Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38676400

RESUMO

Developmental and functional defects in the lymphatic system are responsible for primary lymphoedema (PL). PL is a chronic debilitating disease caused by increased accumulation of interstitial fluid, predisposing to inflammation, infections and fibrosis. There is no cure, only symptomatic treatment is available. Thirty-two genes or loci have been linked to PL, and another 22 are suggested, including Hepatocyte Growth Factor (HGF). We searched for HGF variants in 770 index patients from the Brussels PL cohort. We identified ten variants predicted to cause HGF loss-of-function (six nonsense, two frameshifts, and two splice-site changes; 1.3% of our cohort), and 14 missense variants predicted to be pathogenic in 17 families (2.21%). We studied co-segregation within families, mRNA stability for non-sense variants, and in vitro functional effects of the missense variants. Analyses of the mRNA of patient cells revealed degradation of the nonsense mutant allele. Reduced protein secretion was detected for nine of the 14 missense variants expressed in COS-7 cells. Stimulation of lymphatic endothelial cells with these 14 HGF variant proteins resulted in decreased activation of the downstream targets AKT and ERK1/2 for three of them. Clinically, HGF-associated PL was diverse, but predominantly bilateral in the lower limbs with onset varying from early childhood to adulthood. Finally, aggregation study in a second independent cohort underscored that rare likely pathogenic variants in HGF explain about 2% of PL. Therefore, HGF signalling seems crucial for lymphatic development and/or maintenance in human beings and HGF should be included in diagnostic genetic screens for PL.

2.
Hum Mol Genet ; 32(2): 276-289, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-35972810

RESUMO

Somatic activating Kirsten rat sarcoma viral oncogene homologue (KRAS) mutations have been reported in patients with arteriovenous malformations. By producing LSL-Kras (G12D); Cdh5 (PAC)-CreERT2 [iEC-Kras (G12D*)] mice, we hoped to activate KRAS within vascular endothelial cells (ECs) to generate an arteriovenous malformation mouse model. Neonatal mice were treated daily with tamoxifen from postnatal (PN) days 1-3. Mortality and phenotypes varied amongst iEC-Kras (G12D*) pups, with only 31.5% surviving at PN14. Phenotypes (focal lesions, vessel dilations) developed in a consistent manner, although with unpredictable severity within multiple soft tissues (such as the brain, liver, heart and brain). Overall, iEC-Kras (G12D*) pups developed significantly larger vascular lumen areas compared with control littermates, beginning at PN8. We subsequently tested whether the MEK inhibitor trametinib could effectively alleviate lesion progression. At PN16, iEC-Kras (G12D*) pup survival improved to 76.9%, and average vessel sizes were closer to controls than in untreated and vehicle-treated mutants. In addition, trametinib treatment helped normalize iEC-Kras (G12D*) vessel morphology in PN14 brains. Thus, trametinib could act as an effective therapy for KRAS-induced vascular malformations in patients.


Assuntos
Neoplasias Pancreáticas , Proteínas Proto-Oncogênicas p21(ras) , Camundongos , Animais , Proteínas Proto-Oncogênicas p21(ras)/genética , Neoplasias Pancreáticas/genética , Células Endoteliais/patologia , Modelos Animais de Doenças , Mutação
3.
Am J Med Genet A ; 194(6): e63551, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38321651

RESUMO

Capillary malformations (CMs) are the most common type of vascular anomalies, affecting around 0.3% of newborns. They are usually caused by somatic pathogenic variants in GNAQ or GNA11. PIK3CA and PIK3R1, part of the phosphoinositide 3-kinase-protein kinase B-mammalian target of rapamycin pathway, are mutated in fainter CMs such as diffuse CM with overgrowth and megalencephaly CM. In this study, we present two young patients with a CM-like phenotype associated with cerebral anomalies and severe epilepsy. Pathogenic variants in PIK3CA and PIK3R1, as well as GNAQ and GNA11, were absent in affected cutaneous tissue biopsies. Instead, we identified two somatic pathogenic variants in the AKT3 gene. Subsequent analysis of the DNA obtained from surgically resected brain tissue of one of the two patients confirmed the presence of the AKT3 variant. Focal cortical dysplasia was also detected in this patient. Genetic analysis thus facilitated workup to reach a precise diagnosis for these patients, associating the vascular anomaly with the neurological symptoms. This study underscores the importance of searching for additional signs and symptoms to guide the diagnostic workup, especially in cases with atypical vascular malformations. In addition, it strongly emphasizes the significance of genotype-phenotype correlation studies in guiding clinicians' informed decision-making regarding patient care.


Assuntos
Capilares , Epilepsia , Proteínas Proto-Oncogênicas c-akt , Telangiectasia , Malformações Vasculares , Feminino , Humanos , Recém-Nascido , Masculino , Capilares/anormalidades , Capilares/patologia , Epilepsia/genética , Epilepsia/patologia , Estudos de Associação Genética , Predisposição Genética para Doença , Mosaicismo , Mutação/genética , Fenótipo , Proteínas Proto-Oncogênicas c-akt/genética , Telangiectasia/genética , Telangiectasia/patologia , Telangiectasia/diagnóstico , Malformações Vasculares/genética , Malformações Vasculares/patologia , Malformações Vasculares/diagnóstico , Malformações Vasculares/complicações , Adolescente
4.
J Med Genet ; 60(1): 57-64, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-34876502

RESUMO

BACKGROUND: Hydrops fetalis, a pathological fluid accumulation in two or more body compartments, is aetiologically heterogeneous. We investigated a consanguineous family with recurrent pregnancy loss due to severe early-onset non-immune hydrops fetalis. METHODS AND RESULTS: Whole exome sequencing in four fetuses with hydrops fetalis revealed that they were homozygous for the angiopoietin-2 (ANGPT2) variant Chr8 (GRCh37/Hg19): 6385085T>C, NM_001147.2:c.557A>G. The substitution introduces a cryptic, exonic splice site predicted to result in loss of 10 nucleotides with subsequent shift in reading frame, leading to a premature stop codon. RNA analysis in the heterozygous parents demonstrated loss of detectable mutant allele, indicative of loss-of-function via nonsense-mediated mRNA decay. Serum ANGPT2 levels were reduced in the parents. In a pregnancy with a healthy, heterozygous child, transiently increased fetal nuchal translucency was noted. CONCLUSION: Pathogenic heterozygous ANGPT2 missense variants were recently shown to cause autosomal dominant primary lymphoedema. ANGPT2 is a ligand of the TIE1-TIE2 (tyrosine kinase with immunoglobulin-like and epidermal growth factor-like domains 1 and 2) pathway. It is critical to the formation and remodelling of blood and lymphatic vessels and is involved in vessel maintenance. ANGPT2 knockout mice die from generalised lymphatic dysfunction. We show here that a homozygous pathogenic variant causes loss-of-function and results in severe early-onset hydrops fetalis. This is the first report of an autosomal recessive ANGPT2-related disorder in humans.


Assuntos
Angiopoietina-2 , Hidropisia Fetal , Animais , Feminino , Humanos , Camundongos , Gravidez , Angiopoietina-2/genética , Códon sem Sentido/genética , Heterozigoto , Hidropisia Fetal/genética , Hidropisia Fetal/metabolismo , Mutação de Sentido Incorreto , Recém-Nascido
5.
J Med Genet ; 60(12): 1161-1168, 2023 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-37225411

RESUMO

BACKGROUND: Primary lymphoedema (PL) is a chronic, debilitating disease caused by developmental and functional defects of the lymphatic system. It is marked by an accumulation of interstitial fluid, fat and tissue fibrosis. There is no cure. More than 50 genes and genetic loci have been linked to PL. We sought to study systematically cell polarity signalling protein Cadherin Epidermal Growth Factor Laminin G Seven-pass G-type Receptor 1 (CELSR1) variants linked to PL. METHODS: We investigated 742 index patients from our PL cohort using exome sequencing. RESULTS: We identified nine variants predicted to cause CELSR1 loss of function. Four of them were tested for nonsense-mediated mRNA decay, but none was observed. Most of the truncated CELSR1 proteins would lack the transmembrane domain, if produced. The affected individuals had puberty/late-onset PL on lower extremities. The variants had a statistically significant difference in penetrance between female patients (87%) and male patients (20%). Eight variant carriers had a kidney anomaly, mostly in the form of ureteropelvic junction obstruction, which has not been associated with CELSR1 before. CELSR1 is located in the 22q13.3 deletion locus of the Phelan-McDermid syndrome. As variable renal defects are often seen in patients with the Phelan-McDermid syndrome, CELSR1 may be the long-sought gene for the renal defects. CONCLUSION: PL associated with a renal anomaly suggests a CELSR1-related cause.


Assuntos
Transtornos Cromossômicos , Linfedema , Feminino , Humanos , Masculino , Caderinas/genética , Caderinas/metabolismo , Deleção Cromossômica , Transtornos Cromossômicos/genética , Linfedema/genética
6.
Circ Res ; 129(1): 155-173, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34166070

RESUMO

Vascular and lymphatic malformations represent a challenge for clinicians. The identification of inherited and somatic mutations in important signaling pathways, including the PI3K (phosphoinositide 3-kinase)/AKT (protein kinase B)/mTOR (mammalian target of rapamycin), RAS (rat sarcoma)/RAF (rapidly accelerated fibrosarcoma)/MEK (mitogen-activated protein kinase kinase)/ERK (extracellular signal-regulated kinases), HGF (hepatocyte growth factor)/c-Met (hepatocyte growth factor receptor), and VEGF (vascular endothelial growth factor) A/VEGFR (vascular endothelial growth factor receptor) 2 cascades has led to the evaluation of tailored strategies with preexisting cancer drugs that interfere with these signaling pathways. The era of theranostics has started for the treatment of vascular anomalies. Registration: URL: https://www.clinicaltrialsregister.eu; Unique identifier: 2015-001703-32.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Vasos Sanguíneos/anormalidades , Vasos Sanguíneos/efeitos dos fármacos , Mutação , Neovascularização Fisiológica/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Malformações Vasculares/tratamento farmacológico , Malformações Vasculares/genética , Inibidores da Angiogênese/efeitos adversos , Animais , Vasos Sanguíneos/metabolismo , Predisposição Genética para Doença , Humanos , Terapia de Alvo Molecular , Fenótipo , Inibidores de Proteínas Quinases/efeitos adversos , Transdução de Sinais , Malformações Vasculares/metabolismo , Malformações Vasculares/patologia
7.
Circ Res ; 129(1): 136-154, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34166072

RESUMO

Lymphatic vessels maintain tissue fluid homeostasis by returning to blood circulation interstitial fluid that has extravasated from the blood capillaries. They provide a trafficking route for cells of the immune system, thus critically contributing to immune surveillance. Developmental or functional defects in the lymphatic vessels, their obstruction or damage, lead to accumulation of fluid in tissues, resulting in lymphedema. Here we discuss developmental lymphatic anomalies called lymphatic malformations and complex lymphatic anomalies that manifest as localized or multifocal lesions of the lymphatic vasculature, respectively. They are rare diseases that are caused mostly by somatic mutations and can present with variable symptoms based upon the size and location of the lesions composed of fluid-filled cisterns or channels. Substantial progress has been made recently in understanding the molecular basis of their pathogenesis through the identification of their genetic causes, combined with the elucidation of the underlying mechanisms in animal disease models and patient-derived lymphatic endothelial cells. Most of the solitary somatic mutations that cause lymphatic malformations and complex lymphatic anomalies occur in genes that encode components of oncogenic growth factor signal transduction pathways. This has led to successful repurposing of some targeted cancer therapeutics to the treatment of lymphatic malformations and complex lymphatic anomalies. Apart from the mutations that act as lymphatic endothelial cell-autonomous drivers of these anomalies, current evidence points to superimposed paracrine mechanisms that critically contribute to disease pathogenesis and thus provide additional targets for therapeutic intervention. Here, we review these advances and discuss new treatment strategies that are based on the recently identified molecular pathways.


Assuntos
Linfangiogênese , Anormalidades Linfáticas/genética , Anormalidades Linfáticas/terapia , Vasos Linfáticos/anormalidades , Mutação , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Anormalidades Linfáticas/metabolismo , Anormalidades Linfáticas/patologia , Vasos Linfáticos/metabolismo , Fenótipo , Transdução de Sinais
8.
Eur J Neurol ; 29(10): 3061-3070, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35715928

RESUMO

BACKGROUND AND PURPOSE: Sturge-Weber syndrome (SWS) is a neurocutaneous disorder characterized by clinical manifestations involving the brain, eye and skin. SWS is commonly caused by somatic mutations in G protein subunit Alpha Q (GNAQ). Five cases of subunit Alpha 11 (GNA11) mutations have been reported. We studied phenotypic features of GNA11-SWS and compared them with those of classic SWS. METHODS: Within two European multidisciplinary centers we looked for patients with clinical characteristics of SWS and a GNA11 mutation. Clinical and radiological data were collected retrospectively and prospectively. RESULTS: We identified three patients with SWS associated with a somatic GNA11 mutation. All had disseminated capillary malformation (CM) and hyper- or hypotrophy of an extremity. At birth, the CMs of the face, trunk and limbs were pink and patchy, and slowly darkened with age, evolving to a purple color. Two of the patients had glaucoma. All had neurological symptoms and moderate brain atrophy with a lower degree of severity than that classically associated with SWS. Susceptibility-weighted imaging (SWI) and contrast-enhanced fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging demonstrated the best sensitivity to reveal the pial angiomas. CONCLUSIONS: We have differentiated two distinct clinical/radiological phenotypes of SWS; GNAQ- and GNA11-SWS. The classic GNAQ-SWS is characterized by a homogeneous dark-red CM, commonly associated with underlying soft tissue hypertrophy. The CM in GNA11-SWS is more reticulate and darkens with time, and the neurological picture is milder. SWI and post-contrast FLAIR sequences appear to be necessary to demonstrate leptomeningeal angiomatosis. Anti-epileptic medication or future targeted therapies may be useful, as in classic SWS.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP , Síndrome de Sturge-Weber , Anticonvulsivantes , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Humanos , Imageamento por Ressonância Magnética , Estudos Retrospectivos , Síndrome de Sturge-Weber/complicações , Síndrome de Sturge-Weber/genética , Síndrome de Sturge-Weber/patologia
9.
J Oral Pathol Med ; 51(10): 878-887, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35610188

RESUMO

Vascular anomalies are a heterogenous group of vascular lesions that can be divided, according to the International Society for the Study of Vascular Anomalies Classification, into two main groups: vascular tumors and vascular malformations. Vascular malformations can be further subdivided into slow-flow and fast-flow malformations. This clinical and radiological classification allows for a better understanding of vascular anomalies and aims to offer a more precise final diagnosis. Correct diagnosis is essential to propose the best treatment, which traditionally consists of surgery, embolization, or sclerotherapy. Since a few years, medical treatment has become an important part of multidisciplinary treatment. Genetic and molecular knowledge of vascular anomalies are increasing rapidly and opens the door for a molecular classification of vascular anomalies according to the underlying pathways involved. The main pathways seem to be PI3K/AKT/mTOR and RAS/RAF/MEK/ERK. Knowing the underlying molecular cascades allows us to use targeted medical therapies. The first part of this article aims to review the vascular anomalies seen in the head and neck region and their underlying molecular causes and involved pathways. The second part will propose an overview of the available targeted therapies based on the affected molecular cascade. This article summarizes theragnostic treatments available in vascular anomalies.


Assuntos
Fosfatidilinositol 3-Quinases , Malformações Vasculares , Humanos , Malformações Vasculares/terapia , Malformações Vasculares/diagnóstico por imagem , Malformações Vasculares/patologia , Pescoço/patologia , Escleroterapia , Radiografia
10.
Acta Orthop Belg ; 88(3): 475-481, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36791700

RESUMO

Gorham Stout disease is a very rare monostotic or polyostotic osteolysis and physiopathology of the osteolysis is not yet fully understood. Three new cases are reported with their evolution and treatment. Among these 3 cases, two are very rare cases of polyostotic involvement. One patient finally deceased from respiratory complications despite limb amputation. The two others are alive. Both needed final reconstruction with massive bone allograft for one and with a prosthesis for the other. Monostotic osteolysis is the most frequent presentation of Gorham Stout disease and extensive polyostotic osteolysis is very rare. Treatment methods vary from one clinic to another, from drug treatment to surgical treatment with or without radiotherapy. Sometimes, as a last solution, an amputation of the affected limb is performed. The prognosis depends on the affected region and the reponse to various treatments. Chylothorax seems to be a factor of poor prognosis.


Assuntos
Quilotórax , Osteólise Essencial , Osteólise , Humanos , Osteólise Essencial/diagnóstico , Osteólise Essencial/diagnóstico por imagem , Osteólise/etiologia , Osteólise/complicações , Prognóstico , Quilotórax/complicações , Transplante Ósseo/métodos
11.
J Med Genet ; 57(1): 48-52, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31300548

RESUMO

BACKGROUND: Capillary malformation-arteriovenous malformation is an autosomal dominant disorder, characterised by capillary malformations and increased risk of fast-flow vascular malformations, caused by loss-of-function mutations in the RASA1 or EPHB4 genes. Around 25% of the patients do not seem to carry a germline mutation in either one of these two genes. Even if other genes could be involved, some individuals may have mutations in the known genes that escaped detection by less sensitive techniques. We tested the hypothesis that mosaic mutations could explain some of previously negative cases. METHODS: DNA was extracted from peripheral blood lymphocytes, saliva or vascular malformation tissues from four patients. RASA1 and EPHB4 coding regions and exon/intron boundaries were analysed by targeted custom gene panel sequencing. A second panel and/or Sanger sequencing were used to confirm the identified mutations. RESULTS: Four distinct mosaic RASA1 mutations, with an allele frequency ranging from 3% to 25%, were identified in four index patients with classical capillary malformation-arteriovenous malformation phenotype. Three mutations were known, one was novel. In one patient, a somatic second hit was also identified. One index case had three affected children, illustrating that the mosaicism was also present in the germline. CONCLUSION: This study shows that RASA1 mosaic mutations can cause capillary malformation-arteriovenous malformation. Thus, highly sensitive sequencing techniques should be considered as diagnostic tools, especially for patients with no family history. Even low-level mosaicism can cause the classical phenotype and increased risk for offspring. In addition, our study further supports the second-hit pathophysiological mechanism to explain the multifocality of vascular lesions in this disorder.


Assuntos
Malformações Arteriovenosas/genética , Capilares/anormalidades , Mosaicismo , Mutação , Mancha Vinho do Porto/genética , Proteína p120 Ativadora de GTPase/genética , Malformações Arteriovenosas/diagnóstico , Malformações Arteriovenosas/metabolismo , Capilares/metabolismo , Análise Mutacional de DNA , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mancha Vinho do Porto/diagnóstico , Mancha Vinho do Porto/metabolismo
12.
Curr Opin Hematol ; 26(3): 185-192, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30855337

RESUMO

PURPOSE OF REVIEW: The field of vascular anomalies has seen a fundamental change during the past 10 years. The identification of somatic genetic mutations as the explanation of sporadic vascular anomalies opened the doors to study prospectively and a posteriori the causes of various vascular malformations. This was helped by the rapidly evolving genetic techniques including the highly sensitive next generation sequencing. In parallel, knowledge on signaling alterations occurring in vascular endothelial cells because of the various mutations, development of in-vitro and especially the first in-vivo models, gave the possibility to test preclinically molecular therapies for vascular malformations. RECENT FINDINGS: One of the first molecules, rapamycin, showed clear evidence of interrupting lesion growth. As its safety profile had been established in other conditions, it was quickly accepted for clinical trials on vascular anomalies. Now, with a few trials published and others ongoing, it is establishing itself as a gold standard for molecular therapy for recalcitrant lesions. SUMMARY: Targeted molecular therapies are becoming interesting new additions to the management of vascular anomalies, and rapamycin is establishing itself as a gold standard for venous malformations.


Assuntos
Células Endoteliais , Transdução de Sinais/efeitos dos fármacos , Sirolimo/uso terapêutico , Malformações Vasculares , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Malformações Vasculares/tratamento farmacológico , Malformações Vasculares/metabolismo , Malformações Vasculares/patologia
13.
Circulation ; 136(11): 1037-1048, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28687708

RESUMO

BACKGROUND: Most arteriovenous malformations (AVMs) are localized and occur sporadically. However, they also can be multifocal in autosomal-dominant disorders, such as hereditary hemorrhagic telangiectasia and capillary malformation (CM)-AVM. Previously, we identified RASA1 mutations in 50% of patients with CM-AVM. Herein we studied non-RASA1 patients to further elucidate the pathogenicity of CMs and AVMs. METHODS: We conducted a genome-wide linkage study on a CM-AVM family. Whole-exome sequencing was also performed on 9 unrelated CM-AVM families. We identified a candidate gene and screened it in a large series of patients. The influence of several missense variants on protein function was also studied in vitro. RESULTS: We found evidence for linkage in 2 loci. Whole-exome sequencing data unraveled 4 distinct damaging variants in EPHB4 in 5 families that cosegregated with CM-AVM. Overall, screening of EPHB4 detected 47 distinct mutations in 54 index patients: 27 led to a premature stop codon or splice-site alteration, suggesting loss of function. The other 20 are nonsynonymous variants that result in amino acid substitutions. In vitro expression of several mutations confirmed loss of function of EPHB4. The clinical features included multifocal CMs, telangiectasias, and AVMs. CONCLUSIONS: We found EPHB4 mutations in patients with multifocal CMs associated with AVMs. The phenotype, CM-AVM2, mimics RASA1-related CM-AVM1 and also hereditary hemorrhagic telangiectasia. RASA1-encoded p120RASGAP is a direct effector of EPHB4. Our data highlight the pathogenetic importance of this interaction and indicts EPHB4-RAS-ERK signaling pathway as a major cause for AVMs.


Assuntos
Malformações Arteriovenosas/diagnóstico , Malformações Arteriovenosas/genética , Capilares/anormalidades , Mutação em Linhagem Germinativa/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Mancha Vinho do Porto/diagnóstico , Mancha Vinho do Porto/genética , Receptor EphB4/genética , Proteína p120 Ativadora de GTPase/genética , Bases de Dados Genéticas , Feminino , Estudo de Associação Genômica Ampla/métodos , Humanos , Masculino , Linhagem
14.
Am J Hum Genet ; 97(6): 914-21, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26637981

RESUMO

Somatic mutations in TEK, the gene encoding endothelial cell tyrosine kinase receptor TIE2, cause more than half of sporadically occurring unifocal venous malformations (VMs). Here, we report that somatic mutations in PIK3CA, the gene encoding the catalytic p110α subunit of PI3K, cause 54% (27 out of 50) of VMs with no detected TEK mutation. The hotspot mutations c.1624G>A, c.1633G>A, and c.3140A>G (p.Glu542Lys, p.Glu545Lys, and p.His1047Arg), frequent in PIK3CA-associated cancers, overgrowth syndromes, and lymphatic malformation (LM), account for >92% of individuals who carry mutations. Like VM-causative mutations in TEK, the PIK3CA mutations cause chronic activation of AKT, dysregulation of certain important angiogenic factors, and abnormal endothelial cell morphology when expressed in human umbilical vein endothelial cells (HUVECs). The p110α-specific inhibitor BYL719 restores all abnormal phenotypes tested, in PIK3CA- as well as TEK-mutant HUVECs, demonstrating that they operate via the same pathogenic pathways. Nevertheless, significant genotype-phenotype correlations in lesion localization and histology are observed between individuals with mutations in PIK3CA versus TEK, pointing to gene-specific effects.


Assuntos
Mutação , Neovascularização Patológica/genética , Fosfatidilinositol 3-Quinases/genética , Malformações Vasculares/genética , Alelos , Classe I de Fosfatidilinositol 3-Quinases , Regulação da Expressão Gênica , Frequência do Gene , Estudos de Associação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Receptor TIE-2/antagonistas & inibidores , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transdução de Sinais , Tiazóis/farmacologia , Transfecção , Malformações Vasculares/enzimologia , Malformações Vasculares/patologia , Veias/enzimologia , Veias/patologia
15.
Pediatr Dermatol ; 35(6): e382-e388, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30216524

RESUMO

We herein report the case of a 3-year-old girl with atypical congenital right upper limb lymphedema who developed an angiosarcoma. Only a few cases have been reported following congenital form of lymphedema and only 4 in such a young child. We also summarize all cases of angiosarcoma associated with congenital lymphedema reported in the literature.


Assuntos
Hemangiossarcoma/diagnóstico , Linfangiossarcoma/diagnóstico , Linfedema/complicações , Antineoplásicos/uso terapêutico , Pré-Escolar , Evolução Fatal , Feminino , Hemangiossarcoma/terapia , Humanos , Lactente , Linfangiossarcoma/terapia , Linfedema/congênito , Pele/patologia , Extremidade Superior/patologia
16.
Hum Mol Genet ; 24(22): 6374-89, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26319232

RESUMO

Venous malformations (VMs) are localized defects in vascular morphogenesis frequently caused by mutations in the gene for the endothelial tyrosine kinase receptor TIE2. Here, we report the analysis of a comprehensive collection of 22 TIE2 mutations identified in patients with VM, either as single amino acid substitutions or as double-mutations on the same allele. Using endothelial cell (EC) cultures, mouse models and ultrastructural analysis of tissue biopsies from patients, we demonstrate common as well as mutation-specific cellular and molecular features, on the basis of which mutations cluster into categories that correlate with data from genetic studies. Comparisons of double-mutants with their constituent single-mutant forms identified the pathogenic contributions of individual changes, and their compound effects. We find that defective receptor trafficking and subcellular localization of different TIE2 mutant forms occur via a variety of mechanisms, resulting in attenuated response to ligand. We also demonstrate, for the first time, that TIE2 mutations cause chronic activation of the MAPK pathway resulting in loss of normal EC monolayer due to extracellular matrix (ECM) fibronectin deficiency and leading to upregulation of plasminogen/plasmin proteolytic pathway. Corresponding EC and ECM irregularities are observed in affected tissues from mouse models and patients. Importantly, an imbalance between plasminogen activators versus inhibitors would also account for high d-dimer levels, a major feature of unknown cause that distinguishes VMs from other vascular anomalies.


Assuntos
Receptor TIE-2/genética , Malformações Vasculares/genética , Substituição de Aminoácidos , Animais , Movimento Celular/genética , Células Endoteliais/metabolismo , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligantes , Camundongos , Camundongos SCID , Mutação , Fosforilação , Receptor TIE-2/metabolismo , Transdução de Sinais , Esferoides Celulares , Malformações Vasculares/enzimologia
17.
Am J Hum Genet ; 92(2): 188-96, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23375657

RESUMO

Inherited vascular malformations are commonly autosomal dominantly inherited with high, but incomplete, penetrance; they often present as multiple lesions. We hypothesized that Knudson's two-hit model could explain this multifocality and partial penetrance. We performed a systematic analysis of inherited glomuvenous malformations (GVMs) by using multiple approaches, including a sensitive allele-specific pairwise SNP-chip method. Overall, we identified 16 somatic mutations, most of which were not intragenic but were cases of acquired uniparental isodisomy (aUPID) involving chromosome 1p. The breakpoint of each aUPID is located in an A- and T-rich, high-DNA-flexibility region (1p13.1-1p12). This region corresponds to a possible new fragile site. Occurrences of these mutations render the inherited glomulin variant in 1p22.1 homozygous in the affected tissues without loss of genetic material. This finding demonstrates that a double hit is needed to trigger formation of a GVM. It also suggests that somatic UPID, only detectable by sensitive pairwise analysis in heterogeneous tissues, might be a common phenomenon in human cells. Thus, aUPID might play a role in the pathogenesis of various nonmalignant disorders and might explain local impaired function and/or clinical variability. Furthermore, these data suggest that pairwise analysis of blood and tissue, even on heterogeneous tissue, can be used for localizing double-hit mutations in disease-causing genes.


Assuntos
Tumor Glômico/genética , Paraganglioma Extrassuprarrenal/genética , Dissomia Uniparental/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Quebra Cromossômica , Cromossomos Humanos Par 1/genética , DNA/genética , Feminino , Tumor Glômico/patologia , Humanos , Hibridização in Situ Fluorescente , Masculino , Mutação/genética , Paraganglioma Extrassuprarrenal/patologia , Polimorfismo de Nucleotídeo Único/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
18.
J Am Acad Dermatol ; 75(1): 64-8, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27004804

RESUMO

BACKGROUND: There is limited information regarding early development of soft-tissue and/or bone hypertrophy with facial port-wine stains (PWS). OBJECTIVE: We sought to characterize patients with hypertrophic PWS presenting during childhood. METHODS: Patients with a facial PWS and underlying hypertrophy that developed before the age of 18 years were included in a multicenter retrospective study. Age at onset of the hypertrophy, its location, association with odontologic problems, presence of other associated complications, and response to laser treatment were recorded. RESULTS: A total of 98 patients were included. The mean age at onset of hypertrophy, retrieved for 77 of 98 patients, was 5.6 years. The hypertrophy was congenital in 26%. Odontologic problems were noted in 39.8% of cases. Other complications, including cataract, asymmetric development of the maxillary bone, and speech delay/disorders, were reported in 18.4%. In all, 67 patients received laser treatment. Only 3% achieved complete or nearly complete clearance of the PWS. LIMITATIONS: As only cases of PWS with early-onset hypertrophy were included, we were unable to calculate the prevalence of this manifestation. CONCLUSION: PWS with early-onset hypertrophy are associated with a high rate of complications and a poor response to laser treatment. Periodic monitoring is recommended for early detection and treatment of complications.


Assuntos
Anormalidades Múltiplas , Lasers de Corante/uso terapêutico , Mancha Vinho do Porto/patologia , Mancha Vinho do Porto/cirurgia , Anormalidades Múltiplas/diagnóstico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Face , Feminino , Humanos , Hipertrofia/congênito , Hipertrofia/patologia , Hipertrofia/cirurgia , Lactente , Masculino , Pessoa de Meia-Idade , Pescoço , Mancha Vinho do Porto/complicações , Prognóstico , Estudos Retrospectivos , Adulto Jovem
19.
J Pediatr Orthop ; 36(3): 316-22, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25757206

RESUMO

BACKGROUND: To assess the efficacy of surgical treatment of intra-articular knee venous malformations (VM). METHODS: Between 1998 and 2010, 8 children (mean age: 12.3 y) underwent surgical resection of their vascular malformation (7 venous and 1 capillary venous) involving the knee joint. The lesion was diffuse in 6 cases and well-demarcated in 2 cases. All children were suffering from knee pain and had recurrent hemarthroses. Color-Doppler ultrasonography, magnetic resonance imaging, computed tomography scan, and blood test were performed preoperatively. Preoperative and postoperative physical examination, clinical symptoms, and orthopaedic evaluation were retrospectively reviewed. Surgery consisted in arthrotomy with total excision of the vascular malformation for the 2 well-demarcated lesions and synovectomy with squeezing of the surrounding vascular malformation for 5 diffuse lesions. One patient with an extensive venous malformation associated with severe localized intravascular coagulopathy and mild hemophilia A had undergone synovectomy by knee arthroscopy. RESULTS: Immediate postoperative follow-up was uneventful in 6 patients, whereas 2 patients with diffuse vascular malformation and coagulopathy suffered from postoperative hemarthroses, delaying their rehabilitation. After a mean follow-up of 5.1 years, persistence of the VM within the joint was observed in the 6 initially diffuse lesions. The 2 well-demarcated lesions showed no evidence of disease at latest follow-up. Four patients with preoperative chondropathy and functional impairment were not substantially improved regarding their range of knee motion at latest follow-up, whereas the 4 others were free of symptoms. Only 1 patient presented a recurrent hemarthroses after a 5-year-symptom-free period and had to be reoperated. Patients without preoperative chondropathy recovered normal knee function mobility. CONCLUSIONS: This retrospective study highlights the importance of early surgery in patients with intra-articular venous malformation, even if asymptomatic, to prevent joint impairment. For well-demarcated lesions, total resection by arthrotomy can provide definitive healing without resuming of symptoms. Although diffuse lesions treated by synovectomy still persist in the joint, treatment avoids recurrence of hemarthroses and, therefore, protects the cartilage from further erosion. LEVEL OF EVIDENCE: Level IV-cases series.


Assuntos
Hemartrose/etiologia , Articulação do Joelho/irrigação sanguínea , Complicações Pós-Operatórias/etiologia , Malformações Vasculares/cirurgia , Adolescente , Artralgia/etiologia , Artroscopia , Transtornos da Coagulação Sanguínea/complicações , Doenças das Cartilagens/complicações , Criança , Feminino , Seguimentos , Humanos , Articulação do Joelho/fisiopatologia , Masculino , Amplitude de Movimento Articular , Recidiva , Estudos Retrospectivos , Sinovectomia , Malformações Vasculares/complicações
20.
Hum Mol Genet ; 22(17): 3438-48, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23633549

RESUMO

Mutations in the endothelial cell (EC) tyrosine kinase receptor TIE2 cause inherited and sporadic forms of venous malformation. The recurrent somatic mutation L914F and common germline mutation R849W differ in terms of phosphorylation level, as well as sub-cellular localization and trafficking of the receptor. Previous studies have shed light on certain pathogenic properties of R849W, but the mechanisms of action of L914F are unknown. We used global gene expression profiling to study the effects of L914F on ECs. We found that L914F strongly dysregulates genes involved in vascular development, cell migration and extracellular matrix processing, while R849W has weak effects. We also demonstrate, for the first time, that TIE2-mutant ECs are deficient in the production of PDGFB, both in vitro and ex vivo in patient tissues. This defect is mediated by the chronic, ligand-independent activation of AKT by the mutant receptors. Inadequate secretion of the major mural cell attractant likely plays an important role in the development of abnormal vascular channels, contributing to the characteristic paucity of surrounding vascular smooth muscle cells.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Malformações Vasculares/genética , Malformações Vasculares/metabolismo , Movimento Celular/genética , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Mutação em Linhagem Germinativa , Humanos , Músculo Liso Vascular/metabolismo , Fosforilação , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA