Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
1.
EMBO J ; 43(12): 2308-2336, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38760574

RESUMEN

How cells coordinate morphogenetic cues and fate specification during development remains a fundamental question in organogenesis. The mammary gland arises from multipotent stem cells (MaSCs), which are progressively replaced by unipotent progenitors by birth. However, the lack of specific markers for early fate specification has prevented the delineation of the features and spatial localization of MaSC-derived lineage-committed progenitors. Here, using single-cell RNA sequencing from E13.5 to birth, we produced an atlas of matched mouse mammary epithelium and mesenchyme and reconstructed the differentiation trajectories of MaSCs toward basal and luminal fate. We show that murine MaSCs exhibit lineage commitment just prior to the first sprouting events of mammary branching morphogenesis at E15.5. We identify early molecular markers for committed and multipotent MaSCs and define their spatial distribution within the developing tissue. Furthermore, we show that the mammary embryonic mesenchyme is composed of two spatially restricted cell populations, and that dermal mesenchyme-produced FGF10 is essential for embryonic mammary branching morphogenesis. Altogether, our data elucidate the spatiotemporal signals underlying lineage specification of multipotent MaSCs, and uncover the signals from mesenchymal cells that guide mammary branching morphogenesis.


Asunto(s)
Linaje de la Célula , Células Epiteliales , Glándulas Mamarias Animales , Células Madre Mesenquimatosas , Animales , Ratones , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/metabolismo , Femenino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Diferenciación Celular , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Morfogénesis , Análisis de la Célula Individual , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/embriología
2.
Int Immunopharmacol ; 134: 112165, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38692017

RESUMEN

Particulate matter (PM) is considered the fundamental component of atmospheric pollutants and is associated with the pathogenesis of many respiratory diseases. Fibroblast growth factor 10 (FGF10) mediates mesenchymal-epithelial signaling and has been linked with the repair process of PM-induced lung injury (PMLI). However, the pathogenic mechanism of PMLI and the specific FGF10 protective mechanism against this injury are still undetermined. PM was administered in vivo into murine airways or in vitro to human bronchial epithelial cells (HBECs), and the inflammatory response and ferroptosis-related proteins SLC7A11 and GPX4 were assessed. The present research investigates the FGF10-mediated regulation of ferroptosis in PMLI mice models in vivo and HBECs in vitro. The results showed that FGF10 pretreatment reduced PM-mediated oxidative damage and ferroptosis in vivo and in vitro. Furthermore, FGF10 pretreatment led to reduced oxidative stress, decreased secretion of inflammatory mediators, and activation of the Nrf2-dependent antioxidant signaling. Additionally, silencing of Nrf2 using siRNA in the context of FGF10 treatment attenuated the effect on ferroptosis. Altogether, both in vivo and in vitro assessments confirmed that FGF10 protects against PMLI by inhibiting ferroptosis via the Nrf2 signaling. Thus, FGF10 can be used as a novel ferroptosis suppressor and a potential treatment target in PMLI.


Asunto(s)
Ferroptosis , Factor 10 de Crecimiento de Fibroblastos , Lesión Pulmonar , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Material Particulado , Transducción de Señal , Ferroptosis/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Material Particulado/toxicidad , Humanos , Transducción de Señal/efectos de los fármacos , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Ratones , Estrés Oxidativo/efectos de los fármacos , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Masculino , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Línea Celular , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Modelos Animales de Enfermedad , Sistema de Transporte de Aminoácidos y+
3.
Proc Natl Acad Sci U S A ; 121(11): e2314911121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38442169

RESUMEN

In amniote limbs, Fibroblast Growth Factor 10 (FGF10) is essential for limb development, but whether this function is broadly conserved in tetrapods and/or involved in adult limb regeneration remains unknown. To tackle this question, we established Fgf10 mutant lines in the newt Pleurodeles waltl which has amazing regenerative ability. While Fgf10 mutant forelimbs develop normally, the hindlimbs fail to develop and downregulate FGF target genes. Despite these developmental defects, Fgf10 mutants were able to regenerate normal hindlimbs rather than recapitulating the embryonic phenotype. Together, our results demonstrate an important role for FGF10 in hindlimb formation, but little or no function in regeneration, suggesting that different mechanisms operate during limb regeneration versus development.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos , Animales , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Miembro Posterior/crecimiento & desarrollo , Regeneración , Pleurodeles/genética , Pleurodeles/crecimiento & desarrollo , Pleurodeles/metabolismo
4.
Genetics ; 227(1)2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38386912

RESUMEN

Vertebrate limbs start to develop as paired protrusions from the lateral plate mesoderm at specific locations of the body with forelimb buds developing anteriorly and hindlimb buds posteriorly. During the initiation process, limb progenitor cells maintain active proliferation to form protrusions and start to express Fgf10, which triggers molecular processes for outgrowth and patterning. Although both processes occur in both types of limbs, forelimbs (Tbx5), and hindlimbs (Isl1) utilize distinct transcriptional systems to trigger their development. Here, we report that Sall1 and Sall4, zinc finger transcription factor genes, regulate hindlimb initiation in mouse embryos. Compared to the 100% frequency loss of hindlimb buds in TCre; Isl1 conditional knockouts, Hoxb6Cre; Isl1 conditional knockout causes a hypomorphic phenotype with only approximately 5% of mutants lacking the hindlimb. Our previous study of SALL4 ChIP-seq showed SALL4 enrichment in an Isl1 enhancer, suggesting that SALL4 acts upstream of Isl1. Removing 1 allele of Sall4 from the hypomorphic Hoxb6Cre; Isl1 mutant background caused loss of hindlimbs, but removing both alleles caused an even higher frequency of loss of hindlimbs, suggesting a genetic interaction between Sall4 and Isl1. Furthermore, TCre-mediated conditional double knockouts of Sall1 and Sall4 displayed a loss of expression of hindlimb progenitor markers (Isl1, Pitx1, Tbx4) and failed to develop hindlimbs, demonstrating functional redundancy between Sall1 and Sall4. Our data provides genetic evidence that Sall1 and Sall4 act as master regulators of hindlimb initiation.


Asunto(s)
Proteínas de Unión al ADN , Regulación del Desarrollo de la Expresión Génica , Miembro Posterior , Proteínas con Homeodominio LIM , Factores de Transcripción , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ratones , Miembro Posterior/embriología , Miembro Posterior/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Esbozos de los Miembros/metabolismo , Esbozos de los Miembros/embriología , Ratones Noqueados , Embrión de Mamíferos/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo
5.
Orthod Craniofac Res ; 27(1): 84-94, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37452556

RESUMEN

OBJECTIVE: Dysregulation of Fibroblast Growth Factor 10 (FGF10), a member of the family of Fibroblast Growth Factor (FGF) proteins, has been implicated in craniofacial and dental anomalies, including craniosynostosis, cleft palate, and Lacrimo-Auriculo-Dento-Digital Syndrome. The aim of this murine study was to assess the craniofacial and dental phenotypes associated with a heterozygous FGF10 gene (FGF10+/- ) mutation at skeletal maturity. METHODS: Skulls of 40 skeletally mature mice, comprising two genotypes (heterozygous FGF10+/- mutation, n = 22; wildtype, n = 18) and two sexes (male, n = 23; female, n = 17), were subjected to micro-computed tomography. Landmark-based linear dimensions were measured for the cranial vault, maxilla, mandible, and first molar teeth. Multivariate analysis of variance was performed to assess whether there were significant differences in the craniofacial and dental structures between genotypes and sexes. RESULTS: The craniomaxillary skeleton and the first molar teeth were smaller in the FGF10+/- mice (P < .05), but the mandible was unaffected. Sex did not have a significant effect on these structures (P > .05). Cranial sutural defects were noted in 5/22 (22.7%) mutant versus 2/18 (11.1%) wildtype mice, and cleft palate in only one (4.5%) mutant mouse. None of the mice displayed craniosynostosis, expansive bony lesions, bifid condyles, or impacted teeth. CONCLUSION: The FGF10+/- mutation was associated with craniomaxillary skeletal hypoplasia that probably arose from deficient (delayed) intramembranous ossification of the sutured bones. Overall, the skeletal and dental data suggest that the FGF10 gene plays an important role in the aetiology of craniofacial dysmorphology and malocclusion.


Asunto(s)
Fisura del Paladar , Anomalías Craneofaciales , Craneosinostosis , Ratones , Masculino , Femenino , Animales , Fisura del Paladar/genética , Microtomografía por Rayos X , Factor 10 de Crecimiento de Fibroblastos/genética , Modelos Animales de Enfermedad , Anomalías Craneofaciales/diagnóstico por imagen , Anomalías Craneofaciales/genética , Craneosinostosis/genética , Mutación/genética
6.
Pediatr Pulmonol ; 58(11): 3095-3105, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37560881

RESUMEN

INTRODUCTION: Fibroblast growth factor 10 (FGF10) is a signaling molecule with a well-established role for lung branching morphogenesis. Rare heterozygous, deleterious variants in the FGF10 gene are known causes of the lacrimo-auriculo-dento-digital (LADD) syndrome and aplasia of lacrimal and salivary glands. Previous studies indicate that pathogenic variants in FGF10 can cause childhood Interstitial Lung Disease (chILD) due to severe diffuse developmental disorders of the lung, but detailed reports on clinical presentation and follow-up of affected children are lacking. METHODS: We describe four children with postnatal onset of chILD and heterozygous variants in FGF10, each detected by exome or whole genome sequencing. RESULTS: All children presented with postnatal respiratory failure. Two children died within the first 2 days of life, one patient died at age of 12 years due to right heart failure related to severe pulmonary hypertension (PH) and one patient is alive at age of 6 years, but still symptomatic. Histopathological analysis of lung biopsies from the two children with early postpartum demise revealed diffuse developmental disorder representing acinar dysplasia and interstitial fibrosis. Sequential biopsies of the child with survival until the age of 12 years revealed alveolar simplification and progressive interstitial fibrosis. DISCUSSION: Our report extends the phenotype of FGF10-related disorders to early onset chILD with progressive interstitial lung fibrosis and PH. Therefore, FGF10-related disorder should be considered even without previously described syndromic stigmata in children with postnatal respiratory distress, not only when leading to death in the neonatal period but also in case of persistent respiratory complaints and PH.


Asunto(s)
Enfermedades del Aparato Lagrimal , Enfermedades Pulmonares Intersticiales , Niño , Humanos , Recién Nacido , Factor 10 de Crecimiento de Fibroblastos/genética , Fibrosis , Enfermedades del Aparato Lagrimal/genética , Pulmón , Enfermedades Pulmonares Intersticiales/genética
7.
J Endocrinol ; 259(1)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37417397

RESUMEN

Acute lung injury (ALI) is associated with an increased incidence of respiratory diseases, which are devastating clinical disorders with high global mortality and morbidity. Evidence confirms that fibroblast growth factors (FGFs) play key roles in mediating ALI. Mice were treated with LPS (lipopolysaccharide: 5 mg/kg, intratracheally) to establish an in vivo ALI model. Human lung epithelial BEAS-2B cells cultured in a corresponding medium with LPS were used to mimic the ALI model in vitro. In this study, we characterized FGF10 pretreatment (5 mg/kg, intratracheally) which improved LPS-induced ALI, including histopathological changes, and reduced pulmonary edema. At the cellular level, FGF10 pretreatment (10 ng/mL) alleviated LPS-induced ALI accompanied by reduced reactive oxygen species (ROS) accumulation and inflammatory responses, such as IL-1ß, IL-6, and IL-10, as well as suppressed excessive autophagy. Additionally, immunoblotting and co-immunoprecipitation showed that FGF10 activated nuclear factor erythroid-2-related factor 2 (Nrf2) signaling pathway via Nrf2 nuclear translocation by promoting the interaction between p62 and keap1, thereby preventing LPS-induced ALI. Nrf2 knockout significantly reversed these protective effects of FGF10. Together, FGF10 protects against LPS-induced ALI by restraining autophagy via p62-Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 signaling pathway, implying that FGF10 could be a novel therapy for ALI.


Asunto(s)
Lesión Pulmonar Aguda , Factor 2 Relacionado con NF-E2 , Ratones , Humanos , Animales , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/farmacología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Lipopolisacáridos/toxicidad , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Autofagia , Pulmón/metabolismo , Pulmón/patología
8.
J Biol Chem ; 299(1): 102787, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36509141

RESUMEN

Chemoresistance remains a major challenge in the current treatment of acute myeloid leukemia (AML). The bone marrow microenvironment (BMM) plays a complex role in protecting leukemia cells from chemotherapeutics, and the mechanisms involved are not fully understood. Antileukemia drugs kill AML cells directly but also damage the BMM. Here, we determined antileukemia drugs induce DNA damage in bone marrow stromal cells (BMSCs), resulting in resistance of AML cell lines to adriamycin and idarubicin killing. Damaged BMSCs induced an inflammatory microenvironment through NF-κB; suppressing NF-κB with small molecule inhibitor Bay11-7082 attenuated the prosurvival effects of BMSCs on AML cell lines. Furthermore, we used an ex vivo functional screen of 507 chemokines and cytokines to identify 44 proteins secreted from damaged BMSCs. Fibroblast growth factor-10 (FGF10) was most strongly associated with chemoresistance in AML cell lines. Additionally, expression of FGF10 and its receptors, FGFR1 and FGFR2, was increased in AML patients after chemotherapy. FGFR1 and FGFR2 were also widely expressed by AML cell lines. FGF10-induced FGFR2 activation in AML cell lines operates by increasing P38 MAPK, AKT, ERK1/2, and STAT3 phosphorylation. FGFR2 inhibition with small molecules or gene silencing of FGFR2 inhibited proliferation and reverses drug resistance of AML cells by inhibiting P38 MAPK, AKT, and ERK1/2 signaling pathways. Finally, release of FGF10 was mediated by ß-catenin signaling in damaged BMSCs. Our data indicate FGF10-FGFR2 signaling acts as an effector of damaged BMSC-mediated chemoresistance in AML cells, and FGFR2 inhibition can reverse stromal protection and AML cell chemoresistance in the BMM.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia Mieloide Aguda , Células Madre Mesenquimatosas , Humanos , Células de la Médula Ósea/metabolismo , Daño del ADN , Factor 10 de Crecimiento de Fibroblastos/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Células Madre Mesenquimatosas/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral , Comunicación Paracrina
9.
Int Immunopharmacol ; 113(Pt A): 109398, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36461597

RESUMEN

Particulate matter (PM) is a major environmental contaminant that causes and worsens respiratory diseases. Fibroblast growth factor 10 (FGF10), a paracrine fibroblast growth factor that specifically stimulates repair and regeneration after injury, has been shown to protect against PM-induced lung injury. However, the underlying mechanisms are still unclear. In this study, the protective effects of FGF10 were investigated using a PM-induced lung injury mouse model in vivo and BEAS-2B cells in vitro. According to the findings, FGF10 treatment alleviated PM-induced oxidative damage and pyroptosis in vivo and in vitro. Mechanistically, FGF10 activated antioxidative Nrf2 signaling. Inhibition of PI3K signaling with LY294002 or Nrf2 signaling with ML385 revealed that FGF10-mediated lung protection was mediated by the PI3K/Akt/Nrf2 pathway. These results collectively indicate that FGF10 inhibits oxidative stress-mediated pyroptosis via the PI3K/Akt/Nrf2 pathway, suggesting a possible therapy for PM-induced lung injury.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos , Lesión Pulmonar , Material Particulado , Piroptosis , Animales , Ratones , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/inmunología , Lesión Pulmonar/etiología , Lesión Pulmonar/genética , Lesión Pulmonar/inmunología , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo/genética , Estrés Oxidativo/inmunología , Material Particulado/toxicidad , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Piroptosis/genética , Piroptosis/inmunología , Transducción de Señal
10.
PeerJ ; 10: e14003, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36124135

RESUMEN

FGF10, as an FGFR2b-specific ligand, plays a crucial role during cell proliferation, multi-organ development, and tissue injury repair. The developmental importance of FGF10 has been emphasized by the identification of FGF10 abnormalities in human congenital disorders affecting different organs and systems. Single-nucleotide variants in FGF10 or FGF10-involving copy-number variant deletions have been reported in families with lacrimo-auriculo-dento-digital syndrome, aplasia of the lacrimal and salivary glands, or lethal lung developmental disorders. Abnormalities involving FGF10 have also been implicated in cleft lip and palate, myopia, or congenital heart disease. However, the exact developmental role of FGF10 and large phenotypic heterogeneity associated with FGF10 disruption remain incompletely understood. Here, we review human and animal studies and summarize the data on FGF10 mechanism of action, expression, multi-organ function, as well as its variants and their usefulness for clinicians and researchers.


Asunto(s)
Labio Leporino , Fisura del Paladar , Enfermedades del Aparato Lagrimal , Aparato Lagrimal , Enfermedades Pulmonares , Sindactilia , Animales , Humanos , Aparato Lagrimal/anomalías , Factor 10 de Crecimiento de Fibroblastos/genética
11.
Stem Cells ; 40(6): 605-617, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35437594

RESUMEN

Bronchopulmonary dysplasia (BPD) is a neonatal lung disease developing in premature babies characterized by arrested alveologenesis and associated with decreased Fibroblast growth factor 10 (FGF10) expression. One-week hyperoxia (HYX) exposure of newborn mice leads to a permanent arrest in alveologenesis. To test the role of Fgf10 signaling to promote de novo alveologenesis following hyperoxia, we used transgenic mice allowing inducible expression of Fgf10 and recombinant FGF10 (rFGF10) protein delivered intraperitoneally. We carried out morphometry analysis, and IF on day 45. Alveolospheres assays were performed co-culturing AT2s from normoxia (NOX) with FACS-isolated Sca1Pos resident mesenchymal cells (rMC) from animals exposed to NOX, HYX-PBS, or HYX-FGF10. scRNAseq between rMC-Sca1Pos isolated from NOX and HYX-PBS was also carried out. Transgenic overexpression of Fgf10 and rFGF10 administration rescued the alveologenesis defects following HYX. Alveolosphere assays indicate that the activity of rMC-Sca1Pos is negatively impacted by HYX and partially rescued by rFGF10 treatment. Analysis by IF demonstrates a significant impact of rFGF10 on the activity of resident mesenchymal cells. scRNAseq results identified clusters expressing Fgf10, Fgf7, Pdgfra, and Axin2, which could represent the rMC niche cells for the AT2 stem cells. In conclusion, we demonstrate that rFGF10 administration is able to induce de novo alveologenesis in a BPD mouse model and identified subpopulations of rMC-Sca1Pos niche cells potentially representing its cellular target.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Humanos , Hiperoxia/metabolismo , Recién Nacido , Pulmón/metabolismo , Ratones , Ratones Transgénicos
12.
Cell Rep ; 39(2): 110663, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417692

RESUMEN

Fibroblast growth factor 10 (FGF10) is well established as a mesenchyme-derived growth factor and a critical regulator of fetal organ development in mice and humans. Using a single-cell RNA sequencing (RNA-seq) atlas of salivary gland (SG) and a tamoxifen inducible Fgf10CreERT2:R26-tdTomato mouse, we show that FGF10pos cells are exclusively mesenchymal until postnatal day 5 (P5) but, after P7, there is a switch in expression and only epithelial FGF10pos cells are observed after P15. Further RNA-seq analysis of sorted mesenchymal and epithelial FGF10pos cells shows that the epithelial FGF10pos population express the hallmarks of ancient ionocyte signature Forkhead box i1 and 2 (Foxi1, Foxi2), Achaete-scute homolog 3 (Ascl3), and the cystic fibrosis transmembrane conductance regulator (Cftr). We propose that epithelial FGF10pos cells are specialized SG ionocytes located in ducts and important for the ionic modification of saliva. In addition, they maintain FGF10-dependent gland homeostasis via communication with FGFR2bpos ductal and myoepithelial cells.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Glándulas Salivales , Animales , Células Epiteliales/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Ratones , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Glándulas Salivales/citología , Glándulas Salivales/metabolismo , Transducción de Señal
13.
Eur J Hum Genet ; 30(4): 480-484, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33967277

RESUMEN

Pulmonary acinar hypoplasia (PAH) and lacrimo-auriculo-dento-digital (LADD) syndrome have both been associated with loss-of-function variants in, or deletions of FGF10. Here we report a multi-generational family with seven members manifesting varying features of LADD syndrome, with one individual dying in early infancy of PAH. Whole genome sequencing in one family member identified a 12,158 bp deletion on chromosome 5p12 that removes two of the three exons of FGF10. Allele-specific PCR demonstrated that all affected family members, including the individual with PAH, carried the 12 kb deletion. We conclude the deletion is pathogenic and expands the mutational spectrum of FGF10 variants in LADD syndrome. The common mechanism underlying the variable clinical features of LADD syndrome is defective terminal branching of salivary and lacrimal glands and pulmonary acini, regulated by the TBX4-FGF10-FGFR2 pathway. The variable phenotypic expressivity of FGF10 haploinsufficiency from relatively benign to lethal is likely due to variation at other genetic loci.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos , Enfermedades del Aparato Lagrimal , Sindactilia , Anomalías Dentarias , Anomalías Múltiples , Exones , Factor 10 de Crecimiento de Fibroblastos/genética , Pérdida Auditiva , Humanos , Enfermedades del Aparato Lagrimal/genética , Sindactilia/genética , Anomalías Dentarias/genética
14.
Exp Cell Res ; 406(2): 112762, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34352276

RESUMEN

Keratinocyte growth factor (KGF)-2 has been highlighted to play a significant role in maintaining the endothelial barrier integrity in lung injury induced by ischemia-reperfusion (I/R). However, the underlying mechanism remains largely unknown. The aims of this study were to determine whether dexmedetomidine preconditioning (DexP) modulates pulmonary I/R-induced lung injury through the alteration in KGF-2 expression. In our I/R-modeled mice, DexP significantly inhibited pathological injury, inflammatory response, and inflammatory cell infiltration, while promoted endothelial barrier integrity and KGF-2 promoter activity in lung tissues. Bioinformatics prediction and ChIP-seq revealed that I/R significantly diminished the level of H3K4me3 modification in the KGF-2 promoter, which was significantly reversed by DexP. Moreover, DexP inhibited the expression of histone demethylase JMJD3, which in turn promoted the expression of KGF-2. In addition, overexpression of JMJD3 weakened the protective effect of DexP on lung injury in mice with I/R. Collectively, the present results demonstrated that DexP ameliorates endothelial barrier dysfunction via the JMJD3/KGF-2 axis.


Asunto(s)
Dexmedetomidina/farmacología , Endotelio Vascular/efectos de los fármacos , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Histonas/química , Histona Demetilasas con Dominio de Jumonji/metabolismo , Lesión Pulmonar/prevención & control , Daño por Reperfusión/complicaciones , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , Permeabilidad de la Membrana Celular , Endotelio Vascular/metabolismo , Factor 10 de Crecimiento de Fibroblastos/química , Factor 10 de Crecimiento de Fibroblastos/genética , Histona Demetilasas con Dominio de Jumonji/genética , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Regulación hacia Arriba
15.
PLoS One ; 16(8): e0255344, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34383782

RESUMEN

PURPOSE: The role of fibroblast growth factor (FGF) in orbital fibroblasts (OFs) is rarely known. In this study, we investigated the effect of FGF10 on fibrosis and the inflammation mechanism of Graves' orbitopathy (GO). METHODS: Orbital tissue from GO (n = 15) and non-GO (n = 15) was obtained for this study. The mRNA and protein expression levels of FGF10 and FGF receptor 2b (FGFR2b) in orbital tissue were determined by real-time polymerase chain reaction, western blot analysis, and confocal microscopy. The effects of FGF10 on transforming growth factor (TGF)-ß1 induced fibrotic proteins and interleukin (IL)-1ß- or tumor necrosis factor (TNF)-α- induced inflammatory proteins were investigated using recombinant human (rh) FGF10 and small interfering (si) RNA transfection against FGF10. RESULTS: FGF10 and FGFR2b mRNA expression levels were significantly lower in GO orbital tissues than in non-GO orbital tissues (p = 0.009 and 0.005, respectively). Immunostaining of FGF10 in orbital adipose tissues showed differences in FGF10 expression between GO and control samples. Immunostaining of FGF10 was very weak in the orbital tissues of GO patients. TGF-ß1-induced fibronectin, collagen Iα, α-smooth muscle actin protein expression in GO OFs was attenuated by rhFGF10 treatment and increased by knockdown of FGF10 via siFGF10 transfection. Similarly, IL-1ß- or TNF-α-induced IL-6, IL-8, and cyclooxygenase-2 protein production in GO OFs was either blocked by rhFGF10 treatment or further upregulated by inhibition of FGF10 via siFGF10 transfection. CONCLUSIONS: Our data demonstrate that FGF10 has beneficial effects on the inflammatory and fibrotic mechanisms of GO in primary cultured OFs, providing new insights into GO pathology and the discovery of FGF10 as a promising novel therapeutic application for the treatment of GO.


Asunto(s)
Regulación hacia Abajo , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Oftalmopatía de Graves/inmunología , Adulto , Estudios de Casos y Controles , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Femenino , Oftalmopatía de Graves/genética , Oftalmopatía de Graves/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , Microscopía Confocal , Persona de Mediana Edad , Modelos Biológicos , Cultivo Primario de Células , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
16.
Nat Commun ; 12(1): 3086, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035267

RESUMEN

Meis1 and Meis2 are homeodomain transcription factors that regulate organogenesis through cooperation with Hox proteins. Elimination of Meis genes after limb induction has shown their role in limb proximo-distal patterning; however, limb development in the complete absence of Meis function has not been studied. Here, we report that Meis1/2 inactivation in the lateral plate mesoderm of mouse embryos leads to limb agenesis. Meis and Tbx factors converge in this function, extensively co-binding with Tbx to genomic sites and co-regulating enhancers of Fgf10, a critical factor in limb initiation. Limbs with three deleted Meis alleles show proximal-specific skeletal hypoplasia and agenesis of posterior skeletal elements. This failure in posterior specification results from an early role of Meis factors in establishing the limb antero-posterior prepattern required for Shh activation. Our results demonstrate roles for Meis transcription factors in early limb development and identify their involvement in previously undescribed interaction networks that regulate organogenesis.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas de Homeodominio/genética , Esbozos de los Miembros/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/metabolismo , Esbozos de los Miembros/embriología , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética
17.
Life Sci ; 271: 119198, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33577857

RESUMEN

The aim of this study was to evaluate whether high levels of exogenous testosterone (T) interfere in prostate morphogenesis. Pregnant females were exposed to subcutaneous injections of T cypionate (500 µg/animal) at gestational days 20 and 22. Male and female pups were euthanized at postnatal days 1 and 15. 15-day-old males had only fibroblast growth factor 10 (FGF10) immunostaining and nuclear form factor altered by the treatment, whereas treated females (T1 and T15) had almost all analyzed parameters changed. T1 females showed an increased anogenital distance (AGD), whereas T15 females had both AGD and ovary weight increased. T1 females had a higher number of epithelial buds emerging from the urethral and vaginal epithelium. We observed ectopic prostatic tissue surrounding the vagina in both T1 and T15 females. Moreover, the ectopic acini of T15 females showed delayed luminal formation, and there was a thickening of the periacinar smooth muscle layer (SML). Finally, FGF10 immunostaining intensity decreased in both T15 male and female prostates. Indeed, Sonic hedgehog (Shh) was upregulated in T15 female prostates, whereas no difference was observed between the male groups. These data showed that exogenous T changed the nuclear morphology of prostate epithelial cells in both males and females. Surprisingly, smooth muscle hyperplasia was also observed in the ectopic female prostate. Moreover, T downregulated FGF10 in both male and female prostates. Interestingly, the results suggest that FGF10 downregulation is mediated by the upregulation of Shh in females. In conclusion, exogenous T disrupts prostate development, particularly, affecting, the female.


Asunto(s)
Epitelio/metabolismo , Factor 10 de Crecimiento de Fibroblastos/biosíntesis , Proteínas Hedgehog/biosíntesis , Músculo Liso/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Próstata/metabolismo , Testosterona/toxicidad , Animales , Animales Recién Nacidos , Epitelio/efectos de los fármacos , Epitelio/patología , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Gerbillinae , Proteínas Hedgehog/genética , Masculino , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Próstata/efectos de los fármacos , Próstata/patología
18.
Ophthalmic Genet ; 42(3): 239-242, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33620261

RESUMEN

Purpose: Genetic association between the fibroblast growth factor 10 (FGF10) gene rs339501 single nucleotide polymorphism (SNP) and high myopia remains inconsistent in different studies. This study aimed to investigate the association between FGF10 rs339501 and high myopia in a Han Chinese population.Methods: A total of 675 patients with high myopia (HM), including 246 extreme myopia (EM) patients, and 800 healthy subjects with normal vision from the Chinese Han population were selected as the study subjects. The SNP of FGF10 rs399501 was genotyped by TaqMan allele discrimination assay on the 7300 real-time polymorphism chain reaction system, and the relationship between genotype and allele frequency of FGF10 rs399501 and high myopia was analyzed.Results: In our study, there are statistically significant differences between high myopia patients and controls in the allele frequencies (OR = 1.268, 95%CI = 1.030 ~ 1.560, P = .025), but not in genotype distributions (χ2 = 5.673, P = .059) of rs399501 SNP in the FGF10 gene. In addition, a weak association was found in recessive model (GG vs. AG+AA: OR = 1.929, 95%CI = 1.004 ~ 3.708, P = .045), but not in dominant model (AG+GG vs. AA: OR = 1.239, 95%CI = 0.981 ~ 1.566, P = .072). Moreover, significant associations were also found between FGF10 rs339501 polymorphism and the risk of extreme myopia in all genetic models.Conclusion: Our results do support that the genetic variant of FGF10 rs339501 is associated with susceptibility of high myopia, especially extreme myopia in a Chinese Han population, and further exploration is needed for myopia in other populations.


Asunto(s)
Pueblo Asiatico/genética , Factor 10 de Crecimiento de Fibroblastos/genética , Predisposición Genética a la Enfermedad/genética , Miopía Degenerativa/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Estudios de Casos y Controles , China/epidemiología , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Técnicas de Genotipaje , Humanos , Masculino , Persona de Mediana Edad , Miopía Degenerativa/diagnóstico
19.
Cell Rep ; 34(8): 108772, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33626352

RESUMEN

Evidence regarding intraductal papillary neoplasm of the bile duct (IPNB) as a type of precancerous lesion of cholangiocarcinoma is limited. Moreover, a reproducible in vivo model is lacking, and IPNB pathogenesis remains unclear. Here, we use a doxycycline-inducible tetracycline (Tet)-on mice model to control fibroblast growth factor 10 (FGF10) expression, which regulates branching and tubule formation. FGF10-induced IPNB mimics the multifocal and divergent human IPNB phenotypes via the FGF10-FGF receptor 2 (FGFR2)-RAS-extracellular-signal-regulated kinase (ERK) signaling pathway. A paracrine/autocrine growth factor is sufficient to initiate and maintain IPNB originating from the peribiliary glands, including biliary stem/progenitor cells. With KrasG12D, p53, or p16 mutations or both, Fgf10-induced IPNB shows stepwise carcinogenesis, causing associated invasive carcinoma. Fgf10-induced papillary changes and progression are suppressed by the inhibition of the FGF10-FGFR2-RAS-ERK signaling pathway, demonstrating that the signal is a therapeutic target for IPNB and associated carcinoma.


Asunto(s)
Neoplasias de los Conductos Biliares/enzimología , Carcinoma Papilar/enzimología , Colangiocarcinoma/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Células Madre Neoplásicas/enzimología , Lesiones Precancerosas/enzimología , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Carcinoma Papilar/tratamiento farmacológico , Carcinoma Papilar/genética , Carcinoma Papilar/patología , Células Cultivadas , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Progresión de la Enfermedad , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Regulación Neoplásica de la Expresión Génica , Genes ras , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Células Madre Neoplásicas/patología , Fosforilación , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
20.
Respir Res ; 22(1): 26, 2021 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-33478486

RESUMEN

BACKGROUND: The epithelial-mesenchymal signaling involving SHH-FOXF1, TBX4-FGF10, and TBX2 pathways is an essential transcriptional network operating during early lung organogenesis. However, precise regulatory interactions between different genes and proteins in this pathway are incompletely understood. METHODS: To identify TBX2 and TBX4 genome-wide binding sites, we performed chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq) in human fetal lung fibroblasts IMR-90. RESULTS: We identified 14,322 and 1,862 sites strongly-enriched for binding of TBX2 and TBX4, respectively, 43.95% and 18.79% of which are located in the gene promoter regions. Gene Ontology, pathway enrichment, and DNA binding motif analyses revealed a number of overrepresented cues and transcription factor binding motifs relevant for lung branching that can be transcriptionally regulated by TBX2 and/or TBX4. In addition, TBX2 and TBX4 binding sites were found enriched around and within FOXF1 and its antisense long noncoding RNA FENDRR, indicating that the TBX4-FGF10 cascade may directly interact with the SHH-FOXF1 signaling. CONCLUSIONS: We highlight the complexity of transcriptional network driven by TBX2 and TBX4 and show that disruption of this crosstalk during morphogenesis can play a substantial role in etiology of lung developmental disorders.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina/métodos , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Hedgehog/metabolismo , Pulmón/metabolismo , Proteínas de Dominio T Box/metabolismo , Desarrollo Fetal/fisiología , Factor 10 de Crecimiento de Fibroblastos/genética , Factores de Transcripción Forkhead/genética , Proteínas Hedgehog/genética , Humanos , Pulmón/irrigación sanguínea , Pulmón/embriología , Unión Proteica/inmunología , Proteínas de Dominio T Box/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...