS 781, Hôpital Necker-Enfants Malades, Paris, France
| | - Martin Holzenberger
- INSERM UMRS 938, Hôpital Saint-Antoine, Paris, France
- UPMC, Université Paris 6, Paris, France
- * E-mail:
| |
Collapse
|
46
|
Zhang XQ. Intrauterine growth restriction and genetic determinants - existing findings, problems, and further direction. World J Obstet Gynecol 2012; 1:20-28. [DOI: 10.5317/wjog.v1.i3.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Fetal growth is determined largely by the nutrient supply, placental transport function, and growth hormones. Recently, gene mutation and expression, especially of those genes associated with the proteins that are related to the fetal growth, have been reported to play an important role in the development of intrauterine growth restriction (IUGR). Fetal growth epigenetics, a new concept in fetal growth, has resulted from studies on fetal programing. This paper outlines the findings of our serial studies on IUGR, and summarizes data on IUGR animal models, placental function in transferring nutrients, cell proliferation dynamics in IUGR, and experimental treatment of IUGR. We review genetic approaches to IUGR, especially those relating to growth factor genes, angiotensinogen genes and other gene mutations. We also discuss the epigenetics of fetal growth and future study directions on fetal growth restriction. These should be valuable in elucidating the mechanisms employed by the fetus and in helping to develop interventional strategies that might prevent the development of IUGR.
Collapse
|
47
|
Leal ADC, Canton APM, Montenegro LR, Coutinho DC, Arnhold IJP, Jorge AADL. [Mutations in insulin-like growth factor receptor 1 gene (IGF1R) resulting in intrauterine and postnatal growth retardation]. ACTA ACUST UNITED AC 2012; 55:541-9. [PMID: 22218435 DOI: 10.1590/s0004-27302011000800007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 10/20/2011] [Indexed: 11/22/2022]
Abstract
Approximately 10% of children born small-for-gestational age (SGA) do not show spontaneous growth catch-up. The causes of this deficit in prenatal growth and its maintenance after birth are not completely known, in most cases. Over the past eight years, several heterozygous inactivating mutations and deletions in IGF1R gene have been reported, indicating the role of defects in the IGFs/IGF1R axis as a cause of growth deficit. It has been hypothesized that at least 2.5% of children born SGA may have IGF1R gene defects. The clinical presentation of these patients is highly variable in the severity of growth retardation and hormonal parameters. In the most evident cases, patients have microcephaly, mild cognitive impairment and high levels of IGF-1, associated with short stature of prenatal onset. This review will describe the clinical, molecular and treatment of short stature with hrGH of children with mutations in the IGF1R gene.
Collapse
Affiliation(s)
- Andréa de Castro Leal
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular, Hospital das Clínicas, São Paulo, Brasil
| | | | | | | | | | | |
Collapse
|
48
|
O’Kusky J, Ye P. Neurodevelopmental effects of insulin-like growth factor signaling. Front Neuroendocrinol 2012; 33:230-51. [PMID: 22710100 PMCID: PMC3677055 DOI: 10.1016/j.yfrne.2012.06.002] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/09/2012] [Accepted: 06/07/2012] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factor (IGF) signaling greatly impacts the development and growth of the central nervous system (CNS). IGF-I and IGF-II, two ligands of the IGF system, exert a wide variety of actions both during development and in adulthood, promoting the survival and proliferation of neural cells. The IGFs also influence the growth and maturation of neural cells, augmenting dendritic growth and spine formation, axon outgrowth, synaptogenesis, and myelination. Specific IGF actions, however, likely depend on cell type, developmental stage, and local microenvironmental milieu within the brain. Emerging research also indicates that alterations in IGF signaling likely contribute to the pathogenesis of some neurological disorders. This review summarizes experimental studies and shed light on the critical roles of IGF signaling, as well as its mechanisms, during CNS development.
Collapse
Affiliation(s)
- John O’Kusky
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada V5Z 1M9
| | - Ping Ye
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
49
|
Kawashima Y, Higaki K, Fukushima T, Hakuno F, Nagaishi JI, Hanaki K, Nanba E, Takahashi SI, Kanzaki S. Novel missense mutation in the IGF-I receptor L2 domain results in intrauterine and postnatal growth retardation. Clin Endocrinol (Oxf) 2012; 77:246-54. [PMID: 22309212 DOI: 10.1111/j.1365-2265.2012.04357.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND IGFs play key roles in intrauterine and postnatal growth through the IGF-I receptor (IGF-IR). We identified a family bearing a new heterozygous missense mutation at the L2 domain of IGF-IR (R431L). METHOD We analysed the nucleotide sequences of the IGF1R gene of the family. We prepared R(-) cells (fibroblasts with targeted disruption of the IGF-IR gene) expressing wild-type or R431L IGF-IR and performed functional analyses by evaluating IGF-I binding, IGF-I-stimulated DNA synthesis, tyrosine phosphorylation of IGF-IR and its substrates, and internalization by measuring [(125) I]IGF-I internalization. We also performed confocal microscopy analysis. RESULTS We identified a family bearing a new heterozygous missense mutation at the L2 domain of IGF-IR (R431L) through an 8-year-old girl and her mother, both born with intrauterine growth retardation. In experiments conducted using cells homozygously transfected with the IGF-IR R431L mutation; (i) IGF-I binding was not affected; (ii) DNA synthesis induced by IGF-I was decreased; (iii) IGF-IR internalization stimulated by IGF-I was decreased and (iv) IGF-I-stimulated tyrosine phosphorylation was reduced IGF-IR by low concentrations of IGF-I and on insulin receptor substrate (IRS)-1 and IRS-2. CONCLUSION A missense mutation (R431L) leads to the inhibition of cell proliferation, attenuation of IGF signalling and decrease in internalization of IGF-IR. The results of this study suggest a novel link between a mutation at the IGF-IR L2 domain and intrauterine and postnatal growth retardation.
Collapse
Affiliation(s)
- Yuki Kawashima
- Division of Pediatrics & Perinatology, Research Center for Bioscience and Technology, Tottori University Faculty of Medicine, Yonago, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Backeljauw PF, Chernausek SD. The insulin-like growth factors and growth disorders of childhood. Endocrinol Metab Clin North Am 2012; 41:265-82, v. [PMID: 22682630 DOI: 10.1016/j.ecl.2012.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Specific lesions of the growth hormone (GH)/insulin-like growth factor (IGF) axis have been identified in humans, each of which has distinctive auxologic and biochemical features. Measures of circulating IGF-I are useful in diagnosing growth disorders in childhood and in evaluating response to GH therapy. Recombinant human IGF-I is an effective treatment of severe primary IGF deficiency, which is typical of patients with GH receptor defects (Laron syndrome). Such treatment has been limited to a few severely affected patients. Future studies will provide new insight into IGF-I as treatment and into the nature of growth disorders that involve the IGF axis.
Collapse
Affiliation(s)
- Philippe F Backeljauw
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnett Avenue, Cincinnati, OH 45229, USA
| | | |
Collapse
|