Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Heliyon ; 10(11): e30887, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38841436

RESUMO

Fibroblast growth factors and their receptors (FGFR) have major roles in both human growth and oncogenesis. In adults, therapeutic FGFR inhibitors have been successful against tumors that carry somatic FGFR mutations. In pediatric patients, trials testing these anti-tumor FGFR inhibitor therapeutics are underway, with several recent reports suggesting modest positive responses. Herein, we report an unforeseen outcome in a pre-pubescent child with an FGFR1-mutated glioma who was successfully treated with FDA-approved erdafitinib, a pan-FGFR inhibitor approved for treatment of Bladder tumors. While on treatment with erdafitinib, the patient experienced rapid skeletal and long bone overgrowth resulting in kyphoscoliosis, reminiscent of patients with congenital loss-of-function FGFR3 mutations. We utilized normal dermal fibroblast cells established from the patient as a surrogate model to demonstrate that insulin-like growth factor 1 (IGF-1), a factor important for developmental growth of bones and tissues, can activate the PI3K/AKT pathway in erdafitinib-treated cells but not the MAPK/ERK pathway. The IGF-I-activated PI3K/AKT signaling rescued normal fibroblasts from the cytotoxic effects of erdafitinib by promoting cell survival. We, therefore, postulate that IGF-I-activated P13K/AKT signaling likely continues to promote bone elongation in the growing child, but not in adults, treated with therapeutic pan-FGFR inhibitors. Importantly, since activated MAPK signaling counters bone elongation, we further postulate that prolonged blockage of the MAPK pathway with pan-FGFR inhibitors, together with actions of growth-promoting factors including IGF-1, could explain the abnormal skeletal and axial growth suffered by our pre-pubertal patient during systemic therapeutic use of pan-FGFR inhibitors. Further studies to find more targeted, and/or appropriate dosing, of pan-FGFR inhibitor therapeutics for children are essential to avoid unexpected off-target effects as was observed in our young patient.

2.
Horm Res Paediatr ; 96(5): 449-457, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36646053

RESUMO

INTRODUCTION: The metalloproteinase pregnancy-associated plasma protein A2 (PAPP-A2) cleaves insulin-like growth factor (IGF)-binding proteins 3 and 5 to release bioactive IGF-I from its ternary complex. Patients with mutations in PAPP-A2 have growth failure and low free IGF-I despite elevated total IGF-I. We describe 5-year treatment response to recombinant human IGF-1 (rhIGF-1) in a patient with PAPP-A2 deficiency, and the phenotype of PAPP-A2 deficiency in three siblings. METHODS: Two siblings (P2, P3) with PAPP-A2 deficiency were recruited for rhIGF-1 therapy at 120 µg/kg subcutaneous twice daily, along with a third sibling (P1) for phenotyping. We evaluated efficacy and safety of rhIGF-1 therapy, including effect on metabolic measures and bone mineral density (BMD). RESULTS: Treatment with rhIGF-1 was started in 10.4-year- (P3) and 14.5-year (P2)-old brothers. P2 discontinued therapy due to pseudotumor cerebri. P3 continued rhIGF-1 for 5 years; height velocity increased (3.0 cm/year at baseline; 5.0-7.6 cm/year thereafter) as did height SDS (+0.6). P3's pubertal onset was at 12.4 year. BMD height-adjusted Z-score modestly improved for lumbar spine (+0.4), and decreased in forearm (-0.2) and hip (-0.3). All siblings had hyperinsulinemia. Impaired glucose tolerance (IGT) resolved in P1. P2 showed worsening glucose tolerance (2-h glucose: 225 mg/dL). Impaired fasting glucose and hyperinsulinemia initially resolved for P3, but IGT (2-h glucose: 152 mg/dL) developed during puberty. CONCLUSION: Therapy with rhIGF-1 modestly improved linear growth in one patient with PAPP-A2 deficiency, but without true catch-up. Therapy was associated with pseudotumor cerebri in a sibling. Initial improvement in BMD and glycemic pattern on rhIGF-1 was not sustained during puberty.


Assuntos
Intolerância à Glucose , Hiperinsulinismo , Pseudotumor Cerebral , Masculino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Glucose , Proteínas Recombinantes/uso terapêutico
3.
Am J Med Genet A ; 188(4): 1193-1203, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35001504

RESUMO

Aggrecan is a proteoglycan within the physeal and articular cartilage. Aggrecan deficiency, due to heterozygous mutations in the ACAN gene, causes dominantly inherited short stature and, in many patients, early-onset osteoarthritis and degenerative disc disease. We aimed to further characterize this phenotypic spectrum with an emphasis on musculoskeletal health. Twenty-two individuals from nine families were enrolled. Histories and examinations focused on joint health, gait analysis, joint specific patient reported outcomes, and imaging studies were performed. All patients had dominantly inherited short stature, with the exception of a de novo mutation. Short stature was worse in adults versus children (median height -3.05 SD vs. -2.25 SD). ACAN mutations were not always associated with bone age advancement (median advancement +1.1 years, range 0 to +2 years). Children had subtle disproportionality and clinically silent joint disease-25% with osteochondritis dissecans (OD). Adults had a high prevalence of joint symptomatology-decline in knee function, disability from spinal complaints, and lower physical activity on outcome measures. Osteoarthritis (OA) and OD was detected in 90% of adults, and orthopedic surgeries were reported in 60%. Aggrecan deficiency leads to short stature with progressive decline in height SD, mild skeletal dysplasia, and increasing prevalence of joint pathology over time. Optimal musculoskeletal health and quality of life can be attained with timely identification of pathology and intervention.


Assuntos
Nanismo , Osteoartrite , Agrecanas/genética , Nanismo/genética , Heterozigoto , Humanos , Fenótipo , Qualidade de Vida
4.
Birth Defects Res ; 112(19): 1733-1737, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32935482

RESUMO

Proteus syndrome is a mosaic genetic overgrowth disorder caused by a postzygotic, mosaic activating mutation in AKT1. Rare prenatal presentations include segmental tissue overgrowth, and skeletal and CNS anomalies. We present the first report of prenatally diagnosed and molecularly confirmed Proteus syndrome. Prenatal imaging identified megalencephaly, brain and eye malformations, focal soft tissue enlargement, and ambiguous genitalia. Exome sequencing performed on cultured amniocytes demonstrated an AKT1 pathogenic variant consistent with Proteus syndrome, and postnatal examination confirmed the diagnosis. Postnatal Sanger sequencing could not identify the AKT1 pathogenic variant. This case underscores the importance of prenatal exome sequencing on cultured amniocytes for mosaic overgrowth disorders, as well as provides additional information on the prenatal phenotype of Proteus syndrome, and highlights the impact of prenatal diagnosis on postnatal management.


Assuntos
Síndrome de Proteu , Feminino , Humanos , Mutação , Fenótipo , Gravidez , Diagnóstico Pré-Natal , Síndrome de Proteu/diagnóstico , Síndrome de Proteu/genética , Proteínas Proto-Oncogênicas c-akt/genética
5.
Am J Hum Genet ; 104(4): 758-766, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30929739

RESUMO

By using exome sequencing and a gene matching approach, we identified de novo and inherited pathogenic variants in KDM3B in 14 unrelated individuals and three affected parents with varying degrees of intellectual disability (ID) or developmental delay (DD) and short stature. The individuals share additional phenotypic features that include feeding difficulties in infancy, joint hypermobility, and characteristic facial features such as a wide mouth, a pointed chin, long ears, and a low columella. Notably, two individuals developed cancer, acute myeloid leukemia and Hodgkin lymphoma, in childhood. KDM3B encodes for a histone demethylase and is involved in H3K9 demethylation, a crucial part of chromatin modification required for transcriptional regulation. We identified missense and truncating variants, suggesting that KDM3B haploinsufficiency is the underlying mechanism for this syndrome. By using a hybrid facial-recognition model, we show that individuals with a pathogenic variant in KDM3B have a facial gestalt, and that they show significant facial similarity compared to control individuals with ID. In conclusion, pathogenic variants in KDM3B cause a syndrome characterized by ID, short stature, and facial dysmorphism.


Assuntos
Anormalidades Craniofaciais/genética , Deficiências do Desenvolvimento/genética , Nanismo/genética , Variação Genética , Deficiência Intelectual/genética , Histona Desmetilases com o Domínio Jumonji/genética , Anormalidades Musculoesqueléticas/genética , Estatura , Criança , Exoma , Face , Feminino , Estudos de Associação Genética , Mutação em Linhagem Germinativa , Haploinsuficiência , Histonas/química , Humanos , Masculino , Mutação de Sentido Incorreto , Fenótipo
6.
Nat Genet ; 51(1): 96-105, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30478443

RESUMO

DNA methylation and Polycomb are key factors in the establishment of vertebrate cellular identity and fate. Here we report de novo missense mutations in DNMT3A, which encodes the DNA methyltransferase DNMT3A. These mutations cause microcephalic dwarfism, a hypocellular disorder of extreme global growth failure. Substitutions in the PWWP domain abrogate binding to the histone modifications H3K36me2 and H3K36me3, and alter DNA methylation in patient cells. Polycomb-associated DNA methylation valleys, hypomethylated domains encompassing developmental genes, become methylated with concomitant depletion of H3K27me3 and H3K4me3 bivalent marks. Such de novo DNA methylation occurs during differentiation of Dnmt3aW326R pluripotent cells in vitro, and is also evident in Dnmt3aW326R/+ dwarf mice. We therefore propose that the interaction of the DNMT3A PWWP domain with H3K36me2 and H3K36me3 normally limits DNA methylation of Polycomb-marked regions. Our findings implicate the interplay between DNA methylation and Polycomb at key developmental regulators as a determinant of organism size in mammals.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Nanismo/genética , Mutação com Ganho de Função/genética , Microcefalia/genética , Proteínas do Grupo Polycomb/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , DNA Metiltransferase 3A , Metilases de Modificação do DNA/genética , Feminino , Células HeLa , Histonas/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos/genética , Ligação Proteica/genética , Sequências Reguladoras de Ácido Nucleico/genética
7.
J Clin Endocrinol Metab ; 102(12): 4568-4577, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29029190

RESUMO

Context: The pregnancy-associated plasma protein A2 (PAPP-A2) cleaves insulinlike growth factor binding proteins 3 and 5, releasing free insulinlike growth factor 1 (IGF-1). Homozygous mutations in PAPP-A2 result in growth failure with elevated total but low free IGF-1. Objective: To determine the 24-hour pharmacokinetic (PK) profile of free and total IGF-1 after a dose of recombinant human insulinlike growth factor 1 (rhIGF-1). We describe the growth response and effects on glucose metabolism and bone mineral density (BMD) after 1 year of rhIGF-1 therapy. Design and Patients: Three affected siblings, their heterozygous parents, and two healthy controls participated. The subjects received a dose of rhIGF-1, followed by serial blood samples collected over 24 hours. The two younger siblings were started on rhIGF-1 treatment. An oral glucose tolerance test and dual-energy X-ray absorptiometry scans were obtained at baseline and after 1 year of treatment. Results: Subcutaneous administration of rhIGF-1 increased the concentration of free and total IGF-1 in patients with PAPP-A2 deficiency. The PK profile was comparable in all participants. At baseline, all three subjects demonstrated insulin resistance and below-average BMD. Treatment with rhIGF-1 is ongoing in the youngest patient but was discontinued in his brother because of the development of pseudotumor cerebri. The treated patient had an increase in height velocity from 3.0 to 6.2 cm/y, resolution of insulin resistance, and an increase in total body BMD. Conclusions: rhIGF-1 at standard dosages resulted in similar PK characteristics in patients with PAPP-A2 deficiency, heterozygous relatives, and healthy controls. The youngest affected patient experienced a modest growth response to therapy with rhIGF-1, as well as beneficial effects on glucose metabolism and bone mass.


Assuntos
Glicemia/metabolismo , Densidade Óssea/efeitos dos fármacos , Crescimento/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacocinética , Proteína Plasmática A Associada à Gravidez/deficiência , Absorciometria de Fóton , Adolescente , Adulto , Composição Corporal/efeitos dos fármacos , Criança , Família , Feminino , Teste de Tolerância a Glucose , Humanos , Injeções Subcutâneas , Resistência à Insulina/genética , Masculino , Projetos Piloto , Estudos Prospectivos , Proteínas Recombinantes/farmacocinética , Adulto Jovem
8.
Pediatr Endocrinol Rev ; 14(3): 289-297, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28508599

RESUMO

CDKN1C is a cyclin-dependent kinase Inhibitor and negative regulator of cellular proliferation. Recently, gain-of-function mutations in the PCNA domain of CDKN1C have been reported as the genetic basis of various growth-retarded syndromes including IMAGe syndrome, Russell Silver syndrome as well as a novel undergrowth syndrome that additionally exhibited early adulthood onset diabetes. This review summarizes the key clinical features and the molecular advances that have contributed to our understanding of this complex phenotypic spectrum.


Assuntos
Insuficiência Adrenal/genética , Insuficiência Adrenal/patologia , Inibidor de Quinase Dependente de Ciclina p57/genética , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/patologia , Transtornos do Crescimento/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/patologia , Insuficiência Adrenal/classificação , Retardo do Crescimento Fetal/classificação , Humanos , Masculino , Mutação , Osteocondrodisplasias/classificação , Fenótipo , Síndrome , Anormalidades Urogenitais/classificação
9.
Ann Endocrinol (Paris) ; 78(2): 80-82, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28495324

RESUMO

Normal growth requires that pituitary-secreted growth hormone (GH) bind to its specific receptor and activate a complex signaling cascade, leaving to production of insulin-like growth factor-I (IGF-I), which, in turn, activates its own receptor (IGF1R). The GH receptor (GHR) is preformed as a dimer and is transported in a nonligand bound state to the cell surface. Binding of GH to the GHR dimer, results in a conformational change of the dimer, activation of the intracellular Janus Kinase 2 (JAK2) and phosphorylation of signal transducer and activator of transcription (STAT) 5B. Phosphorylated STAT5B dimers are then translocated to the nucleus, where they transcriptionally activate multiple genes, including those for IGF-I, IGF binding protein-3 and the acid-labile subunit (ALS).


Assuntos
Somatotrofos/fisiologia , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Doenças da Hipófise/genética , Doenças da Hipófise/metabolismo , Receptor IGF Tipo 1/genética , Fatores de Transcrição STAT/genética , Somatotrofos/metabolismo
10.
J Clin Endocrinol Metab ; 102(2): 460-469, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27870580

RESUMO

CONTEXT: Heterozygous mutations in the aggrecan gene (ACAN) cause autosomal dominant short stature with accelerated skeletal maturation. OBJECTIVE: We sought to characterize the phenotypic spectrum and response to growth-promoting therapies. PATIENTS AND METHODS: One hundred three individuals (57 females, 46 males) from 20 families with autosomal dominant short stature and heterozygous ACAN mutations were identified and confirmed using whole-exome sequencing, targeted next-generation sequencing, and/or Sanger sequencing. Clinical information was collected from the medical records. RESULTS: Identified ACAN variants showed perfect cosegregation with phenotype. Adult individuals had mildly disproportionate short stature [median height, -2.8 standard deviation score (SDS); range, -5.9 to -0.9] and a history of early growth cessation. The condition was frequently associated with early-onset osteoarthritis (12 families) and intervertebral disc disease (9 families). No apparent genotype-phenotype correlation was found between the type of ACAN mutation and the presence of joint complaints. Childhood height was less affected (median height, -2.0 SDS; range, -4.2 to -0.6). Most children with ACAN mutations had advanced bone age (bone age - chronologic age; median, +1.3 years; range, +0.0 to +3.7 years). Nineteen individuals had received growth hormone therapy with some evidence of increased growth velocity. CONCLUSIONS: Heterozygous ACAN mutations result in a phenotypic spectrum ranging from mild and proportionate short stature to a mild skeletal dysplasia with disproportionate short stature and brachydactyly. Many affected individuals developed early-onset osteoarthritis and degenerative disc disease, suggesting dysfunction of the articular cartilage and intervertebral disc cartilage. Additional studies are needed to determine the optimal treatment strategy for these patients.


Assuntos
Agrecanas/genética , Nanismo/genética , Mutação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antropometria/métodos , Braquidactilia/genética , Criança , Pré-Escolar , Análise Mutacional de DNA/métodos , Nanismo/tratamento farmacológico , Feminino , Crescimento/genética , Hormônio do Crescimento/uso terapêutico , Heterozigoto , Humanos , Lactente , Degeneração do Disco Intervertebral/genética , Deslocamento do Disco Intervertebral/genética , Masculino , Pessoa de Meia-Idade , Osteocondrite Dissecante/congênito , Osteocondrite Dissecante/genética , Linhagem , Fenótipo , Adulto Jovem
11.
Am J Hum Genet ; 98(6): 1235-1242, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27259054

RESUMO

Whole-exome sequencing (WES) is increasingly being utilized to diagnose individuals with undiagnosed disorders. Developmental delay and short stature are common clinical indications for WES. We performed WES in three families, using proband-parent trios and two additional affected siblings. We identified a syndrome due to an autosomal-recessively inherited deficiency of transketolase, encoded by TKT, on chromosome 3p21. Our series includes three families with a total of five affected individuals, ranging in age from 4 to 25 years. Two families of Ashkenazi Jewish ancestry were homozygous for an 18 base pair in-frame insertion in TKT. The third family was compound heterozygous for nonsense and missense variants in TKT. All affected individuals had short stature and were developmentally delayed. Congenital heart defects were noted in four of the five affected individuals, and there was a history of chronic diarrhea and cataracts in the older individuals with the homozygous 18 base pair insertion. Enzymatic testing confirmed significantly reduced transketolase activity. Elevated urinary excretion of erythritol, arabitol, ribitol, and pent(ul)ose-5-phosphates was detected, as well as elevated amounts of erythritol, arabitol, and ribitol in the plasma of affected individuals. Transketolase deficiency reduces NADPH synthesis and nucleic acid synthesis and cell division and could explain the problems with growth. NADPH is also critical for maintaining cerebral glutathione, which might contribute to the neurodevelopmental delays. Transketolase deficiency is one of a growing list of inborn errors of metabolism in the non-oxidative part of the pentose phosphate pathway.


Assuntos
Deficiências do Desenvolvimento/etiologia , Nanismo/etiologia , Cardiopatias Congênitas/etiologia , Mutação/genética , Transcetolase/genética , Adulto , Criança , Pré-Escolar , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Nanismo/metabolismo , Nanismo/patologia , Feminino , Glutationa/metabolismo , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/patologia , Humanos , Masculino , NADP/metabolismo , Linhagem , Síndrome , Adulto Jovem
12.
Horm Res Paediatr ; 86(5): 342-348, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27245183

RESUMO

BACKGROUND: Acromicric dysplasia (AD) and geleophysic dysplasia 2 (GD2) belong to the category of acromelic dysplasia syndromes, consisting of severe short stature, short hands and feet and skin thickening. Both can result from missense mutations in the transforming growth factor beta 5 domain of the fibrillin-1 gene (FBN1). METHODS: Two patients (P1 age 10, and P2 age 7) from unrelated families presented to their endocrinologist with severe short stature (approx. -4 SDS). They were otherwise asymptomatic and only had mild facial dysmorphisms. Extensive endocrine work-up did not reveal an underlying etiology. Exome sequencing was performed in each family. RESULTS: Exome sequencing identified the presence of the same heterozygous missense variant c.C5183T (p.Ala1728Val) in the FBN1 gene in both P1 and P2. This variant was previously reported in a patient with GD2 and associated cardiac valvulopathy and hepatomegaly. Detailed clinical re-examination, cardiac and skeletal imaging did not reveal any abnormalities in P1 or P2 other than mild hip dysplasia. CONCLUSION: This report broadens the phenotypic spectrum of growth disorders associated with FBN1 mutations. Identical mutations give rise to a wide phenotypic spectrum, ranging from isolated short stature to a more classic picture of GD2 with cardiac involvement, distinct facial dysmorphisms and various skeletal anomalies.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Exoma , Fibrilina-1/genética , Transtornos do Crescimento/genética , Deformidades Congênitas dos Membros/genética , Mutação de Sentido Incorreto , Adulto , Substituição de Aminoácidos , Doenças do Desenvolvimento Ósseo/patologia , Criança , Pré-Escolar , Feminino , Transtornos do Crescimento/patologia , Humanos , Lactente , Deformidades Congênitas dos Membros/patologia , Masculino
13.
Horm Res Paediatr ; 85(5): 358-62, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26789720

RESUMO

BACKGROUND: ACP5 deficiency is known to cause spondyloenchondrodysplasia (SPENCD), which is characterized by various autoimmune and neurological symptoms in addition to short stature. METHODS: Two siblings from a consanguineous Turkish family, a girl aged 13 years (P1) and a boy aged 8 years (P2), presented to their endocrinologist with progressive growth failure and severe short stature (-5 SDS). They had no comorbid conditions and, on physical examination, there were no signs of an overt skeletal dysplasia with normal appearance of extremities. Other than a low baseline IGF-1, extensive laboratory workup, including growth hormone stimulation and IGF-1 generation tests, was normal. Exome sequencing was performed. RESULTS: Exome sequencing identified the presence of a homozygous frameshift mutation (p.Ser258Trpfs*39) in ACP5 in both siblings, which was confirmed by Sanger sequencing. This specific mutation has previously been described in patients with SPENCD. Additional workup in the two siblings showed distinct features of skeletal dysplasia on X-rays consistent with SPENCD, but none of the common autoimmune or neurological abnormalities associated with this condition. CONCLUSION: Severe short stature can be the only presenting sign of ACP5 deficiency and the latter could therefore be considered as a rare cause in the differential diagnosis of severe, proportionate growth failure.


Assuntos
Doenças Autoimunes/genética , Mutação da Fase de Leitura , Transtornos do Crescimento/genética , Osteocondrodisplasias/genética , Irmãos , Fosfatase Ácida Resistente a Tartarato/deficiência , Adolescente , Doenças Autoimunes/fisiopatologia , Criança , Feminino , Transtornos do Crescimento/fisiopatologia , Humanos , Masculino , Osteocondrodisplasias/fisiopatologia , Turquia
15.
J Clin Endocrinol Metab ; 100(5): E789-98, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25742519

RESUMO

CONTEXT: Severe short stature can be caused by defects in numerous biological processes including defects in IGF-1 signaling, centromere function, cell cycle control, and DNA damage repair. Many syndromic causes of short stature are associated with medical comorbidities including hypogonadism and microcephaly. OBJECTIVE: To identify an underlying genetic etiology in two siblings with severe short stature and gonadal failure. DESIGN: Clinical phenotyping, genetic analysis, complemented by in vitro functional studies of the candidate gene. SETTING: An academic pediatric endocrinology clinic. PATIENTS OR OTHER PARTICIPANTS: Two adult siblings (male patient [P1] and female patient 2 [P2]) presented with a history of severe postnatal growth failure (adult heights: P1, -6.8 SD score; P2, -4 SD score), microcephaly, primary gonadal failure, and early-onset metabolic syndrome in late adolescence. In addition, P2 developed a malignant gastrointestinal stromal tumor at age 28. INTERVENTION(S): Single nucleotide polymorphism microarray and exome sequencing. RESULTS: Combined microarray analysis and whole exome sequencing of the two affected siblings and one unaffected sister identified a homozygous variant in XRCC4 as the probable candidate variant. Sanger sequencing and mRNA studies revealed a splice variant resulting in an in-frame deletion of 23 amino acids. Primary fibroblasts (P1) showed a DNA damage repair defect. CONCLUSIONS: In this study we have identified a novel pathogenic variant in XRCC4, a gene that plays a critical role in non-homologous end-joining DNA repair. This finding expands the spectrum of DNA damage repair syndromes to include XRCC4 deficiency causing severe postnatal growth failure, microcephaly, gonadal failure, metabolic syndrome, and possibly tumor predisposition.


Assuntos
Estatura/genética , Proteínas de Ligação a DNA/genética , Hipogonadismo/genética , Síndrome Metabólica/genética , Polimorfismo de Nucleotídeo Único , Adulto , Exoma , Feminino , Humanos , Masculino , Mutação , Irmãos
16.
J Clin Endocrinol Metab ; 99(10): E2117-22, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25057881

RESUMO

CONTEXT: CDKN1C, a cyclin-dependent kinase inhibitor and negative regulator of cellular proliferation, is paternally imprinted and has been shown to regulate ß-cell proliferation. CDKN1C mutations are associated with growth disorders, including Beckwith-Wiedemann syndrome and IMAGe syndrome. OBJECTIVE: To investigate the genetic basis for a familial disorder characterized by intrauterine growth restriction, short stature, and early-adulthood-onset diabetes. DESIGN, SETTING, AND PARTICIPANTS: Genomic DNA samples (15 affected and 26 unaffected from a six-generation pedigree) were analyzed by genome-wide single nucleotide polymorphism arrays, whole exome and Sanger sequencing, and multiplex ligation-dependent probe amplification. MAIN OUTCOME MEASURE(S): Subjects were assessed for height, weight, adrenal gland size, ACTH, diabetes status, and testis volume. Linkage and sequence analyses were performed, and the identified genetic variant was functionally evaluated in reconstitution studies. RESULTS: The pedigree followed a paternally imprinted pattern of inheritance, and genetic linkage analysis identified a single significant 2.6-megabase locus on chromosome 11p15, within the imprinting center region 2. Multiplex ligation-dependent probe amplification did not detect copy number variants or methylation abnormalities. Whole exome sequencing revealed a single novel variant in the proliferating cell nuclear antigen-binding region of CDKN1C (c.842G>T, p.R281I) that co-segregated with affected status and, unlike variants found in IMAGe, did not entirely abrogate proliferating cell nuclear antigen binding. Clinical assessments revealed that affected individuals had low testicular volume but normal adrenal function. CONCLUSIONS: We report a novel CDKN1C mutation associated with features of IMAGe syndrome, but without adrenal insufficiency or metaphyseal dysplasia, and characterized by early-adulthood-onset diabetes. Our data expand the range of phenotypes observed with CDKN1C defects and suggest that CDKN1C mutations may represent a novel monogenic form of diabetes.


Assuntos
Insuficiência Adrenal/genética , Síndrome de Beckwith-Wiedemann/genética , Inibidor de Quinase Dependente de Ciclina p57/genética , Diabetes Mellitus Tipo 2/genética , Retardo do Crescimento Fetal/genética , Estudo de Associação Genômica Ampla , Osteocondrodisplasias/genética , Anormalidades Urogenitais/genética , Glândulas Suprarrenais/patologia , Glândulas Suprarrenais/fisiologia , Insuficiência Adrenal/patologia , Hormônio Adrenocorticotrópico/sangue , Adulto , Idade de Início , Síndrome de Beckwith-Wiedemann/patologia , Estatura/genética , Peso Corporal/genética , Diabetes Mellitus Tipo 2/patologia , Feminino , Retardo do Crescimento Fetal/patologia , Ligação Genética , Variação Genética , Humanos , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Masculino , Tamanho do Órgão/genética , Osteocondrodisplasias/patologia , Linhagem , Fenótipo , Testículo/patologia , Testículo/fisiologia , Anormalidades Urogenitais/patologia
17.
J Clin Endocrinol Metab ; 98(8): E1428-37, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23771920

RESUMO

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.


Assuntos
Estatura/genética , Transtornos do Crescimento/genética , Mutação , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Variação Genética , Hormônio do Crescimento Humano/deficiência , Humanos , Masculino , Síndrome de Noonan/genética , Osteocondrodisplasias/genética , Estudos Prospectivos , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Receptor IGF Tipo 1/genética , Análise de Sequência de DNA , Adulto Jovem
18.
Clin Immunol ; 148(2): 227-36, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23773921

RESUMO

STAT5A and STAT5B are highly homologous proteins whose distinctive roles in human immunity remain unclear. However, STAT5A sufficiency cannot compensate for STAT5B defects, and human STAT5B deficiency, a rare autosomal recessive primary immunodeficiency, is characterized by chronic lung disease, growth failure and autoimmunity associated with regulatory T cell (Treg) reduction. We therefore hypothesized that STAT5A and STAT5B play unique roles in CD4(+) T cells. Upon knocking down STAT5A or STAT5B in human primary T cells, we found differentially regulated expression of FOXP3 and IL-2R in STAT5B knockdown T cells and down-regulated Bcl-X only in STAT5A knockdown T cells. Functional ex vivo studies in homozygous STAT5B-deficient patients showed reduced FOXP3 expression with impaired regulatory function of STAT5B-null Treg cells, also of increased memory phenotype. These results indicate that STAT5B and STAT5A act partly as non-redundant transcription factors and that STAT5B is more critical for Treg maintenance and function in humans.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fator de Transcrição STAT5/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Adolescente , Adulto , Doenças Autoimunes/genética , Doenças Autoimunes/metabolismo , Células Cultivadas , Criança , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/metabolismo , Fator de Transcrição STAT5/genética , Linfócitos T Reguladores/fisiologia , Proteínas Supressoras de Tumor/genética , Adulto Jovem , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
19.
J Clin Endocrinol Metab ; 97(11): E2140-51, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22933543

RESUMO

CONTEXT: Microcephalic primordial dwarfism (MPD) is a rare, severe form of human growth failure in which growth restriction is evident in utero and continues into postnatal life. Single causative gene defects have been identified in a number of patients with MPD, and all involve genes fundamental to cellular processes including centrosome functions. OBJECTIVE: The objective of the study was to find the genetic etiology of a novel presentation of MPD. DESIGN: The design of the study was whole-exome sequencing performed on two affected sisters in a single family. Molecular and functional studies of a candidate gene were performed using patient-derived primary fibroblasts and a zebrafish morpholino oligonucleotides knockdown model. PATIENTS: Two sisters presented with a novel subtype of MPD, including severe intellectual disabilities. MAIN OUTCOME MEASURES: NIN, encoding Ninein, a centrosomal protein critically involved in asymmetric cell division, was identified as a candidate gene, and functional impacts in fibroblasts and zebrafish were studied. RESULTS: From 34,606 genomic variants, two very rare missense variants in NIN were identified. Both probands were compound heterozygotes. In the zebrafish, ninein knockdown led to specific and novel defects in the specification and morphogenesis of the anterior neuroectoderm, resulting in a deformity of the developing cranium with a small, squared skull highly reminiscent of the human phenotype. CONCLUSION: We identified a novel clinical subtype of MPD in two sisters who have rare variants in NIN. We show, for the first time, that reduction of ninein function in the developing zebrafish leads to specific deficiencies of brain and skull development, offering a developmental basis for the myriad phenotypes in our patients.


Assuntos
Proteínas do Citoesqueleto/genética , Nanismo/genética , Deficiência Intelectual/genética , Microcefalia/genética , Proteínas Nucleares/genética , Adolescente , Feminino , Humanos , Mutação de Sentido Incorreto , Adulto Jovem
20.
J Clin Endocrinol Metab ; 97(5): E830-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22419735

RESUMO

CONTEXT: Signal transducer and activator of transcription 5b (STAT5b) deficiency, first reported in a patient who carried a p.Ala630Pro missense mutation in the Src homology 2 (SH2) domain, results in a rare clinical condition of GH insensitivity (GHI), IGF-I deficiency (IGFD), and severe immune dysregulation manifesting as progressive worsening of pulmonary function. PATIENT: The new patient presented with severe cutaneous eczema, episodic infections in the first years of life, and autoimmune thyroiditis. Immunological evaluation revealed T lymphopenia, but severe pulmonary symptoms were notably absent. She concomitantly exhibited pronounced growth failure, reaching an adult height of 124.7 cm [-5.90 SD score (SDS)]. Endocrine evaluations (normal provocative GH tests; low serum IGF-I, -3.7 SDS, and IGF-binding protein-3, -4.5 SDS) were consistent with GHI and IGFD. RESULTS: Analysis of the STAT5B gene revealed a novel homozygous missense mutation, p.Phe646Ser, located within the ßD' strand of the SH2 domain. Reconstitution studies demonstrated expression of the p.Phe646Ser variant was less robust than wild type but, in contrast to the previously described STAT5B p.Ala630Pro SH2 mutation, could be phosphorylated in response to GH and interferon-γ. The phosphorylated p.Phe646Ser, however, could not drive transcription. CONCLUSION: A novel STAT5B p.Phe646Ser mutation has been identified in a patient with clinical characteristics of STAT5b deficiency. Only the second STAT5B missense mutation identified, its lack of transcriptional activities despite GH-induced phosphorylation, confirms the crucial role of STAT5b for regulating the expression of IGF1 and provides insights into the importance of the SH2 ßD' strand for full STAT5b transcriptional activities. Whether the phosphorylated p.Phe646Ser variant retained functions that prevented pulmonary distress remains unresolved.


Assuntos
Nanismo Hipofisário/genética , Doenças do Sistema Imunitário/genética , Fator de Crescimento Insulin-Like I/deficiência , Mutação de Sentido Incorreto , Fator de Transcrição STAT5/genética , Tireoidite Autoimune/genética , Análise Mutacional de DNA , Feminino , Humanos , Fator de Crescimento Insulin-Like I/genética , Pneumopatias/genética , Adulto Jovem , Domínios de Homologia de src/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA