Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 33(14): 1250-1261, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38676400

RESUMEN

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.


Asunto(s)
Factor de Crecimiento de Hepatocito , Linfedema , Humanos , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Masculino , Femenino , Niño , Adulto , Linfedema/genética , Linfedema/patología , Adolescente , Persona de Mediana Edad , Animales , Mutación Missense/genética , Mutación con Pérdida de Función , Edad de Inicio , Preescolar , Células COS , Chlorocebus aethiops , Células Endoteliales/metabolismo , Células Endoteliales/patología , Adulto Joven
2.
Hum Mol Genet ; 32(2): 276-289, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-35972810

RESUMEN

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.


Asunto(s)
Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas p21(ras) , Ratones , Animales , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias Pancreáticas/genética , Células Endoteliales/patología , Modelos Animales de Enfermedad , Mutación
3.
Am J Med Genet A ; 194(6): e63551, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38321651

RESUMEN

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.


Asunto(s)
Capilares , Epilepsia , Proteínas Proto-Oncogénicas c-akt , Telangiectasia , Malformaciones Vasculares , Femenino , Humanos , Recién Nacido , Masculino , Capilares/anomalías , Capilares/patología , Epilepsia/genética , Epilepsia/patología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mosaicismo , Mutación/genética , Fenotipo , Proteínas Proto-Oncogénicas c-akt/genética , Telangiectasia/genética , Telangiectasia/patología , Telangiectasia/diagnóstico , Malformaciones Vasculares/genética , Malformaciones Vasculares/patología , Malformaciones Vasculares/diagnóstico , Malformaciones Vasculares/complicaciones , Adolescente
4.
J Med Genet ; 60(1): 57-64, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-34876502

RESUMEN

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.


Asunto(s)
Angiopoyetina 2 , Hidropesía Fetal , Animales , Femenino , Humanos , Ratones , Embarazo , Angiopoyetina 2/genética , Codón sin Sentido/genética , Heterocigoto , Hidropesía Fetal/genética , Hidropesía Fetal/metabolismo , Mutación Missense , Recién Nacido
5.
Circ Res ; 129(1): 155-173, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34166070

RESUMEN

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.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Vasos Sanguíneos/anomalías , Vasos Sanguíneos/efectos de los fármacos , Mutación , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Malformaciones Vasculares/tratamiento farmacológico , Malformaciones Vasculares/genética , Inhibidores de la Angiogénesis/efectos adversos , Animales , Vasos Sanguíneos/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Terapia Molecular Dirigida , Fenotipo , Inhibidores de Proteínas Quinasas/efectos adversos , Transducción de Señal , Malformaciones Vasculares/metabolismo , Malformaciones Vasculares/patología
6.
Circ Res ; 129(1): 136-154, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34166072

RESUMEN

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.


Asunto(s)
Linfangiogénesis , Anomalías Linfáticas/genética , Anomalías Linfáticas/terapia , Vasos Linfáticos/anomalías , Mutación , Animales , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Anomalías Linfáticas/metabolismo , Anomalías Linfáticas/patología , Vasos Linfáticos/metabolismo , Fenotipo , Transducción de Señal
7.
J Med Genet ; 57(1): 48-52, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31300548

RESUMEN

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.


Asunto(s)
Malformaciones Arteriovenosas/genética , Capilares/anomalías , Mosaicismo , Mutación , Mancha Vino de Oporto/genética , Proteína Activadora de GTPasa p120/genética , Malformaciones Arteriovenosas/diagnóstico , Malformaciones Arteriovenosas/metabolismo , Capilares/metabolismo , Análisis Mutacional de ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mancha Vino de Oporto/diagnóstico , Mancha Vino de Oporto/metabolismo
8.
Curr Opin Hematol ; 26(3): 185-192, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30855337

RESUMEN

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.


Asunto(s)
Células Endoteliales , Transducción de Señal/efectos de los fármacos , Sirolimus/uso terapéutico , Malformaciones Vasculares , Animales , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Malformaciones Vasculares/tratamiento farmacológico , Malformaciones Vasculares/metabolismo , Malformaciones Vasculares/patología
9.
Circulation ; 136(11): 1037-1048, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28687708

RESUMEN

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.


Asunto(s)
Malformaciones Arteriovenosas/diagnóstico , Malformaciones Arteriovenosas/genética , Capilares/anomalías , Mutación de Línea Germinal/genética , Sistema de Señalización de MAP Quinasas/fisiología , Mancha Vino de Oporto/diagnóstico , Mancha Vino de Oporto/genética , Receptor EphB4/genética , Proteína Activadora de GTPasa p120/genética , Bases de Datos Genéticas , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Linaje
10.
Am J Hum Genet ; 97(6): 914-21, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26637981

RESUMEN

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.


Asunto(s)
Mutación , Neovascularización Patológica/genética , Fosfatidilinositol 3-Quinasas/genética , Malformaciones Vasculares/genética , Alelos , Fosfatidilinositol 3-Quinasa Clase I , Regulación de la Expresión Génica , Frecuencia de los Genes , Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Receptor TIE-2/antagonistas & inhibidores , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Transducción de Señal , Tiazoles/farmacología , Transfección , Malformaciones Vasculares/enzimología , Malformaciones Vasculares/patología , Venas/enzimología , Venas/patología
11.
Pediatr Dermatol ; 35(6): e382-e388, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30216524

RESUMEN

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.


Asunto(s)
Hemangiosarcoma/diagnóstico , Linfangiosarcoma/diagnóstico , Linfedema/complicaciones , Antineoplásicos/uso terapéutico , Preescolar , Resultado Fatal , Femenino , Hemangiosarcoma/terapia , Humanos , Lactante , Linfangiosarcoma/terapia , Linfedema/congénito , Piel/patología , Extremidad Superior/patología
12.
Hum Mol Genet ; 24(22): 6374-89, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26319232

RESUMEN

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.


Asunto(s)
Receptor TIE-2/genética , Malformaciones Vasculares/genética , Sustitución de Aminoácidos , Animales , Movimiento Celular/genética , Células Endoteliales/metabolismo , Femenino , Productos de Degradación de Fibrina-Fibrinógeno , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ligandos , Ratones , Ratones SCID , Mutación , Fosforilación , Receptor TIE-2/metabolismo , Transducción de Señal , Esferoides Celulares , Malformaciones Vasculares/enzimología
13.
Am J Hum Genet ; 92(2): 188-96, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23375657

RESUMEN

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.


Asunto(s)
Tumor Glómico/genética , Paraganglioma Extraadrenal/genética , Disomía Uniparental/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Rotura Cromosómica , Cromosomas Humanos Par 1/genética , ADN/genética , Femenino , Tumor Glómico/patología , Humanos , Hibridación Fluorescente in Situ , Masculino , Mutación/genética , Paraganglioma Extraadrenal/patología , Polimorfismo de Nucleótido Simple/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
J Pediatr Orthop ; 36(3): 316-22, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25757206

RESUMEN

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.


Asunto(s)
Hemartrosis/etiología , Articulación de la Rodilla/irrigación sanguínea , Complicaciones Posoperatorias/etiología , Malformaciones Vasculares/cirugía , Adolescente , Artralgia/etiología , Artroscopía , Trastornos de la Coagulación Sanguínea/complicaciones , Enfermedades de los Cartílagos/complicaciones , Niño , Femenino , Estudios de Seguimiento , Humanos , Articulación de la Rodilla/fisiopatología , Masculino , Rango del Movimiento Articular , Recurrencia , Estudios Retrospectivos , Sinovectomía , Malformaciones Vasculares/complicaciones
15.
Hum Mol Genet ; 22(17): 3438-48, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23633549

RESUMEN

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.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Malformaciones Vasculares/genética , Malformaciones Vasculares/metabolismo , Movimiento Celular/genética , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Mutación de Línea Germinal , Humanos , Músculo Liso Vascular/metabolismo , Fosforilación , Transducción de Señal/genética , Transducción de Señal/fisiología
16.
Eur J Pediatr ; 174(7): 855-65, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26021855

RESUMEN

UNLABELLED: With a prevalence of 2.6-4.5 %, infantile haemangiomas (IH) represent the most common tumour of infancy. While the majority of IH does not require therapy and regresses spontaneously, about 10 % of IH exhibit complications such as obstruction, ulceration or disfigurement. With the advent of oral propranolol, many conventional treatment options have become obsolete. This paper summarizes current recommendations for management of complicated IH. These recommendations have been written by an expert group after a consensus process including bibliographic review, several drafts of synthesis, meetings with quantitative voting system and redaction of an approved final manuscript. CONCLUSION: Oral propranolol is the first-line agent for the treatment of complicated IH. WHAT IS KNOWN: • Infantile haemangiomas (IH) are the most common tumours of infancy. Within a very short period after its discovery and long before the publication of randomized controlled trials, propranolol has become the number one agent for the treatment of complicated IH. What is New: • We report IH treatment recommendations of an international, interdisciplinary team of experts, based on an up-to-date review of the literature.


Asunto(s)
Hemangioma/terapia , Administración Tópica , Antagonistas Adrenérgicos beta/uso terapéutico , Coartación Aórtica/complicaciones , Crioterapia , Diagnóstico Diferencial , Estética , Anomalías del Ojo/complicaciones , Glucocorticoides/uso terapéutico , Hemangioma/diagnóstico , Humanos , Inmunosupresores/uso terapéutico , Lactante , Terapia por Láser , Síndromes Neurocutáneos/complicaciones , Fototerapia , Propranolol/uso terapéutico , Factores de Riesgo , Sirolimus/uso terapéutico , Neoplasias Vasculares/diagnóstico , Espera Vigilante
17.
Res Pract Thromb Haemost ; 8(3): 102400, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38738172

RESUMEN

Background: Venous malformations (VMs) are commonly associated with localized intravascular coagulopathy leading to elevated D-dimer and risks of hemorrhagic and thromboembolic events, particularly in extensive lesions. While low-molecular-weight heparin (LMWH) has been effective in managing coagulopathy and pain, direct oral anticoagulants (DOACs) emerge as a promising alternative. Objectives: This study aims to evaluate the efficacy and safety of DOACs in treating VMs associated with localized intravascular coagulopathy, offering a comparative perspective to LMWH. Methods: A retrospective study was conducted on 29 patients with VMs and secondary localized intravascular coagulopathy treated with DOACs between 2013 and 2023 in a single tertiary center specialized in vascular anomalies. Data were collected from February 24, 2023, to September 1, 2023. Results: Patients' median age was 40 years (range, 22-76 years), with a female predominance of 66%. Descriptive statistical analysis showed that 85% of patients experienced pain improvement, and 86% showed a reduction in D-dimer by at least 25%, with a mean reduction of 57% (SD, ±32%; IQR, [38-81%]). Additionally, 37% of patients reported a bleeding event, mostly minor. Conclusion: The study findings suggests that DOACs may serve as an alternative to LMWH for patients with VMs associated with pain management and reduced D-dimer, alongside a low observed risk of major bleeding. Tailored dosing considering the location of the malformation, bleeding and thrombotic tendencies, and laboratory abnormalities is recommended. Future studies with larger cohorts and extended follow-up are necessary for more conclusive evidence on DOACs' role in this patient population.

18.
Orphanet J Rare Dis ; 19(1): 199, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38750525

RESUMEN

Repurposing anticancer drugs to vascular malformations has significantly improved patient outcomes. Complex Lymphatic Anomalies (CLA) are part of the spectrum of lymphatic malformations (LMs) that share similar oncogenic mutations to cancer. We report the case of a young patient with highly symptomatic CLA who was initially treated with sirolimus, due to the frequent involvement of the PI3K-AKT-mTOR pathway in CLA pathogenesis. Despite an initial reduction in symptoms, sirolimus progressively lost its effectiveness. After an unsuccessful attempt with trametinib alone, sirolimus was added to trametinib and resulted in a significant, rapid and sustained improvement in symptoms. This suggests that, contrary to current dogmas, combination therapy using sub-therapeutic doses targeting both the PI3K and RAS pathways retains efficacy without generating the toxicity known for combination therapies, and is beneficial in the management of CLAs and potentially other vascular anomalies.


Asunto(s)
Anomalías Linfáticas , Piridonas , Pirimidinonas , Sirolimus , Humanos , Anomalías Linfáticas/tratamiento farmacológico , Anomalías Linfáticas/patología , Piridonas/uso terapéutico , Pirimidinonas/uso terapéutico , Sirolimus/uso terapéutico
19.
Plast Reconstr Surg ; 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38739875

RESUMEN

BACKGROUND: To assess the impact of propranolol as the first-line treatment of infantile hemangioma (IH) on the need for surgery in the management of IH. METHODS: Retrospective study of 420 patients, with IH, referred to our multidisciplinary center between January 2005 and August 2014. Clinical data including sex, age at first consultation and at treatment initiation, location, size, number, aspect, and complication of IH, as well as the type of treatment were collected. Statistical analyses were conducted considering each patient and each tumor independently. RESULTS: A total of 625 IH(420 patients (P))were reviewed, 113 patients had more than one IH (26.91%). Median age at first consultation was 7 months old. Overall, 243 patients were treated (57.86%) using either surgery (n=128 P/141 IH), propranolol (n=79 P/89 IH), corticosteroids (n=51 P/56 IH), and/or laser (n=34 P /36 IH). Propranolol was effective in all but 2 infants with IH. Seven patients (n=7/79 P; 8.86%) initially treated with propranolol, still required surgery, in contrast to 18 patients (n=18/51 P; 35.29%) initially treated with corticosteroids, and 103 patients (n=103/290 P; 35.51%) with no medical treatment. Since the availability of propranolol, patients were less likely to undergo surgery (48 P versus 80 P; P-Value < 0.001). This demonstrated that the use of propranolol reduced the need for surgery (P-Value < 0.001 with an OR of 0.177: CI 95% 0.079-0.396). CONCLUSION: Propranolol has dramatically reduced the need for surgery, regarding indications and number of patients. Surgical correction remains important for sequelae management, non-responders or strawberry-like IH.

20.
J Invest Dermatol ; 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38431221

RESUMEN

Common capillary malformations are red vascular skin lesions, most commonly associated with somatic activating GNAQ or GNA11 mutations. We focused on capillary malformations lacking such a mutation to identify previously unreported genetic causes. We used targeted next-generation sequencing on 82 lesions. Bioinformatic analysis allowed the identification of 9 somatic pathogenic variants in PIK3R1 and PIK3CA, encoding for the regulatory and catalytic subunits of phosphoinositide 3-kinase, respectively. Recharacterization of these lesions unraveled a common phenotype: a pale capillary malformation associated with visible dilated veins. Primary endothelial cells from 2 PIK3R1-mutated lesions were isolated, and PI3k-Akt-mTOR and RAS-RAF-MAPK signaling were assessed by western blot. This unveiled an abnormal increase in Akt phosphorylation, effectively reduced by PI3K pathway inhibitors, such as mTOR, Akt, and PIK3CA inhibitors. The effects of mutant PIK3R1 were further studied using zebrafish embryos. Endothelium-specific expression of PIK3R1 mutants resulted in abnormal development of the posterior capillary-venous plexus. In summary, capillary malformation associated with visible dilated veins emerges as a clinical entity associated with somatic pathogenic variants in PIK3R1 or PIK3CA (nonhotspot). Our findings suggest that the activated Akt signaling can be effectively reversed by PI3K pathway inhibitors. In addition, the proposed zebrafish model holds promise as a valuable tool for future drug screening aimed at developing patient-tailored treatments.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA