Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
3.
Eur J Med Genet ; 64(10): 104295, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34371190

RESUMO

Defects in the PIEZO1 gene cause lymphatic dysplasia in an autosomal recessive manner, mostly by loss-of-function variants. Moreover, since 2019, the role of PIEZO1 in bone formation has been established, but there have been no PIEZO1-related cases presenting definite skeletal involvement to date. A 21-year-old male with primary lymphatic dysplasia had some other distinctive clinical features, including multiple fracture history during infancy, thoracolumbar scoliosis, short stature, and left-sided facial bone hypoplasia. We analyzed the whole exome of the patient and found two novel pathogenic variants of PIEZO1 in trans: a 93.7 kb heterozygous deletion (chr16:88,782,477-88,876,207; exon 1-50) and c.2858G>A (p.Arg953His). Sanger sequencing validated the deletion with breakpoints, and each variant was inherited from a different parent. This study presented an extremely rare case of a patient with lymphatic dysplasia caused by compound heterozygous variants of PIEZO1, along with additional clinical manifestations including several skeletal phenotypes.


Assuntos
Anormalidades Craniofaciais/genética , Fraturas Ósseas/genética , Canais Iônicos/genética , Linfangiectasia Intestinal/genética , Linfedema/genética , Mutação , Fenótipo , Escoliose/genética , Anormalidades Craniofaciais/patologia , Fraturas Ósseas/patologia , Heterozigoto , Humanos , Linfangiectasia Intestinal/patologia , Linfedema/patologia , Masculino , Escoliose/patologia , Adulto Jovem
4.
ScientificWorldJournal ; 2021: 6642626, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34234628

RESUMO

Hennekam lymphangiectasia-lymphedema syndrome has been linked to single-nucleotide polymorphisms in the CCBE1 (collagen and calcium-binding EGF domains 1) gene. Several bioinformatics methods were used to find the most dangerous nsSNPs that could affect CCBE1 structure and function. Using state-of-the-art in silico tools, this study examined the most pathogenic nonsynonymous single-nucleotide polymorphisms (nsSNPs) that disrupt the CCBE1 protein and extracellular matrix remodeling and migration. Our results indicate that seven nsSNPs, rs115982879, rs149792489, rs374941368, rs121908254, rs149531418, rs121908251, and rs372499913, are deleterious in the CCBE1 gene, four (G330E, C102S, C174R, and G107D) of which are the highly deleterious, two of them (G330E and G107D) have never been seen reported in the context of Hennekam syndrome. Twelve missense SNPs, rs199902030, rs267605221, rs37517418, rs80008675, rs116596858, rs116675104, rs121908252, rs147974432, rs147681552, rs192224843, rs139059968, and rs148498685, are found to revert into stop codons. Structural homology-based methods and sequence homology-based tools revealed that 8.8% of the nsSNPs are pathogenic. SIFT, PolyPhen2, M-CAP, CADD, FATHMM-MKL, DANN, PANTHER, Mutation Taster, LRT, and SNAP2 had a significant score for identifying deleterious nsSNPs. The importance of rs374941368 and rs200149541 in the prediction of post-translation changes was highlighted because it impacts a possible phosphorylation site. Gene-gene interactions revealed CCBE1's association with other genes, showing its role in a number of pathways and coexpressions. The top 16 deleterious nsSNPs found in this research should be investigated further in the future while researching diseases caused CCBE1 gene specifically HS. The FT web server predicted amino acid residues involved in the ligand-binding site of the CCBE1 protein, and two of the substitutions (R167W and T153N) were found to be involved. These highly deleterious nsSNPs can be used as marker pathogenic variants in the mutational diagnosis of the HS syndrome, and this research also offers potential insights that will aid in the development of precision medicines. CCBE1 proteins from Hennekam syndrome patients should be tested in animal models for this purpose.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Anormalidades Craniofaciais/genética , Linfangiectasia Intestinal/genética , Linfedema/genética , Mutação de Sentido Incorreto/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Supressoras de Tumor/genética , Biologia Computacional , Previsões , Humanos
6.
Eur J Med Genet ; 64(1): 104106, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33227434

RESUMO

Generalized lymphatic dysplasia (GLD), characterized by lymphedema, lymphangiectasias, chylothorax, effusions, represents a recognized cause of fetal hydrops. We describe for the first time recurrent pregnancies showing different ultrasound presentations of lymphatic dysplasia. The first fetus displayed diffuse subcutaneous cysts and septations while the second one presented fetal hydrops. Exome sequencing results at 18 gestational weeks in the second pregnancy showed compound heterozygosity for two novel PIEZO1 variants, afterwards detected also in the first fetus and in the heterozygous parents. Both ultrasound and genetic findings expand the current knowledge of PIEZO1-related GLD. We suggest exome sequencing in hydropic fetuses with normal cytogenetics and in pregnancies with recurrent hydrops/lymphatic dysplasia.


Assuntos
Anormalidades Craniofaciais/genética , Testes Genéticos , Hidropisia Fetal/genética , Canais Iônicos/genética , Linfangiectasia Intestinal/genética , Linfedema/genética , Ultrassonografia Pré-Natal , Adulto , Anormalidades Craniofaciais/diagnóstico por imagem , Anormalidades Craniofaciais/patologia , Feminino , Humanos , Hidropisia Fetal/diagnóstico por imagem , Hidropisia Fetal/patologia , Linfangiectasia Intestinal/diagnóstico por imagem , Linfangiectasia Intestinal/patologia , Linfedema/diagnóstico por imagem , Linfedema/patologia , Gravidez , Sequenciamento do Exoma
7.
Am J Med Genet A ; 182(9): 2152-2160, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32618121

RESUMO

The etiology of nonimmune hydrops fetalis is extensive and includes genetic disorders. We describe a term-born female neonate with late onset extensive nonimmune hydrops, that is, polyhydramnios, edema, and congenital bilateral chylothorax. This newborn was successfully treated with repetitive thoracocentesis, total parenteral feeding, octreotide intravenously and finally surgical pleurodesis and corticosteroids. A genetic cause seemed plausible as the maternal history revealed a fatal nonimmune hydrops fetalis. A homozygous truncating variant in GDF2 (c.451C>T, p.(Arg151*)) was detected with exome sequencing. Genetic analysis of tissue obtained from the deceased fetal sibling revealed the same homozygous variant. The parents and two healthy siblings were heterozygous for the GDF2 variant. Skin and lung biopsies in the index patient, as well as the revised lung biopsy of the deceased fetal sibling, showed lymphatic dysplasia and lymphangiectasia. To the best of our knowledge, this is the first report of an association between a homozygous variant in GDF2 with lymphatic dysplasia, hydrothorax and nonimmune hydrops fetalis.


Assuntos
Anormalidades Craniofaciais/genética , Fator 2 de Diferenciação de Crescimento/genética , Hidropisia Fetal/genética , Linfangiectasia Intestinal/genética , Linfedema/genética , Poli-Hidrâmnios/genética , Anormalidades Craniofaciais/diagnóstico , Anormalidades Craniofaciais/patologia , Feminino , Homozigoto , Humanos , Hidropisia Fetal/diagnóstico , Hidropisia Fetal/patologia , Recém-Nascido , Linfangiectasia Intestinal/diagnóstico , Linfangiectasia Intestinal/patologia , Linfedema/diagnóstico , Linfedema/patologia , Poli-Hidrâmnios/diagnóstico , Poli-Hidrâmnios/patologia , Gravidez , Toracentese , Ultrassonografia Pré-Natal , Sequenciamento do Exoma
8.
Medicine (Baltimore) ; 99(27): e20995, 2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32629717

RESUMO

RATIONAL: Intestinal lymphangiectasia (IL) is a rare disease characterized by dilatation and rupture of intestinal lymphatic channels leading to protein-losing enteropathy. IL is classified as primary and secondary types. PATIENT CONCERNS: A 3-month-old girl born at term from vaginal delivery with an APGAR score of 10/10 and birth weight of 4.310 g (>97° percentile) was admitted to our hospital because of increasing abdominal tenderness and diarrhea. At first examination, she presented an abdominal circumference of 60 cm, edema of the lower extremities and vulva, and facial dysmorphisms (hypertelorism, flat nasal bridge, flat mid-face). DIAGNOSIS: Once admitted, ultrasonography showed a large amount of ascites, while blood laboratory investigations revealed severe hypoproteinemia, hypoalbuminemia and hypogammaglobulinemia. Lymphoscintigraphy with 99m-Tc-nanocolloid demonstrated abnormal leakage of the tracer in the abdomen as evidence of IL. To detect a possible secondary, exams were performed and demonstrated positive antibody titres for CMV-IgM and IgG in blood and CMV-DNA positivity in blood, urine, saliva, maternal milk, and gastric and duodenal biopsies. Genetic investigations identified the genomic variant c.472C>T of the CCBE1 gene, coding for a protein variant (p.Arg158Cys), in homozygosity. INTERVENTIONS: Total parenteral nutrition was started and continued for a total of 18 days, then gradually bridged by enteral nutrition with a special formula. In addition, antiviral therapy for CMV infection was added first with intravenous ganciclovir for 14 days, resulting in the disappearance of blood viral load after 7 days of therapy and then with valganciclovir per os for another 30 days. OUTCOMES: The clinical course of the child gradually improved. A few days after starting treatments, lower extremities and vulvar edema disappeared, and abdominal circumference gradually decreased to a stable value of 38 cm, without any ultrasonographic signs of ascites left. Moreover, serum albumin and IgG rose to normal values after 3 months (4.3 g/dL and 501 mg/dL, respectively). LESSONS: This case suggests that in presence of IL both primary and secondary causes should be evaluated. On the other hand, genetic diagnosis is crucial not only for diagnosis but also for prognosis in HS. Life expectancy and quality could deeply vary among different gene mutations and protein variants of the same gene. Further studies and case reports are needed to better understand the clinical meaning of these genetic results and the role of CMV as trigger of IL.


Assuntos
Anormalidades Craniofaciais/genética , Linfangiectasia Intestinal/genética , Linfedema/genética , Proteínas de Ligação ao Cálcio/genética , Anormalidades Craniofaciais/diagnóstico , Feminino , Humanos , Lactente , Linfangiectasia Intestinal/diagnóstico , Linfedema/diagnóstico , Mutação , Proteínas Supressoras de Tumor/genética
9.
Am J Med Genet A ; 182(1): 189-194, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31633297

RESUMO

Hennekam lymphangiectasia-lymphedema syndrome is an autosomal recessive disorder characterized by congenital lymphedema, intestinal lymphangiectasia, facial dysmorphism, and variable intellectual disability. Known disease genes include CCBE1, FAT4, and ADAMTS3. In a patient with clinically diagnosed Hennekam syndrome but without mutations or copy-number changes in the three known disease genes, we identified a homozygous single-exon deletion affecting FBXL7. Specifically, exon 3, which encodes the F-box domain and several leucine-rich repeats of FBXL7, is eliminated. Our analyses of databases representing >100,000 control individuals failed to identify biallelic loss-of-function variants in FBXL7. Published studies in Drosophila indicate Fbxl7 interacts with Fat, of which human FAT4 is an ortholog, and mutation of either gene yields similar morphological consequences. These data suggest that FBXL7 may be the fourth gene for Hennekam syndrome, acting via a shared pathway with FAT4.


Assuntos
Anormalidades Craniofaciais/genética , Proteínas F-Box/genética , Predisposição Genética para Doença , Linfangiectasia Intestinal/genética , Linfedema/genética , Proteínas ADAMTS/genética , Alelos , Animais , Pré-Escolar , Anormalidades Craniofaciais/complicações , Anormalidades Craniofaciais/patologia , Drosophila melanogaster/genética , Genótipo , Homozigoto , Humanos , Linfangiectasia Intestinal/complicações , Linfangiectasia Intestinal/patologia , Linfedema/complicações , Linfedema/patologia , Masculino , Técnicas de Diagnóstico Molecular/métodos , Mutação/genética , Linhagem , Fenótipo , Pró-Colágeno N-Endopeptidase/genética
10.
Immunol Rev ; 287(1): 20-32, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30565236

RESUMO

Primary intestinal lymphangiectasia (PIL) or Waldmann's disease was described in 1961 as an important cause of protein-losing enteropathy (PLE). PIL can be the sole finding in rare individuals or occur as part of a multisystemic genetic syndrome. Although genetic etiologies of many lymphatic dysplasia syndromes associated with PIL have been identified, the pathogenesis of isolated PIL (with no associated syndromic features) remains unknown. Familial cases and occurrence at birth suggest genetic etiologies in certain cases. Recently, CD55 deficiency with hyperactivation of complement, angiopathic thrombosis, and PLE (the CHAPLE syndrome) has been identified as a monogenic form of PIL. Surprisingly, loss of CD55, a key regulator of complement system leads to a predominantly gut condition. Similarly to other complement disorders, namely paroxysmal nocturnal and hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS), CHAPLE disease involves pathogenic cross-activation of the coagulation system, predisposing individuals to severe thrombosis. The observation that complement system is overly active in CHAPLE disease introduced a novel concept into the management of PLE; anti-complement therapy. While CD55 deficiency constitutes a treatable subgroup in the larger pool of patients with isolated PIL, the etiology remains to be identified in the remaining patients with intact CD55.


Assuntos
Antígenos CD55/genética , Proteínas do Sistema Complemento/metabolismo , Linfangiectasia Intestinal/genética , Linfedema/genética , Coagulação Sanguínea/genética , Reações Cruzadas , Humanos , Enteropatias Perdedoras de Proteínas , Síndrome
11.
Am J Med Genet A ; 176(12): 2858-2861, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30450763

RESUMO

Hennekam lymphangiectasia-lymphedema syndrome (HKLLS) is a genetically heterogeneous lymphatic dysplasia with characteristic of facial dysmorphism, neurocognitive impairments, and abnormalities of the pericardium, intestinal tract, and extremities. It is an autosomal recessive condition caused by biallelic mutations in CCBE1 (collagen- and calcium-binding epidermal growth factor domain-containing protein 1) (HKLLS1; OMIM 235510) or FAT4 (HKLLS2; OMIM 616006). CCBE1 acts via ADAMTS3 (a disintegrin and metalloprotease with thrombospondin motifs-3 protease) to enhance vascular endothelial growth factor C signaling. There is report of one family supporting mutations in ADAMTS3 as causative for the phenotype labeled as HKLLS3. Here, we report an additional case of HKLLS that appears to be associated with homozygous nonsense mutation of ADAMTS3.


Assuntos
Proteínas ADAMTS/genética , Anormalidades Craniofaciais/diagnóstico , Anormalidades Craniofaciais/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Mutação com Perda de Função , Linfangiectasia Intestinal/diagnóstico , Linfangiectasia Intestinal/genética , Linfedema/diagnóstico , Linfedema/genética , Pró-Colágeno N-Endopeptidase/genética , Alelos , Biópsia , Estudos de Associação Genética/métodos , Genótipo , Humanos , Recém-Nascido , Masculino , Fenótipo , Sequenciamento do Exoma
12.
J Exp Med ; 215(9): 2339-2353, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-30115739

RESUMO

We report the first case of nonimmune hydrops fetalis (NIHF) associated with a recessive, in-frame deletion of V205 in the G protein-coupled receptor, Calcitonin Receptor-Like Receptor (hCALCRL). Homozygosity results in fetal demise from hydrops fetalis, while heterozygosity in females is associated with spontaneous miscarriage and subfertility. Using molecular dynamic modeling and in vitro biochemical assays, we show that the hCLR(V205del) mutant results in misfolding of the first extracellular loop, reducing association with its requisite receptor chaperone, receptor activity modifying protein (RAMP), translocation to the plasma membrane and signaling. Using three independent genetic mouse models we establish that the adrenomedullin-CLR-RAMP2 axis is both necessary and sufficient for driving lymphatic vascular proliferation. Genetic ablation of either lymphatic endothelial Calcrl or nonendothelial Ramp2 leads to severe NIHF with embryonic demise and placental pathologies, similar to that observed in humans. Our results highlight a novel candidate gene for human congenital NIHF and provide structure-function insights of this signaling axis for human physiology.


Assuntos
Sequência de Aminoácidos , Proteína Semelhante a Receptor de Calcitonina , Anormalidades Craniofaciais , Hidropisia Fetal , Linfangiectasia Intestinal , Linfedema , Camundongos Transgênicos , Deleção de Sequência , Animais , Proteína Semelhante a Receptor de Calcitonina/genética , Proteína Semelhante a Receptor de Calcitonina/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Heterozigoto , Homozigoto , Humanos , Hidropisia Fetal/genética , Hidropisia Fetal/metabolismo , Hidropisia Fetal/patologia , Linfangiectasia Intestinal/genética , Linfangiectasia Intestinal/metabolismo , Linfangiectasia Intestinal/patologia , Linfedema/genética , Linfedema/metabolismo , Linfedema/patologia , Masculino , Camundongos , Placenta , Gravidez
13.
Clin Med (Lond) ; 17(6): 552-557, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29196357

RESUMO

Lymphoedema is the build-up of lymphatic fluid leading to swelling in the tissues. Most commonly it affects the peripheries. Diagnosis is based on clinical assessment and imaging with lymphoscintigraphy. Treatment is supportive with compression garments, massage, good skin hygiene and prompt use of antibiotics to avoid the complication of cellulitis. Most commonly, lymphoedema occurs as a result of damage to the lymphatic system following surgery, trauma, radiation or infection. However, it can be primary, often associated with a genetic defect that causes disruption to the development of the lymphatic system. Common genetic conditions associated with lymphoedema include Turner syndrome and Noonan syndrome; however, there are numerous others that can be classified based on their clinical presentation and associated features. Herein we discuss how to diagnose and classify the known primary lymphoedema conditions and how best to investigate and manage this group of patients.


Assuntos
Linfedema/diagnóstico , Idade de Início , Anormalidades Craniofaciais/classificação , Anormalidades Craniofaciais/diagnóstico , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/fisiopatologia , Testes Genéticos , Humanos , Canais Iônicos/genética , Linfangiectasia Intestinal/classificação , Linfangiectasia Intestinal/diagnóstico , Linfangiectasia Intestinal/genética , Linfangiectasia Intestinal/fisiopatologia , Linfedema/classificação , Linfedema/genética , Linfedema/fisiopatologia , Síndrome de Noonan/genética , Síndrome de Noonan/fisiopatologia , Receptor EphB4/genética , Síndrome de Turner/genética , Síndrome de Turner/fisiopatologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética
14.
Arterioscler Thromb Vasc Biol ; 37(9): 1732-1735, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28705793

RESUMO

OBJECTIVE: The purpose of this study was to investigate the role of Fat4 and Dachsous1 signaling in the lymphatic vasculature. APPROACH AND RESULTS: Phenotypic analysis of the lymphatic vasculature was performed in mice lacking functional Fat4 or Dachsous1. The overall architecture of lymphatic vasculature is unaltered, yet both genes are specifically required for lymphatic valve morphogenesis. Valve endothelial cells (Prox1high [prospero homeobox protein 1] cells) are disoriented and failed to form proper valve leaflets. Using Lifeact-GFP (green fluorescent protein) mice, we revealed that valve endothelial cells display prominent actin polymerization. Finally, we showed the polarized recruitment of Dachsous1 to membrane protrusions and cellular junctions of valve endothelial cells in vivo and in vitro. CONCLUSIONS: Our data demonstrate that Fat4 and Dachsous1 are critical regulators of valve morphogenesis. This study highlights that valve defects may contribute to lymphedema in Hennekam syndrome caused by Fat4 mutations.


Assuntos
Caderinas/metabolismo , Movimento Celular , Células Endoteliais/metabolismo , Endotélio Linfático/metabolismo , Linfangiogênese , Vasos Linfáticos/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Caderinas/deficiência , Caderinas/genética , Células Cultivadas , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Células Endoteliais/patologia , Endotélio Linfático/patologia , Imunofluorescência , Predisposição Genética para Doença , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Linfangiectasia Intestinal/genética , Linfangiectasia Intestinal/metabolismo , Linfangiectasia Intestinal/patologia , Vasos Linfáticos/patologia , Linfedema/genética , Linfedema/metabolismo , Linfedema/patologia , Camundongos Knockout , Mutação , Fenótipo , Multimerização Proteica , Transdução de Sinais , Transfecção , Proteínas Supressoras de Tumor/genética
16.
Am J Med Genet A ; 170(10): 2694-7, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27345729

RESUMO

Hennekam lymphangiectasia-lymphedema syndrome is an autosomal recessive disorder, with 25% of patients having mutations in CCBE1. We identified a family with two brothers presenting with primary lymphedema, and performed exome sequencing to determine the cause of their disease. Analysis of four family members showed that both affected brothers had the same rare compound heterozygous mutations in CCBE1. The presumed paternally inherited NM_133459.3:c.310G>A; p.(Asp104Asn), lies adjacent to other known pathogenic CCBE1 mutations, while the maternally inherited NM_133459.3:c.80T>C; p.(Leu27Pro) lies in the CCBE1 signal peptide, which has not previously been associated with disease. Functional analysis in a zebrafish model of lymphatic disease showed that both mutations lead to CCBE1 loss of function, confirming the pathogenicity of these variants and expanding the genotypic spectrum of lymphatic disorders. © 2016 Wiley Periodicals, Inc.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Anormalidades Craniofaciais/diagnóstico , Anormalidades Craniofaciais/genética , Genótipo , Linfangiectasia Intestinal/diagnóstico , Linfangiectasia Intestinal/genética , Linfedema/diagnóstico , Linfedema/genética , Mutação , Proteínas Supressoras de Tumor/genética , Alelos , Sequência de Aminoácidos , Animais , Linhagem Celular , Expressão Gênica , Frequência do Gene , Humanos , Recém-Nascido , Masculino , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Peixe-Zebra
17.
Am J Med Genet A ; 170(11): 2993-2997, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27214123

RESUMO

Protein-losing enteropathy is a primary or secondary manifestation of a group of conditions, and etiologies which are broadly divisible into those with mucosal injury on the basis of inflammatory and ulcerative conditions, mucosal injury without erosions or ulcerations, and lymphatic abnormalities. We describe the first case of protein-losing enteropathy in a pediatric patient, with severe skeletal dysplasia consistent with thanatophoric dysplasia type I and DNA analysis that revealed a c.1949A>T (p.Lys650Met) in exon 15 of the FGFR3 gene. She presented with protein-losing enteropathy in her 6th month. Post-mortem examination revealed lymphangiectasia in the small intestine. To our knowledge, this is the first report of intestinal lymphangiectasia as a complication of skeletal dysplasia resulting in severe protein-losing enteropathy. © 2016 Wiley Periodicals, Inc.


Assuntos
Linfangiectasia Intestinal/diagnóstico , Linfangiectasia Intestinal/genética , Mutação , Enteropatias Perdedoras de Proteínas/diagnóstico , Enteropatias Perdedoras de Proteínas/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/deficiência , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Displasia Tanatofórica/diagnóstico , Displasia Tanatofórica/genética , Alelos , Substituição de Aminoácidos , Autopsia , Encéfalo/patologia , Códon , Evolução Fatal , Feminino , Estudos de Associação Genética , Humanos , Recém-Nascido , Mucosa Intestinal/patologia , Pulmão/patologia , Imageamento por Ressonância Magnética , Fenótipo , Radiografia
18.
J Clin Immunol ; 36(1): 19-27, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26686525

RESUMO

Collagen and calcium-binding EGF domain-containing protein 1 (CCBE1) bi-allelic mutations have been associated with syndromes of widespread congenital lymphatic dysplasia, including Hennekam Syndrome (HS). HS is characterized by lymphedema, lymphangiectasia, and intellectual disability. CCBE1 encodes a putative extracellular matrix protein but the HS-causing mutations have not been studied biochemically. We report two HS siblings, born to consanguineous parents of Turkish ancestry, whose clinical phenotype also includes protein losing enteropathy, painful relapsing chylous ascites, and hypogammaglobulinemia. We identified by whole exome and Sanger sequencing the homozygous CCBE1 C174Y mutation in both siblings. This mutation had been previously reported in another HS kindred from the Netherlands. In over-expression studies, we found increased intracellular expression of all forms (monomers, dimers, trimers) of the CCBE1 C174Y mutant protein, by Western blot, despite mutant mRNA levels similar to wild-type (WT). In addition, we detected increased secretion of the mutant CCBE1 protein by ELISA. We further found the mutant and WT proteins to be evenly distributed in the cytoplasm, by immunofluorescence and confocal microscopy. Finally, we found a strong decrease of lymphatic vessels, with a corresponding diminished expression of CCBE1, by immunohistochemistry of the patients' intestinal biopsies. In contrast, mucosal blood vessels and muscularis mucosae showed normal CCBE1 staining. Our findings show that the mutant CCBE1 C174Y protein is not loss-of-function by loss-of-expression.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Anormalidades Craniofaciais/diagnóstico , Linfangiectasia Intestinal/diagnóstico , Linfedema/diagnóstico , Proteínas Supressoras de Tumor/metabolismo , Adulto , Proteínas de Ligação ao Cálcio/genética , Consanguinidade , Anormalidades Craniofaciais/genética , Análise Mutacional de DNA , Feminino , Regulação da Expressão Gênica/genética , Homozigoto , Humanos , Linfangiectasia Intestinal/genética , Linfedema/genética , Masculino , Mutação/genética , Linhagem , Irmãos , Proteínas Supressoras de Tumor/genética , Turquia , Adulto Jovem
19.
Nat Commun ; 6: 8085, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26333996

RESUMO

Generalized lymphatic dysplasia (GLD) is a rare form of primary lymphoedema characterized by a uniform, widespread lymphoedema affecting all segments of the body, with systemic involvement such as intestinal and/or pulmonary lymphangiectasia, pleural effusions, chylothoraces and/or pericardial effusions. This may present prenatally as non-immune hydrops. Here we report homozygous and compound heterozygous mutations in PIEZO1, resulting in an autosomal recessive form of GLD with a high incidence of non-immune hydrops fetalis and childhood onset of facial and four limb lymphoedema. Mutations in PIEZO1, which encodes a mechanically activated ion channel, have been reported with autosomal dominant dehydrated hereditary stomatocytosis and non-immune hydrops of unknown aetiology. Besides its role in red blood cells, our findings indicate that PIEZO1 is also involved in the development of lymphatic structures.


Assuntos
Anemia Hemolítica Congênita/genética , Anormalidades Craniofaciais/genética , Hidropisia Fetal/genética , Canais Iônicos/genética , Linfangiectasia Intestinal/genética , Linfedema/genética , Adolescente , Adulto , Western Blotting , Criança , Pré-Escolar , Anormalidades Craniofaciais/diagnóstico por imagem , Feminino , Heterozigoto , Humanos , Recém-Nascido , Linfangiectasia Intestinal/diagnóstico por imagem , Linfedema/diagnóstico por imagem , Linfocintigrafia , Masculino , Mutação , Análise de Sequência de DNA
20.
Am J Med Genet A ; 167A(9): 2209-12, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25943403

RESUMO

Primary intestinal lymphangiectasia (PIL) is a rare protein-losing enteropathy characterized by a congenital malformation of the lymphatic vessels of the small intestine causing insufficient drainage and leakage of lymph fluid. Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder characterized by benign hamartomas in multiple organ systems. While the lymphatic system has been implicated in TSC through lymphangioleiomyomatosis (LAM) and lymphedema, this paper reports the first case of PIL in TSC, a female patient with a TSC2 mutation. She developed persistent and significant abdominal distension with chronic diarrhea during her first year of life. Due to lack of treatment options and the involvement of the mTOR pathway in TSC, a trial of an mTOR inhibitor, rapamycin, was initiated. This treatment was highly effective, with improvement in clinical symptoms of PIL as well as abnormal laboratory values including VEGF-C, which was elevated to over seven times the normal upper limit before treatment. This case suggests that PIL is a rare manifestation of TSC, warranting the use of mTOR inhibitors in future studies.


Assuntos
Linfangiectasia Intestinal/tratamento farmacológico , Linfangiectasia Intestinal/genética , Linfedema/tratamento farmacológico , Linfedema/genética , Sirolimo/uso terapêutico , Esclerose Tuberosa/genética , Adulto , Feminino , Humanos , Mutação/genética , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA