Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 237
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 194(8): 1581-1591, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38705382

RESUMEN

Melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. While available treatments have improved survival, long-term benefits are still unsatisfactory. The mitogen-activated protein kinase extracellular signal-regulated kinase 5 (ERK5) promotes melanoma growth, and ERK5 inhibition determines cellular senescence and the senescence-associated secretory phenotype. Here, latent-transforming growth factor ß-binding protein 1 (LTBP1) mRNA was found to be up-regulated in A375 and SK-Mel-5 BRAF V600E melanoma cells after ERK5 inhibition. In keeping with a key role of LTBP1 in regulating transforming growth factor ß (TGF-ß), TGF-ß1 protein levels were increased in lysates and conditioned media of ERK5-knockdown (KD) cells, and were reduced upon LTBP1 KD. Both LTBP1 and TGF-ß1 proteins were increased in melanoma xenografts in mice treated with the ERK5 inhibitor XMD8-92. Moreover, treatment with conditioned media from ERK5-KD melanoma cells reduced cell proliferation and invasiveness, and TGF-ß1-neutralizing antibodies impaired these effects. In silico data sets revealed that higher expression levels of both LTBP1 and TGF-ß1 mRNA were associated with better overall survival of melanoma patients. Increased LTBP1 or TGF-ß1 expression played a beneficial role in patients treated with anti-PD1 immunotherapy, making a possible immunosuppressive role of LTBP1/TGF-ß1 unlikely upon ERK5 inhibition. This study, therefore, identifies additional desirable effects of ERK5 targeting, providing evidence of an ERK5-dependent tumor-suppressive role of TGF-ß in melanoma.


Asunto(s)
Proliferación Celular , Proteínas de Unión a TGF-beta Latente , Melanoma , Proteína Quinasa 7 Activada por Mitógenos , Factor de Crecimiento Transformador beta1 , Melanoma/metabolismo , Melanoma/patología , Melanoma/genética , Melanoma/tratamiento farmacológico , Humanos , Proteínas de Unión a TGF-beta Latente/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Animales , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/genética , Ratones , Factor de Crecimiento Transformador beta1/metabolismo , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/genética , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Med Genet ; 61(2): 109-116, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-37734846

RESUMEN

BACKGROUND: Weill-Marchesani syndrome (WMS) belongs to the group of acromelic dysplasias, defined by short stature, brachydactyly and joint limitations. WMS is characterised by specific ophthalmological abnormalities, although cardiovascular defects have also been reported. Monoallelic variations in FBN1 are associated with a dominant form of WMS, while biallelic variations in ADAMTS10, ADAMTS17 and LTBP2 are responsible for a recessive form of WMS. OBJECTIVE: Natural history description of WMS and genotype-phenotype correlation establishment. MATERIALS AND METHODS: Retrospective multicentre study and literature review. INCLUSION CRITERIA: clinical diagnosis of WMS with identified pathogenic variants. RESULTS: 61 patients were included: 18 individuals from our cohort and 43 patients from literature. 21 had variants in ADAMTS17, 19 in FBN1, 19 in ADAMTS10 and 2 in LTBP2. All individuals presented with eye anomalies, mainly spherophakia (42/61) and ectopia lentis (39/61). Short stature was present in 73% (from -2.2 to -5.5 SD), 10/61 individuals had valvulopathy. Regarding FBN1 variants, patients with a variant located in transforming growth factor (TGF)-ß-binding protein-like domain 5 (TB5) domain were significantly smaller than patients with FBN1 variant outside TB5 domain (p=0.0040). CONCLUSION: Apart from the ophthalmological findings, which are mandatory for the diagnosis, the phenotype of WMS seems to be more variable than initially described, partially explained by genotype-phenotype correlation.


Asunto(s)
Enanismo , Anomalías del Ojo , Síndrome de Weill-Marchesani , Humanos , Síndrome de Weill-Marchesani/genética , Síndrome de Weill-Marchesani/diagnóstico , Síndrome de Weill-Marchesani/patología , Enanismo/genética , Fenotipo , Estudios de Asociación Genética , Fibrilina-1/genética , Proteínas de Unión a TGF-beta Latente/genética , Estudios Multicéntricos como Asunto
3.
Am J Physiol Lung Cell Mol Physiol ; 327(3): L304-L318, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38915286

RESUMEN

Extracellular matrix (ECM) remodeling has been implicated in the irreversible obstruction of airways and destruction of alveolar tissue in chronic obstructive pulmonary disease (COPD). Studies investigating differences in the lung ECM in COPD have mainly focused on some collagens and elastin, leaving an array of ECM components unexplored. We investigated the differences in the ECM landscape comparing severe-early onset (SEO)-COPD and moderate COPD to control lung tissue for collagen type I α chain 1 (COL1A1), collagen type VI α chain 1 (COL6A1); collagen type VI α chain 2 (COL6A2), collagen type XIV α chain 1 (COL14A1), fibulin 2 and 5 (FBLN2 and FBLN5), latent transforming growth factor ß binding protein 4 (LTBP4), lumican (LUM), versican (VCAN), decorin (DCN), and elastin (ELN) using image analysis and statistical modeling. Percentage area and/or mean intensity of expression of LUM in the parenchyma, and COL1A1, FBLN2, LTBP4, DCN, and VCAN in the airway walls, was proportionally lower in COPD compared to controls. Lowered levels of most ECM proteins were associated with decreasing forced expiratory volume in 1 s (FEV1) measurements, indicating a relationship with disease severity. Furthermore, we identified six unique ECM signatures where LUM and COL6A1 in parenchyma and COL1A1, FBLN5, DCN, and VCAN in airway walls appear essential in reflecting the presence and severity of COPD. These signatures emphasize the need to examine groups of proteins to represent an overall difference in the ECM landscape in COPD that are more likely to be related to functional effects than individual proteins. Our study revealed differences in the lung ECM landscape between control and COPD and between SEO and moderate COPD signifying distinct pathological processes in the different subgroups.NEW & NOTEWORTHY Our study identified chronic obstructive pulmonary disease (COPD)-associated differences in the lung extracellular matrix (ECM) composition. We highlight the compartmental differences in the ECM landscape in different subtypes of COPD. The most prominent differences were observed for severe-early onset COPD. Moreover, we identified unique ECM signatures that describe airway walls and parenchyma providing insight into the intertwined nature and complexity of ECM changes in COPD that together drive ECM remodeling and may contribute to disease pathogenesis.


Asunto(s)
Decorina , Elastina , Proteínas de la Matriz Extracelular , Matriz Extracelular , Pulmón , Enfermedad Pulmonar Obstructiva Crónica , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Humanos , Masculino , Persona de Mediana Edad , Pulmón/metabolismo , Pulmón/patología , Femenino , Proteínas de la Matriz Extracelular/metabolismo , Elastina/metabolismo , Decorina/metabolismo , Anciano , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Versicanos/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Lumican/metabolismo , Colágeno Tipo I/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Índice de Severidad de la Enfermedad , Colágeno Tipo VI/metabolismo
4.
Hum Mol Genet ; 31(19): 3281-3289, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-35567544

RESUMEN

A disproportionate tall stature is the most evident manifestation in Marfan syndrome (MFS), a multisystem condition caused by mutations in the extracellular protein and TGFß modulator, fibrillin-1. Unlike cardiovascular manifestations, there has been little effort devoted to unravel the molecular mechanism responsible for long bone overgrowth in MFS. By combining the Cre-LoxP recombination system with metatarsal bone cultures, here we identify the outer layer of the perichondrium as the tissue responsible for long bone overgrowth in MFS mice. Analyses of differentially expressed genes in the fibrillin-1-deficient perichondrium predicted that loss of TGFß signaling may influence chondrogenesis in the neighboring epiphyseal growth plate (GP). Immunohistochemistry revealed that fibrillin-1 deficiency in the outer perichondrium is associated with decreased accumulation of latent TGFß-binding proteins (LTBPs)-3 and -4, and reduced levels of phosphorylated (activated) Smad2. Consistent with these findings, mutant metatarsal bones grown in vitro were longer and released less TGFß than the wild-type counterparts. Moreover, addition of recombinant TGFß1 normalized linear growth of mutant metatarsal bones. We conclude that longitudinal bone overgrowth in MFS is accounted for by diminished sequestration of LTBP-3 and LTBP-4 into the fibrillin-1-deficient matrix of the outer perichondrium, which results in less TGFß signaling locally and improper GP differentiation distally.


Asunto(s)
Síndrome de Marfan , Animales , Fibrilina-1/genética , Fibrilina-2 , Fibrilinas , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Síndrome de Marfan/genética , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
5.
Funct Integr Genomics ; 24(1): 30, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38358412

RESUMEN

LTBP1 is closely related to TGF-ß1 function as an essential component, which was unclear in gastric cancer (GC). Harbin Medical University (HMU)-GC cohort and The Cancer Genome Atlas (TCGA) dataset were combined to form a training cohort to calculate the connection between LTBP1 mRNA expression, prognosis and clinicopathological features. The training cohort was also used to verify the biological function of LTBP1 and its relationship with immune microenvironment and chemosensitivity. In the tissue microarrays (TMAs), immunohistochemical (IHC) staining was performed to observe LTBP1 protein expression. The correlation between LTBP1 protein expression level and prognosis was also analyzed, and a nomogram model was constructed. Western blotting (WB) was used in cell lines to assess LTBP1 expression. Transwell assays and CCK-8 were employed to assess LTBP1's biological roles. In compared to normal gastric tissues, LTBP1 expression was upregulated in GC tissues, and high expression was linked to a bad prognosis for GC patients. Based on a gene enrichment analysis, LTBP1 was primarily enriched in the TGF-ß and EMT signaling pathways. Furthermore, high expression of LTBP1 in the tumor microenvironment was positively correlated with an immunosuppressive response. We also found that LTBP1 expression (p = 0.006) and metastatic lymph node ratio (p = 0.044) were independent prognostic risk factors for GC patients. The prognostic model combining LTBP1 expression and lymph node metastasis ratio reliably predicted the prognosis of GC patients. In vitro proliferation and invasion of MKN-45 GC cells were inhibited and their viability was decreased by LTBP1 knockout. LTBP1 plays an essential role in the development and progression of GC, and is a potential prognostic biomarker and therapeutic target for GC.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Pronóstico , Transición Epitelial-Mesenquimal , Línea Celular , Metástasis Linfática , Microambiente Tumoral , Proteínas de Unión a TGF-beta Latente/genética
6.
Am J Hum Genet ; 108(6): 1095-1114, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33991472

RESUMEN

Latent transforming growth factor ß (TGFß)-binding proteins (LTBPs) are microfibril-associated proteins essential for anchoring TGFß in the extracellular matrix (ECM) as well as for correct assembly of ECM components. Variants in LTBP2, LTBP3, and LTBP4 have been identified in several autosomal recessive Mendelian disorders with skeletal abnormalities with or without impaired development of elastin-rich tissues. Thus far, the human phenotype associated with LTBP1 deficiency has remained enigmatic. In this study, we report homozygous premature truncating LTBP1 variants in eight affected individuals from four unrelated consanguineous families. Affected individuals present with connective tissue features (cutis laxa and inguinal hernia), craniofacial dysmorphology, variable heart defects, and prominent skeletal features (craniosynostosis, short stature, brachydactyly, and syndactyly). In vitro studies on proband-derived dermal fibroblasts indicate distinct molecular mechanisms depending on the position of the variant in LTBP1. C-terminal variants lead to an altered LTBP1 loosely anchored in the microfibrillar network and cause increased ECM deposition in cultured fibroblasts associated with excessive TGFß growth factor activation and signaling. In contrast, N-terminal truncation results in a loss of LTBP1 that does not alter TGFß levels or ECM assembly. In vivo validation with two independent zebrafish lines carrying mutations in ltbp1 induce abnormal collagen fibrillogenesis in skin and intervertebral ligaments and ectopic bone formation on the vertebrae. In addition, one of the mutant zebrafish lines shows voluminous and hypo-mineralized vertebrae. Overall, our findings in humans and zebrafish show that LTBP1 function is crucial for skin and bone ECM assembly and homeostasis.


Asunto(s)
Colágeno/metabolismo , Cutis Laxo/etiología , Variación Genética , Proteínas de Unión a TGF-beta Latente/genética , Adolescente , Alelos , Animales , Células Cultivadas , Niño , Preescolar , Cutis Laxo/patología , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Lactante , Masculino , Linaje , Piel/metabolismo , Piel/patología , Pez Cebra
7.
BMC Cancer ; 24(1): 1252, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39390437

RESUMEN

BACKGROUND: Gliomas are the highly aggressive brain tumor and also the most devastating human tumors. The latent TGF binding proteins (LTBP) had been found to be involved in malignant biological process and could be used as potent biomarkers in several solid tumors. While the role of LTBP family in human glioma remain to be elucidated. METHODS: Normalized gene expression and corresponding clinical data of 2407 gliomas samples in public datasets were downloaded from Gliovis. Kaplan-Meier methods and Cox regression analysis was used for survival analyses.Western blot (WB) and Immunohistochemical (IHC) testing were employed to test LTBPs protein level in 154 gliomas samples. Correlation between LTBP2 expression and immune infiltration was evaluated by immunofluorescence (IF) and IHC in glioma tissues. CCK8 and flow cytometric analysis were used to detect the effect of LTBP2 on glioma cells. Orthotopic glioma- mouse models were utilized to evaluate effects in vivo. RESULTS: LTBP2 mRNA level was dramatically higher in glioma samples compared with non-tumor brain tissues in XENA-TCGA_GTEx, Gill and Gravendeel datasets (all P < 0.01), and its expression positively correlated with glioma WHO grade, IDH1/2 wildtype and mesenchymal subtypes. These results were confirmed by In-house cohort which was detected by WB and IHC. We found that gliomas patients with high LTBP2 level had shorter OS than those with low LTBP2 level. LTBP2 expression significantly associated with glioma immune score (Spearman r = 0.68, P < 0.01)) and strongly correlated with infiltration degreee of macrophages both in lower grade gliomas (LGG) and GBM. Knocking down LTBP2 obviously reduced proliferation and enhanced sensitivity to temozolomide in U87 and U251 cells. Nude mice with lower expression of LTBP2 had slower tumor growth, and accompanied by less tumor-associated macrophages (TAMs) infiltration detected by IHC staining in vivo. Finally, low LTBP2 expression glioma patients who received chemotherapy survived longer than patients with high LTBP2 expression. CONCLUSION: LTBP2 could be used as a prognostic marker, and high LTBP2 expression related to abundant TAMs infiltration and with a worse response to chemotherapy.


Asunto(s)
Neoplasias Encefálicas , Biología Computacional , Glioma , Proteínas de Unión a TGF-beta Latente , Humanos , Glioma/genética , Glioma/patología , Glioma/inmunología , Glioma/metabolismo , Animales , Ratones , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Biología Computacional/métodos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Femenino , Línea Celular Tumoral , Progresión de la Enfermedad , Masculino , Regulación Neoplásica de la Expresión Génica , Pronóstico , Proliferación Celular , Persona de Mediana Edad
8.
Ophthalmic Res ; 67(1): 340-347, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38772353

RESUMEN

INTRODUCTION: Weill-Marchesani syndrome (WMS) is a hereditary connective tissue disorder with substantial heterogeneity in clinical features and genetic etiology, so it is essential to define the full mutation spectrum for earlier diagnosis. In this study, we report Weill-Marchesani-like syndrome (WMS-like) change to autosomal dominance inheritance caused by novel haplotypic mutations in latent transforming growth factor beta-binding protein 2 (LTBP2). METHODS: Twenty-five members from a 4-generation Chinese family were recruited from Guangzhou, of whom nine were diagnosed with WMS-like disease, nine were healthy, and seven were of "uncertain" clinical status because of their young age. All members received detailed physical and ocular examinations. Whole-exome sequencing, Sanger sequencing, and real-time PCR were used to identify and verify the causative mutations in family members. RESULTS: Genetic sequencing revealed novel haplotypic mutations on the same LTBP2 chromosome associated with WMS-like, c. 2657C>A/p.T886K in exon 16 and deletion of exons 25-36. Real-time PCR and Sanger sequencing verified both mutations in patients with clinically diagnosed WMS-like, and in one "uncertain" child. In these patients, the haplotypic mutations led to ectopia lentis, short stature, and obesity. CONCLUSION: Our study revealed that WMS-like may be associated with haplotypic LTBP2 mutations with autosomal dominant inheritance.


Asunto(s)
Proteínas de Unión a TGF-beta Latente , Mutación , Linaje , Síndrome de Weill-Marchesani , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , China/epidemiología , Análisis Mutacional de ADN , Pueblos del Este de Asia/genética , Secuenciación del Exoma , Haplotipos , Proteínas de Unión a TGF-beta Latente/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Síndrome de Weill-Marchesani/genética , Síndrome de Weill-Marchesani/diagnóstico
9.
Am J Hum Genet ; 106(4): 559-569, 2020 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-32197075

RESUMEN

Negative regulator of reactive oxygen species (NRROS) is a leucine-rich repeat-containing protein that uniquely associates with latent transforming growth factor beta-1 (TGF- ß1) and anchors it on the cell surface; this anchoring is required for activation of TGF-ß1 in macrophages and microglia. We report six individuals from four families with bi-allelic variants in NRROS. All affected individuals had neurodegenerative disease with refractory epilepsy, developmental regression, and reduced white matter volume with delayed myelination. The clinical course in affected individuals began with normal development or mild developmental delay, and the onset of seizures occurred within the first year of life, followed by developmental regression. Intracranial calcification was detected in three individuals. The phenotypic features in affected individuals are consistent with those observed in the Nrros knockout mouse, and they overlap with those seen in the human condition associated with TGF-ß1 deficiency. The disease-causing NRROS variants involve two significant functional NRROS domains. These variants result in aberrant NRROS proteins with impaired ability to anchor latent TGF-ß1 on the cell surface. Using confocal microscopy in HEK293T cells, we demonstrate that wild-type and mutant NRROS proteins co-localize with latent TGF-ß1 intracellularly. However, using flow cytometry, we show that our mutant NRROS proteins fail to anchor latent TGF-ß1 at the cell surface in comparison to wild-type NRROS. Moreover, wild-type NRROS rescues the defect of our disease-associated mutants in presenting latent TGF-ß1 to the cell surface. Taken together, our findings suggest that loss of NRROS function causes a severe childhood-onset neurodegenerative condition with features suggestive of a disordered response to inflammation.


Asunto(s)
Encefalopatías/genética , Calcinosis/genética , Variación Genética/genética , Proteínas de Unión a TGF-beta Latente/genética , Enfermedades Neurodegenerativas/genética , Factor de Crecimiento Transformador beta1/genética , Alelos , Femenino , Células HEK293 , Humanos , Lactante , Macrófagos/patología , Masculino , Microglía/patología
10.
Mol Vis ; 29: 169-179, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38222456

RESUMEN

Purpose: To evaluate the frequency of LTBP2 mutations and to elaborate on LTBP2-related clinical phenotypes in a Chinese congenital ectopia lentis (CEL) cohort. Methods: In total, 145 Chinese probands with CEL were recruited for this study and underwent ocular and systemic examinations. Whole-exome sequencing was used to identify mutations, and Sanger sequencing and bioinformatics analysis were further performed to verify pathogenic mutations. Results: Overall, biallelic mutations in LTBP2 involving eight novel mutations (c.4370-7_4370-9delTCT, c.4370-5C>G, c.3452G>A, c.2253delG, c.4114T>C, c.1251G>A, c.4760G>A, and c.620G>A) were identified in four CEL probands (4/145, 2.76%). Patients with LTBP2 mutations were characterized by a megalocornea, spherophakia, high myopia, and glaucoma instead of a flat cornea, high corneal astigmatism, cardiovascular and skeletal abnormalities that were reported in other gene mutations. A novel homozygous frameshift mutation was detected, and this type of mutation was found to cause more complicated ocular symptoms than others, ranging from the anterior segment to the fundus. Conclusion: This study reported the mutation frequency of the LTBP2 gene in a Chinese CEL cohort and provided novel insight into LTBP2-related genotype-phenotype associations in CEL.


Asunto(s)
Enfermedades de la Córnea , Desplazamiento del Cristalino , Glaucoma , Proteínas de Unión a TGF-beta Latente , Humanos , China , Enfermedades de la Córnea/genética , Análisis Mutacional de ADN , Desplazamiento del Cristalino/genética , Desplazamiento del Cristalino/complicaciones , Glaucoma/genética , Proteínas de Unión a TGF-beta Latente/genética , Mutación , Linaje , Fenotipo
11.
Exp Eye Res ; 234: 109606, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37506754

RESUMEN

Weill-Marchesani syndrome (WMS) manifests as ectopia lentis (EL), microspherophakia and short stature, which is caused by ADAMTS10, LTBP2, or ADAMTS17 gene defects. This study aims to investigate the characteristics and genotype-phenotype correlations of WMS with ADAMTS17 mutations. WMS patients with ADAMTS17 variants were identified by whole-exome sequencing from 185 patients with EL. All the included patients underwent comprehensive ocular and systemic examinations. ADAMTS17 variants were reviewed from included patients, published literature, and public databases. Bioinformatics analysis, co-segregation analysis, species sequence analysis, and protein silico modeling were used to verify the pathogenic mutations. A total of six novel ADAMTS17 mutations (c.1297C > T, c.2948C > T, c.1322+2T > C, c.1716C > G, c.1630G > A, and c.1669C > T) were identified in four WMS probands in our EL cohort (4/185, 2.16%). All probands and their biological parents presented with apparent short stature compared with the standard value. In particular, one child was detected with valvular heart disease, which has not previously been reported in patients with ADAMTS17 mutations. Conserved residues were greatly affected by the substitution of amino acids caused by these six mutations. Short stature could be considered a clue for EL patients with ADAMTS17 mutations, and much more attention needs to be paid to heart disorders among these patients. This study not only reported the characteristics of ADAMTS17 mutation-related WMS but also helped to recognize the genotype-phenotype correlations in these patients.


Asunto(s)
Desplazamiento del Cristalino , Glaucoma , Síndrome de Weill-Marchesani , Humanos , Síndrome de Weill-Marchesani/genética , Mutación , Desplazamiento del Cristalino/genética , Estudios de Asociación Genética , Proteínas ADAMTS/genética , Proteínas de Unión a TGF-beta Latente/genética
12.
Eur Heart J ; 43(17): 1668-1680, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35245370

RESUMEN

AIMS: Mitral valve prolapse (MVP) is a common valvular heart disease with a prevalence of >2% in the general adult population. Despite this high incidence, there is a limited understanding of the molecular mechanism of this disease, and no medical therapy is available for this disease. We aimed to elucidate the genetic basis of MVP in order to better understand this complex disorder. METHODS AND RESULTS: We performed a meta-analysis of six genome-wide association studies that included 4884 cases and 434 649 controls. We identified 14 loci associated with MVP in our primary analysis and 2 additional loci associated with a subset of the samples that additionally underwent mitral valve surgery. Integration of epigenetic, transcriptional, and proteomic data identified candidate MVP genes including LMCD1, SPTBN1, LTBP2, TGFB2, NMB, and ALPK3. We created a polygenic risk score (PRS) for MVP and showed an improved MVP risk prediction beyond age, sex, and clinical risk factors. CONCLUSION: We identified 14 genetic loci that are associated with MVP. Multiple analyses identified candidate genes including two transforming growth factor-ß signalling molecules and spectrin ß. We present the first PRS for MVP that could eventually aid risk stratification of patients for MVP screening in a clinical setting. These findings advance our understanding of this common valvular heart disease and may reveal novel therapeutic targets for intervention.


Asunto(s)
Prolapso de la Válvula Mitral , Adulto , Sitios Genéticos/genética , Estudio de Asociación del Genoma Completo , Humanos , Proteínas de Unión a TGF-beta Latente/genética , Prolapso de la Válvula Mitral/genética , Proteómica , Factores de Riesgo
13.
Dev Dyn ; 251(1): 95-104, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33742701

RESUMEN

The purpose of this review is to discuss the transforming growth factor beta (TGFB) binding proteins (LTBP) with respect to their participation in the activity of TGFB. We first describe pertinent aspects of the biology and cell function of the LTBPs. We then summarize the physiological consequences of LTBP loss in humans and mice. Finally, we consider a number of outstanding questions relating to LTBP function.


Asunto(s)
Proteínas de Unión a TGF-beta Latente , Animales , Humanos , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Ratones , Unión Proteica , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
14.
Harefuah ; 162(6): 352-358, 2023 Jun.
Artículo en Hebreo | MEDLINE | ID: mdl-37394436

RESUMEN

BACKGROUND: Short stature is a common finding among the general population, mostly presented as an isolated phenotype. The syndromic short statute is rare and complex. Recently, we examined several patients from related families sharing both short stature and congenital dental abnormalities. OBJECTIVES: 1. Clinical characterization of syndromic short stature; 2. To find the disease mutation and evaluate the carrier state in the particular community. METHODS: Clinical characterization- by medical history, medical records and physical examination; Homozygosity mapping - by using the Single nucleotide polymorphism (SNP) chromosomal microarrays (CMA) analysis and gene mutation detection by ABI Sanger sequence. RESULTS: All patients present with short stature severe dental anomalies including enamel formation and mineralization defect, oligodontia, abnormal shape and retarded eruption. CMA analysis in 3 patients and 2 healthy members of four families was normal. One homozygote region in chromosome 11 (11p11.2- 11q13.3) was found in all patients. By using the candidate gene approach, amongst the 301 genes found within this region, only one, the LTBP3 gene (Latent Transforming Growth Factor-Beta-Binding Protein-3) has high priority for sequence. Hence, LTBP3 (OMIM-602090) pathogenic variant is responsible for "brachyolmia with amelogenesis imperfecta" also known as "Dental Anomalies and Short Stature (DASS)" (OMIM- 601216). We sequenced all 29 LTBP3 exons and a novel splice pathogenic variant, c.1346-1G>A chr11:65319629, in exon 8 was identified. The variant segregated well within healthy tested family members. We found a high carrier rate in the village (1:15). CONCLUSIONS: We identified a novel and common LTBP3 gene pathogenic variant responsible for short stature, brachyolmia and amelogenesis imperfecta in Druze Arab patients.


Asunto(s)
Amelogénesis Imperfecta , Osteocondrodisplasias , Humanos , Amelogénesis Imperfecta/genética , Amelogénesis Imperfecta/patología , Árabes , Mutación , Osteocondrodisplasias/genética , Proteínas de Unión a TGF-beta Latente/genética
15.
Dev Biol ; 470: 95-107, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33245870

RESUMEN

During heart formation, the heart grows and undergoes dramatic morphogenesis to achieve efficient embryonic function. Both in fish and amniotes, much of the growth occurring after initial heart tube formation arises from second heart field (SHF)-derived progenitor cell addition to the arterial pole, allowing chamber formation. In zebrafish, this process has been extensively studied during embryonic life, but it is unclear how larval cardiac growth occurs beyond 3 days post-fertilisation (dpf). By quantifying zebrafish myocardial growth using live imaging of GFP-labelled myocardium we show that the heart grows extensively between 3 and 5 dpf. Using methods to assess cell division, cellular development timing assay and Kaede photoconversion, we demonstrate that proliferation, CM addition, and hypertrophy contribute to ventricle growth. Mechanistically, we show that reduction in Mef2c activity (mef2ca+/-;mef2cb-/-), downstream or in parallel with Nkx2.5 and upstream of Ltbp3, prevents some CM addition and differentiation, resulting in a significantly smaller ventricle by 3 dpf. After 3 dpf, however, CM addition in mef2ca+/-;mef2cb-/- mutants recovers to a normal pace, and the heart size gap between mutants and their siblings diminishes into adulthood. Thus, as in mice, there is an early time window when SHF contribution to the myocardium is particularly sensitive to loss of Mef2c activity.


Asunto(s)
Ventrículos Cardíacos/embriología , Corazón/embriología , Factores de Transcripción MEF2/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Factores de Transcripción MEF2/genética , Proteínas Musculares/genética , Mutación , Tamaño de los Órganos , Organogénesis , Pez Cebra , Proteínas de Pez Cebra/genética
16.
J Biol Chem ; 297(3): 101055, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34411563

RESUMEN

Fibrillin-1 (FBN1) is the major component of extracellular matrix microfibrils, which are required for proper development of elastic tissues, including the heart and lungs. Through protein-protein interactions with latent transforming growth factor (TGF) ß-binding protein 1 (LTBP1), microfibrils regulate TGF-ß signaling. Mutations within the 47 epidermal growth factor-like (EGF) repeats of FBN1 cause autosomal dominant disorders including Marfan Syndrome, which is characterized by disrupted TGF-ß signaling. We recently identified two novel protein O-glucosyltransferases, Protein O-glucosyltransferase 2 (POGLUT2) and 3 (POGLUT3), that modify a small fraction of EGF repeats on Notch. Here, using mass spectral analysis, we show that POGLUT2 and POGLUT3 also modify over half of the EGF repeats on FBN1, fibrillin-2 (FBN2), and LTBP1. While most sites are modified by both enzymes, some sites show a preference for either POGLUT2 or POGLUT3. POGLUT2 and POGLUT3 are homologs of POGLUT1, which stabilizes Notch proteins by addition of O-glucose to Notch EGF repeats. Like POGLUT1, POGLUT2 and 3 can discern a folded versus unfolded EGF repeat, suggesting POGLUT2 and 3 are involved in a protein folding pathway. In vitro secretion assays using the N-terminal portion of recombinant FBN1 revealed reduced FBN1 secretion in POGLUT2 knockout, POGLUT3 knockout, and POGLUT2 and 3 double-knockout HEK293T cells compared with wild type. These results illustrate that POGLUT2 and 3 function together to O-glucosylate protein substrates and that these modifications play a role in the secretion of substrate proteins. It will be interesting to see how disease variants in these proteins affect their O-glucosylation.


Asunto(s)
Fibrilina-1/metabolismo , Fibrilina-2/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , Síndrome de Marfan/metabolismo , Secuencias de Aminoácidos , Fibrilina-1/química , Fibrilina-1/genética , Fibrilina-2/química , Fibrilina-2/genética , Glicosilación , Humanos , Proteínas de Unión a TGF-beta Latente/química , Proteínas de Unión a TGF-beta Latente/genética , Síndrome de Marfan/enzimología , Síndrome de Marfan/genética , Sistemas de Translocación de Proteínas , Transducción de Señal
17.
Clin Genet ; 102(1): 66-71, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35352826

RESUMEN

Mutations in LTBP3 are associated with Dental Anomalies and Short Stature syndrome (DASS; MIM 601216), which is characterized by hypoplastic type amelogenesis imperfecta, hypodontia, underdeveloped maxilla, short stature, brachyolmia, aneurysm and dissection of the thoracic aorta. Here we report a novel (p.Arg545ProfsTer22) and a recurrent (c.3107-2A > G) LTBP3 variants, in a Turkish family affected with DASS. The proband, who carried compound heterozygous variant c.3107-2A > G, p.Arg545ProfsTer22, was most severely affected with DASS. The proband's father, who carried the heterozygous variant c.3107-2A > G had short stature and prognathic mandible. The mother and brother of the proband carried the heterozygous variant p.Arg545ProfsTer22, but only the mother showed any DASS characteristics. The c.3107-2A > G and the p.Arg545ProfsTer22 variants are expected to result in abnormal LTPB3 protein, failure of TGFß-LAP-LTBP3 complex formation, and subsequent disruption of TGFß secretion and activation. This is the first report of heterozygous carriers of LTBP3 variants showing phenotypes. The new findings of DASS found in this family include taurodontism, single-rooted molars, abnormal dentin, calcified dental pulp blood vessels, prognathic mandible, failure of mandibular tooth eruption, interatrial septal aneurysm, secundum atrial septal defect, tricuspid valve prolapse, and a recurrent glenohumeral joint dislocation.


Asunto(s)
Amelogénesis Imperfecta , Enanismo , Osteocondrodisplasias , Anomalías Dentarias , Amelogénesis Imperfecta/genética , Enanismo/genética , Humanos , Proteínas de Unión a TGF-beta Latente/genética , Masculino , Osteocondrodisplasias/genética , Fenotipo , Anomalías Dentarias/genética , Factor de Crecimiento Transformador beta/genética
18.
Am J Med Genet A ; 188(11): 3343-3349, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35972031

RESUMEN

Cutis laxa (CL) is a rare connective tissue disorder characterized by wrinkled, abundant and sagging skin, sometimes associated with systemic impairment. Biallelic alterations in latent transforming growth factor beta-binding protein 4 gene (LTBP4) cause autosomal recessive type 1C cutis laxa (ARCL1C, MIM #613177). The present report describes the case of a 17-months-old girl with cutis laxa together with a literature review of previous ARCL1C cases. Based on proband main clinical signs (cutis laxa and pulmonary emphysema), clinical exome sequencing (CES) was performed and showed a new nine base-pairs homozygous in-frame deletion in LTBP4 gene. RT-PCR and cDNA Sanger sequencing were performed in order to clarify its impact on RNA. This report demonstrates that a genetic alteration in the EGF-like 14 domain calcium-binding motif of LTBP4 gene is likely responsible for cutis laxa in our patient.


Asunto(s)
Cutis Laxo , Calcio , Enfermedades de los Cartílagos , Cutis Laxo/genética , ADN Complementario , Factor de Crecimiento Epidérmico , Femenino , Enfermedades Gastrointestinales , Humanos , Lactante , Proteínas de Unión a TGF-beta Latente/genética , ARN , Enfermedades Respiratorias , Factor de Crecimiento Transformador beta , Enfermedades Urológicas
19.
Mol Cell Biochem ; 477(6): 1669-1679, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35235125

RESUMEN

Dysregulated circRNAs have potential roles in the progression of various cancer types, including cervical cancer (CaCx). The carcinogenic roles of circRNA Wolf-Hirschhorn syndrome candidate gene-1 (circWHSC1) are described in the development of diverse cancers. The objective of this study was to investigate the expression and the underlying role of circWHSC1 in CaCx. The expression of circWHSC1 was detected by real-time PCR. After the suppression of circWHSC1 expression, the changes in the proliferation, migration, invasion, and apoptosis capacities were detected by CCK-8 assay, colony formation assay, Transwell assays, flow cytometry, and the determination of apoptosis-related proteins. The interplay among circWHSC1, miR-532-3p, and latent transforming growth factor-ß binding protein 2 (LTBP2) was confirmed by luciferase reporter and biotinylated RNA pull-down assays. A nude mice xenograft tumor model was established to evaluate the anti-tumorigenic role of circWHSC1 silencing in vivo. CircWHSC1 was overexpressed in CaCx tissues and cell lines and its high expression was inversely associated with the survival rate of patients with CaCx. CircWHSC1 silencing was capable of suppressing the proliferation, metastasis, and invasion of tumor cells and inducing apoptosis. Investigation to its molecular mechanism revealed that circWHSC1 functioned as a competitive endogenous RNA (ceRNA), mediating LTBP2 expression by targeting miR-532-3p. The in vivo experiments further confirmed the inhibition of tumor growth and metastasis by circWHSC1 knockdown. The circWHSC1-mediated miR-532-3p/LTBP2 signaling axis might be a novel therapeutic target for CaCx.


Asunto(s)
MicroARNs , Neoplasias del Cuello Uterino , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , Neoplasias del Cuello Uterino/genética
20.
Mol Biol Rep ; 49(5): 4135-4140, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35445908

RESUMEN

BACKGROUND: Autosomal recessive cutis laxa (ARCL) is a heterogeneous disorder with three primary forms (ARCL 1, ARCL 2 and ARCL 3). Latent transforming growth factor beta binding protein 4 (LTBP4) anomalies cause ARCL1C and are connected to different problems in the skin and other organs. Herein, we present a seven month old Iranian boy with a clinical manifestation of ARCL1 with literature review of previous cases with attributes of ARCL1C. METHODS: Considering the craniofacial characteristics and respiratory distress of the proband, cutis laxa (CL) was expected and whole-exome sequencing (WES) was performed. RESULTS: In the proband, signs of CL were mainly located in the face, thorax, and abdomen. The prenatal investigation revealed a diaphragmatic hernia and certain uncommon signs, such as an atrial septal defect and pyloric stenosis. The WES showed a novel homozygous mutation (c.533-1G > A) in exon six of the LTBP4 gene. CONCLUSION: This report showed a new variant with uncommon clinical features, such as a stenosis atrial septal defect and pyloric stenosis, which causes ARCL1C. Unfortunately, the proband developed several heart problems and died at the age of seven months and seven days. Thus, a more in-depth evaluation is needed to clarify the different aspects of CL related to LTBP4 disorder.


Asunto(s)
Cutis Laxo , Defectos del Tabique Interatrial , Estenosis Pilórica , Enfermedades de los Cartílagos , Cutis Laxo/genética , Enfermedades Gastrointestinales , Humanos , Lactante , Irán , Proteínas de Unión a TGF-beta Latente/genética , Masculino , Enfermedades Respiratorias , Enfermedades Urológicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA