Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Life Sci ; 271: 119198, 2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33577857

RESUMO

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.


Assuntos
Epitélio/metabolismo , Fator 10 de Crescimento de Fibroblastos/biossíntese , Proteínas Hedgehog/biossíntese , Músculo Liso/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Próstata/metabolismo , Testosterona/toxicidade , Animais , Animais Recém-Nascidos , Epitélio/efeitos dos fármacos , Epitélio/patologia , Feminino , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Gerbillinae , Proteínas Hedgehog/genética , Masculino , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Próstata/efeitos dos fármacos , Próstata/patologia
2.
Stem Cell Res Ther ; 11(1): 2, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31900217

RESUMO

BACKGROUND: ALI/ARDS is the major cause of acute respiratory failure in critically ill patients. As human chorionic villi-derived MSCs (hCMSCs) could attenuate ALI in the airway injury model, and liraglutide, glucagon-like peptide 1 (GLP-1) agonist, possesses anti-inflammatory and proliferation promotion functions, we proposed to probe the potential combinatory effect of hCMSCs and liraglutide on ALI. METHODS: We examined the time- and dose-dependent manner of GLP-1R, SPC, Ang-1, and FGF-10 with LPS via western blot and qRT-PCR. Western blot and chromatin immunoprecipitation assay detected the effects of liraglutide on GLP-1R, SPC, Ang-1, and FGF-10 through PKAc/ß-catenin pathway and cAMP pathway. In the ALI animal model, we detected the effects of MSC and liraglutide combination on ALI symptoms by H&E staining, western blot, ELISA assays, calculating wet-to-dry ratio of the lung tissue, and counting neutrophils, leukocytes, and macrophages in mouse bronchoalveolar lavage fluid (BALF). RESULTS: The data demonstrated that LPS reduced hCMSC proliferation and GLP-1R, SPC, Ang-1, and FGF-10 levels in a dose- and time-dependent manner. Liraglutide significantly dampened the reduction of GLP-1R, SPC, Ang-1, and FGF-10 and reversed the effect of LPS on hCMSCs, which could be regulated by GLP-1R and its downstream cAMP/PKAc/ß-catenin-TCF4 signaling. Combination of hCMSCs with liraglutide showed more therapeutic efficacy than liraglutide alone in reducing LPS-induced ALI in the animal model. CONCLUSIONS: These results reveal that the combination of hCMSCs and liraglutide might be an effective strategy for ALI treatment.


Assuntos
Lesão Pulmonar Aguda/terapia , Hipoglicemiantes/uso terapêutico , Liraglutida/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Animais , Vilosidades Coriônicas , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fator 10 de Crescimento de Fibroblastos/biossíntese , Peptídeo 1 Semelhante ao Glucagon/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/biossíntese , Humanos , Hipoglicemiantes/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transfecção , beta Catenina/metabolismo
3.
J Clin Invest ; 130(2): 641-654, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31845906

RESUMO

Pituitary develops from oral ectoderm in contact with adjacent ventral hypothalamus. Impairment in this process results in congenital pituitary hypoplasia (CPH); however, there have been no human disease models for CPH thus far, prohibiting the elucidation of the underlying mechanisms. In this study, we established a disease model of CPH using patient-derived induced pluripotent stem cells (iPSCs) and 3D organoid technique, in which oral ectoderm and hypothalamus develop simultaneously. Interestingly, patient iPSCs with a heterozygous mutation in the orthodenticle homeobox 2 (OTX2) gene showed increased apoptosis in the pituitary progenitor cells, and the differentiation into pituitary hormone-producing cells was severely impaired. As an underlying mechanism, OTX2 in hypothalamus, not in oral ectoderm, was essential for progenitor cell maintenance by regulating LHX3 expression in oral ectoderm via FGF10 expression in the hypothalamus. Convincingly, the phenotype was reversed by the correction of the mutation, and the haploinsufficiency of OTX2 in control iPSCs revealed a similar phenotype, demonstrating that this mutation was responsible. Thus, we established an iPSC-based congenital pituitary disease model, which recapitulated interaction between hypothalamus and oral ectoderm and demonstrated the essential role of hypothalamic OTX2.


Assuntos
Haploinsuficiência , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Fatores de Transcrição Otx/metabolismo , Doenças da Hipófise/metabolismo , Hipófise/metabolismo , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Fatores de Transcrição Otx/genética , Doenças da Hipófise/congênito , Doenças da Hipófise/patologia , Hipófise/patologia
4.
J Pediatr Urol ; 16(1): 41.e1-41.e10, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31718875

RESUMO

INTRODUCTION: Fibroblast growth factors (FGFs) play a crucial role in early embryogenesis of the genital tubercle and are involved in the development of hypospadias, affecting both endo- and ectodermally derived tissues. It was hypothesized that expression of FGFs could be qualitatively or quantitatively altered in skin of children with hypospadias. OBJECTIVE: The objective of the study was to investigate expression patterns and transcription levels of FGF8, FGF10, and FGF Receptor 2 (FGFR2) in patients with hypospadias compared to normal controls. PATIENTS AND METHODS: Skin samples from the ventro-lateral aspect of the foreskin of 32 patients with hypospadias (17 distal and 15 proximal, mean age 25 months) and 10 normal foreskin samples (mean age 77 months) were analyzed by immunohistochemistry. Staining, localization, and distribution of positive cells in epidermis and dermis were categorized independently by two researchers. Complementary DNA (cDNA) samples prepared from messenger RNA (mRNA) isolates of the same samples were analyzed by quantitative polymerase chain reaction (qPCR), comparing expressions of FGF8, FGF10, and FGFR2 with loading controls. RESULTS: Patients with hypospadias consistently showed aberrant immunohistochemical staining patterns for FGF8/FGF10/FGFR2 in epidermis and dermis compared to patients without penile malformation (p < 0.01 for all markers). qPCR displayed no difference in expression levels on mRNA level (FGFR2 p = 0.44, FGF8 p = 0.77, and FGF10 p = 0.17) comparing normal foreskin with foreskin from patients with hypospadias. Figure. DISCUSSION: The results point at an impact of FGF signaling during embryological development of hypospadias on skin, as an ectodermally derived tissue. Similar to the urethral development, this might be a result of mesothelial-epithelial interactions. The differing expression patterns in immunohistochemistry are not matched by a quantitative difference in marker expression on the mRNA level, putatively caused by post-translational modifications or alterations of the downstream pathway. FGFs, particularly FGF10 and FGFR2, are critically involved in wound healing. CONCLUSIONS: There are significant differences in localization and distribution of FGF8, FGF10, and FGFR2 in comparisons of normal foreskin to foreskin of patients with hypospadias, whereas there is no difference in the quantitative expression of these markers on the mRNA level. This confirms the notion that penile skin is affected as well by the embryological aberrations during the embryogenesis of hypospadias.


Assuntos
Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/genética , Prepúcio do Pênis/metabolismo , Hipospadia/genética , Hipospadia/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transcrição Gênica , Criança , Pré-Escolar , Fator 10 de Crescimento de Fibroblastos/análise , Fator 8 de Crescimento de Fibroblasto/análise , Prepúcio do Pênis/química , Expressão Gênica , Humanos , Hipospadia/patologia , Imuno-Histoquímica , Lactente , Masculino , Estudos Prospectivos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/análise
5.
Physiol Rep ; 6(16): e13794, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30155985

RESUMO

TGF beta is a multifunctional cytokine that is important in the pathogenesis of pulmonary fibrosis. The ability of TGF beta to stimulate smooth muscle actin and extracellular matrix gene expression in fibroblasts is well established. In this report, we evaluated the effect of TGF beta on the expression of HGF, FGF7 (KGF), and FGF10, important growth and survival factors for the alveolar epithelium. These growth factors are important for maintaining type II cells and for restoration of the epithelium after lung injury. Under conditions of normal serum supplementation or serum withdrawal TGF beta inhibited fibroblast expression of HGF, FGF7, and FGF10. We confirmed these observations with genome wide RNA sequencing of the response of control and IPF fibroblasts to TGF beta. In general, gene expression in IPF fibroblasts was similar to control fibroblasts. Reduced expression of HGF, FGF7, and FGF10 is another means whereby TGF beta impairs epithelial healing and promotes fibrosis after lung injury.


Assuntos
Fibroblastos/efeitos dos fármacos , Fibrose Pulmonar Idiopática/patologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Fator de Crescimento Transformador beta/farmacologia , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Meios de Cultura Livres de Soro , Feminino , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Fator 7 de Crescimento de Fibroblastos/biossíntese , Fator 7 de Crescimento de Fibroblastos/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Pulmão/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , Fator de Crescimento Transformador beta/fisiologia
6.
Hum Mol Genet ; 27(1): 107-119, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29126155

RESUMO

Genital malformations are among the most common human birth defects, and both genetic and environmental factors can contribute to these malformations. Development of the external genitalia in mammals relies on complex signaling networks, and disruption of these signaling pathways can lead to genital defects. Islet-1 (ISL1), a member of the LIM/Homeobox family of transcription factors, has been identified as a major susceptibility gene for classic bladder exstrophy in humans, a common form of the bladder exstrophy-epispadias complex (BEEC), and is implicated in a role in urinary tract development. We report that deletion of Isl1 from the genital mesenchyme in mice led to hypoplasia of the genital tubercle and prepuce, with an ectopic urethral opening and epispadias-like phenotype. These mice also developed hydroureter and hydronephrosis. Identification of ISL1 transcriptional targets via ChIP-Seq and expression analyses revealed that Isl1 regulates several important signaling pathways during embryonic genital development, including the BMP, WNT, and FGF cascades. An essential function of Isl1 during development of the external genitalia is to induce Bmp4-mediated apoptosis in the genital mesenchyme. Together, these studies demonstrate that Isl1 plays a critical role during development of the external genitalia and forms the basis for a greater understanding of the molecular mechanisms underlying the pathogenesis of BEEC and urinary tract defects in humans.


Assuntos
Proteína Morfogenética Óssea 4/genética , Fator 10 de Crescimento de Fibroblastos/genética , Genitália/anormalidades , Genitália/embriologia , Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição/genética , Proteína Wnt-5a/genética , Animais , Extrofia Vesical/genética , Extrofia Vesical/metabolismo , Proteína Morfogenética Óssea 4/biossíntese , Proteína Morfogenética Óssea 4/metabolismo , Desenvolvimento Embrionário , Feminino , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genitália/metabolismo , Proteínas com Homeodomínio LIM/biossíntese , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Organogênese/genética , Transdução de Sinais , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/metabolismo , Proteína Wnt-5a/biossíntese , Proteína Wnt-5a/metabolismo
7.
Protein Expr Purif ; 138: 7-12, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26384708

RESUMO

Fibroblast growth factor 10 (FGF10) is a member of the FGF superfamily. It exhibits diverse biological functions, and is extensively used for fundamental research and clinical applications involving hair growth, tissue repair, and burn wounds. Oil bodies, obtained from oil seeds, have been exploited for a variety of biotechnology applications. The use of oil bodies reduces purification steps and costs associated with the production of heterogonous proteins. Here, recombinant human FGF10 (rhFGF10) was expressed in safflower (Carthamus tinctorius L.) seeds using oilbody-oleosin technology. A plant expression vector, pOTBar-oleosin-rhFGF10, was constructed and introduced into safflower using Agrobacterium tumefaciens transformation, and mature safflower plants were obtained by grafting. Oleosin-rhFGF10 was successfully transformed and expressed in safflower seeds and inherited to the T3 generation. Moreover, MTT assays demonstrated that oil bodies expressed oleosin-FGF10 had a dose-dependent effect on cellular proliferation. In conclusion, this may provide a method of producing oleosin-rhFGF10, and help us meet the increasing pharmacological demands for the protein.


Assuntos
Carthamus tinctorius/genética , Fator 10 de Crescimento de Fibroblastos/genética , Vetores Genéticos/química , Proteínas de Plantas/genética , Sementes/genética , Agrobacterium tumefaciens/genética , Agrobacterium tumefaciens/metabolismo , Animais , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Carthamus tinctorius/química , Carthamus tinctorius/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/isolamento & purificação , Fator 10 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Vetores Genéticos/metabolismo , Humanos , Gotículas Lipídicas/química , Gotículas Lipídicas/metabolismo , Camundongos , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Sementes/química , Sementes/metabolismo
8.
J Assist Reprod Genet ; 34(3): 383-390, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28000057

RESUMO

PURPOSE: In an attempt to improve in vitro embryo production, we investigated the effect of fibroblast growth factor 10 (FGF10) during in vitro maturation on the developmental capacity of bovine oocytes. MATERIAL AND METHODS: Cumulus-oocyte complexes (COCs) were aspirated from follicles of 3-8 mm diameter. After selection, the COCs were matured in medium with or without 0.5 ng/mL of FGF10. The effect of FGF10 during in vitro maturation (IVM) on nuclear maturation kinetics and expansion of the cumulus cells was investigated. Oocyte competence was assessed by the production and development speed of embryos and the relative expression of genes associated with embryo quality. RESULTS: FGF10 delayed the resumption of meiosis from 8 h onwards, but did not affect the percentage of oocytes reaching metaphase II, nor did it increase cumulus expansion at 22 h of maturation. We found no difference between treatments regarding embryo production, developmental speed, and gene expression. CONCLUSION: In conclusion, the presence of FGF10 during IVM had no effect on embryo production, developmental speed, and gene expression.


Assuntos
Desenvolvimento Embrionário/genética , Fator 10 de Crescimento de Fibroblastos/biossíntese , Técnicas de Maturação in Vitro de Oócitos , Oócitos/crescimento & desenvolvimento , Animais , Blastocisto/metabolismo , Bovinos , Células do Cúmulo/metabolismo , Técnicas de Cultura Embrionária , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Meiose/genética , Oócitos/metabolismo
9.
Stem Cell Res ; 17(3): 489-497, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27771497

RESUMO

The continuous growth of mouse incisors depends on epithelial stem cells (SCs) residing in the SC niche, called labial cervical loop (LaCL). The homeostasis of the SCs is subtly regulated by complex signaling networks. In this study, we focus on retinoic acid (RA), a derivative of Vitamin A and a known pivotal signaling molecule in controlling the functions of stem cells (SCs). We analyzed the expression profiles of several key molecules of the RA signaling pathway in cultured incisor explants upon exogenous RA treatment. The expression patterns of these molecules suggested a negative feedback regulation of RA signaling in the developing incisor. We demonstrated that exogenous RA had negative effects on incisor SCs and that this was accompanied by downregulation of Fgf10, a mesenchymally expressed SC survival factor in the mouse incisor. Supplement of Fgf10 in incisor cultures completely blocked RA effects by antagonizing apoptosis and increasing proliferation in LaCL epithelial SCs. In addition, Fgf10 obviously antagonized RA-induced downregulation of the SC marker Sox2 in incisor epithelial SCs. Our findings suggest that the negative effects of RA on incisor SCs result from inhibition of mesenchymal Fgf10.


Assuntos
Incisivo/citologia , Incisivo/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Tretinoína/toxicidade , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator 10 de Crescimento de Fibroblastos/biossíntese , Incisivo/metabolismo , Camundongos , Transdução de Sinais
10.
Gene Expr Patterns ; 19(1-2): 45-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26220830

RESUMO

Lhx9 is a member of the LIM-homeodomain gene family necessary for the correct development of many organs including gonads, limbs, heart and the nervous system. In the context of limb development, Lhx9 has been implicated as an integrator for Fibroblast growth factor (FGF) and Sonic hedgehog (Shh) signalling required for proximal-distal (PD) and anterior-posterior (AP) development of the limb. Three splice variants of the Lhx9 transcript are expressed during development, two of which are predicted to act in a dominant negative fashion, competing with the DNA binding version of Lhx9 for binding to cofactors via the LIM-domain. We examined the expression pattern for the three alternative splice forms of Lhx9; Lhx9α, Lhx9ß and Lhx9c during early limb development. We have found that of the three Lhx9 isoforms, only Lhx9α and Lhx9c (intact homeodomain) are expressed during early limb development, each with their own distinct expression pattern. Additionally we determined that Lhx9 expression overlaps with FGF10 expression in the developing limb bud mesenchyme. Limb bud explant cultures, in the presence of signalling pathway inhibitors, also indicated that Lhx9 mRNA expression in the limb bud was dependent on FGF signalling.


Assuntos
Fator 10 de Crescimento de Fibroblastos/metabolismo , Proteínas com Homeodomínio LIM/biossíntese , Proteínas com Homeodomínio LIM/genética , Botões de Extremidades/fisiologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Animais , Extremidades/embriologia , Feminino , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Botões de Extremidades/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Isoformas de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Transativadores/metabolismo
11.
Dev Biol ; 394(1): 181-90, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25109552

RESUMO

Fibroblast growth factors (FGFs) expressed in the apical ectodermal ridge (AER) and FGF10 expressed in the underlying mesoderm are essential for limb bud outgrowth. Their expression is maintained through a positive feedback loop. We identified the cis-regulatory element and trans-acting factors involved in the AER-FGF-dependent transactivation of Fgf10. Etv1 and Ewsr1 stimulated transcription from the Fgf10 promoter in the sub-AER mesenchyme of mouse and chick limb buds in a conserved AGAAAR cluster-dependent manner. We found that both Etv1 and Ewsr1 were necessary for Fgf10 expression and elongation of the limb bud. In addition, Etv1 and AER-FGF synergistically stimulated Fgf10 promoter activity in an Ewsr1-dependent manner. We also found that Etv1 and Ewsr1 bound to the segment of DNA containing the AGAAAR cluster in vivo and in vitro. Moreover, Etv1 directly bound to the AGAAAR sequence in vitro. Our results suggest that Etv1 and Ewsr1 transactivate Fgf10 directly and cooperatively in response to AER-FGFs.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/metabolismo , Botões de Extremidades/crescimento & desenvolvimento , Proteína EWS de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação/genética , Células Cultivadas , Embrião de Galinha , Ectoderma/metabolismo , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , Proteínas Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno
12.
Biomed Res Int ; 2013: 815895, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23841093

RESUMO

Despite a pivotal role in salivary gland development, homeostasis, and disease, the role of salivary gland mesenchyme is not well understood. In this study, we used the Col1a1-GFP mouse model to characterize the salivary gland mesenchyme in vitro and in vivo. The Col1a1-GFP transgene was exclusively expressed in the salivary gland mesenchyme. Ex vivo culture of mixed salivary gland cells in DMEM plus serum medium allowed long-term expansion of salivary gland epithelial and mesenchymal cells. The role of TGF-ß1 in salivary gland development and disease is complex. Therefore, we used this in vitro culture system to study the effects of TGF-ß1 on salivary gland cell differentiation. TGF-ß1 induced the expression of collagen, and inhibited the formation of acini-like structures in close proximity to mesenchymal cells, which adapted a fibroblastic phenotype. In contrast, TGF-ßR1 inhibition increased acini genes and fibroblast growth factors (Fgf-7 and Fgf-10), decreased collagen and induced formation of larger, mature acini-like structures. Thus, inhibition of TGF-ß signaling may be beneficial for salivary gland differentiation; however, due to differential effects of TGF-ß1 in salivary gland epithelial versus mesenchymal cells, selective inhibition is desirable. In conclusion, this mixed salivary gland cell culture system can be used to study epithelial-mesenchymal interactions and the effects of differentiating inducers and inhibitors.


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular , Glândulas Salivares/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 7 de Crescimento de Fibroblastos/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Mesenquimais , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Glândulas Salivares/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/antagonistas & inibidores
13.
J Biol Chem ; 288(15): 10440-50, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23460641

RESUMO

Cleft palate represents one of the most common congenital birth defects in humans. TGFß signaling, which is mediated by Smad-dependent and Smad-independent pathways, plays a crucial role in regulating craniofacial development and patterning, particularly in palate development. However, it remains largely unknown whether the Smad-independent pathway contributes to TGFß signaling function during palatogenesis. In this study, we investigated the function of TGFß activated kinase 1 (Tak1), a key regulator of Smad-independent TGFß signaling in palate development. We show that Tak1 protein is expressed in both the epithelium and mesenchyme of the developing palatal shelves. Whereas deletion of Tak1 in the palatal epithelium or mesenchyme did not give rise to a cleft palate defect, inactivation of Tak1 in the neural crest lineage using the Wnt1-Cre transgenic allele resulted in failed palate elevation and subsequently the cleft palate formation. The failure in palate elevation in Wnt1-Cre;Tak1(F/F) mice results from a malformed tongue and micrognathia, resembling human Pierre Robin sequence cleft of the secondary palate. We found that the abnormal tongue development is associated with Fgf10 overexpression in the neural crest-derived tongue tissue. The failed palate elevation and cleft palate were recapitulated in an Fgf10-overexpressing mouse model. The repressive effect of the Tak1-mediated noncanonical TGFß signaling on Fgf10 expression was further confirmed by inhibition of p38, a downstream kinase of Tak1, in the primary cell culture of developing tongue. Tak1 thus functions to regulate tongue development by controlling Fgf10 expression and could represent a candidate gene for mutation in human PRS clefting.


Assuntos
Linhagem da Célula , Fissura Palatina/embriologia , Embrião de Mamíferos/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Crista Neural/embriologia , Língua/anormalidades , Língua/embriologia , Animais , Fissura Palatina/genética , Fissura Palatina/patologia , Modelos Animais de Doenças , Embrião de Mamíferos/patologia , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Mutantes , Crista Neural/patologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Dev Biol ; 374(1): 210-22, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23220102

RESUMO

The analysis of Fgf10 mouse mutants has demonstrated a critical role for this ligand in neurosensory development of the vertebrate inner ear, and we have been looking to define the direct upstream regulators of Fgf10 in this sensory organ, as part of constructing the programme of early inner ear development. Through the analysis of reporter constructs in transgenic mouse embryos and neonatal mice, in this report we define a minimal 1400 bp enhancer from the 5' flanking region of Fgf10. This enhancer drives reporter transgene expression in a manner that recapitulates endogenous expression of Fgf10, from its initial onset in the invaginating otic placode and onwards throughout gestation, controlling Fgf10 expression in all developing sensory patches and in the developing VIIIth ganglion. This regulatory region includes three putative Gata3 binding sites that we demonstrate directly interacts with Gata3 protein through the DNA binding domain with differing affinities. Site directed mutagenesis of all three sites and functional testing in transgenic embryos using reporter transgenes reveals an absolute requirement for Gata3 in controlling Fgf10 expression. Transgenic analysis of individual Gata3 binding site mutations illustrates that only one of these binding sites is necessary for reporter expression. Together these data demonstrate that Gata3 directly activates Fgf10 in the early inner ear, and does so through a single binding site.


Assuntos
Orelha Interna/embriologia , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator de Transcrição GATA3/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Animais , Sítios de Ligação , Elementos Facilitadores Genéticos , Fator 10 de Crescimento de Fibroblastos/genética , Fator de Transcrição GATA3/metabolismo , Gânglios/metabolismo , Genes Reporter , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Transgenes , Tretinoína/metabolismo
15.
Development ; 138(11): 2359-68, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21558382

RESUMO

Blood vessels have been shown to play perfusion-independent roles in organogenesis. Here, we examined whether blood vessels determine branching stereotypy of the mouse lung airways in which coordinated branching of epithelial and vascular tubes culminates in their co-alignment. Using different ablative strategies to eliminate the lung vasculature, both in vivo and in lung explants, we show that proximity to the vasculature is indeed essential for patterning airway branching. Remarkably, although epithelial branching per se proceeded at a nearly normal rate, branching stereotypy was dramatically perturbed following vascular ablation. Specifically, branching events requiring a rotation to change the branching plane were selectively affected. This was evidenced by either the complete absence or the shallow angle of their projections, with both events contributing to an overall flat lung morphology. Vascular ablation also led to a high frequency of ectopic branching. Regain of vascularization fully rescued arrested airway branching and restored normal lung size and its three-dimensional architecture. This role of the vasculature is independent of perfusion, flow or blood-borne substances. Inhibition of normal branching resulting from vascular loss could be explained in part by perturbing the unique spatial expression pattern of the key branching mediator FGF10 and by misregulated expression of the branching regulators Shh and sprouty2. Together, these findings uncovered a novel role of the vasculature in organogenesis, namely, determining stereotypy of epithelial branching morphogenesis.


Assuntos
Pulmão/irrigação sanguínea , Pulmão/embriologia , Organogênese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Comunicação Celular , Células Endoteliais/fisiologia , Fator 10 de Crescimento de Fibroblastos/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/biossíntese , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Morfogênese , Neovascularização Fisiológica , Técnicas de Cultura de Órgãos , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Assay Drug Dev Technol ; 9(4): 403-19, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21395400

RESUMO

Embryonic stem (ES) cell technology may serve as a platform for the discovery of drugs to treat diseases such as diabetes. However, because of difficulties in establishing reliable ES cell differentiation methods and in creating cost-effective plating conditions for the high-throughput format, screening for molecules that regulate pancreatic beta cells and their immediate progenitors has been limited. A relatively simple and inexpensive differentiation protocol that allows efficient generation of insulin-expressing cells from murine ES cells was previously established in our laboratories. In this report, this system is characterized in greater detail to map developmental cell stages for future screening experiments. Our results show that sequential activation of multiple gene markers for undifferentiated ES cells, epiblast, definitive endoderm, foregut, and pancreatic lineages was found to follow the sequence of events that mimics pancreatic ontogeny. Cells that expressed enhanced green fluorescent protein, driven by pancreatic and duodenal homeobox 1 or insulin 1 promoter, correctly expressed known beta cell lineage markers. Overexpression of Sox17, an endoderm fate-determining transcription factor, at a very early stage of differentiation (days 2-3) enhanced pancreatic gene expression. Overexpression of neurogenin3, an endocrine progenitor cell marker, induced glucagon expression at stages when pancreatic and duodenal homeobox 1 message was present (days 10-16). Forced expression (between days 16 and 25) of MafA, a pancreatic maturation factor, resulted in enhanced expression of insulin genes, glucose transporter 2 and glucokinase, and glucose-responsive insulin secretion. Day 20 cells implanted in vivo resulted in pancreatic-like cells. Together, our differentiation assay recapitulates the proceedings and behaviors of pancreatic development and will be valuable for future screening of beta cell effectors.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Células Secretoras de Insulina/metabolismo , Pâncreas/crescimento & desenvolvimento , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/genética , Células Cultivadas , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Endoderma/metabolismo , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/análise , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/biossíntese , Insulina/genética , Insulina/metabolismo , Masculino , Camundongos , Camundongos SCID , Pâncreas/citologia , Pâncreas/metabolismo , Fatores de Transcrição SOXF/análise , Fatores de Transcrição SOXF/biossíntese , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Transativadores/análise , Transativadores/biossíntese , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/análise , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Development ; 138(5): 873-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21247964

RESUMO

The pituitary gland is an endocrine organ that is developmentally derived from a fold in the oral ectoderm and a juxtaposed fold in the neural ectoderm. Here, we show that the absence of Vax1, a homeodomain transcription factor known for its role in eye and optic chiasm development, causes the rostral oral ectoderm to form an ectopic fold that eventually develops into a separate second pituitary with all the pituitary cell types and neuronal fibers characteristic of the normal pituitary. The induction of the second pituitary is associated with a localized ectopic expression of Fgf10, a gene encoding a growth factor known to recruit oral ectodermal cells into the pituitary. Interestingly, there are rare cases of pituitary duplications in humans that are also associated with optic nerve dysplasia, suggesting that VAX1 might be involved in the pathogenesis of this disorder.


Assuntos
Ectoderma/citologia , Neuropeptídeos/deficiência , Hipófise/crescimento & desenvolvimento , Fatores de Transcrição/deficiência , Animais , Fator 10 de Crescimento de Fibroblastos/biossíntese , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Fibras Nervosas , Doenças do Nervo Óptico/etiologia , Hipófise/inervação , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
18.
J Immunol ; 185(8): 4896-903, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20861353

RESUMO

Bronchopulmonary dysplasia (BPD) is a frequent complication of preterm birth. This chronic lung disease results from arrested saccular airway development and is most common in infants exposed to inflammatory stimuli. In experimental models, inflammation inhibits expression of fibroblast growth factor-10 (FGF-10) and impairs epithelial-mesenchymal interactions during lung development; however, the mechanisms connecting inflammatory signaling with reduced growth factor expression are not yet understood. In this study we found that soluble inflammatory mediators present in tracheal fluid from preterm infants can prevent saccular airway branching. In addition, LPS treatment led to local production of mediators that inhibited airway branching and FGF-10 expression in LPS-resistant C.C3-Tlr4(Lpsd)/J fetal mouse lung explants. Both direct NF-κB activation and inflammatory cytokines (IL-1ß and TNF-α) that activate NF-κB reduced FGF-10 expression, whereas chemokines that signal via other inflammatory pathways had no effect. Mutational analysis of the FGF-10 promoter failed to identify genetic elements required for direct NF-κB-mediated FGF-10 inhibition. Instead, NF-κB activation appeared to interfere with the normal stimulation of FGF-10 expression by Sp1. Chromatin immunoprecipitation and nuclear coimmunoprecipitation studies demonstrated that the RelA subunit of NF-κB and Sp1 physically interact at the FGF-10 promoter. These findings indicate that inflammatory signaling through NF-κB disrupts the normal expression of FGF-10 in fetal lung mesenchyme by interfering with the transcriptional machinery critical for lung morphogenesis.


Assuntos
Fator 10 de Crescimento de Fibroblastos/biossíntese , Pulmão/embriologia , NF-kappa B/metabolismo , Proteínas Quinases/metabolismo , Animais , Corioamnionite/metabolismo , Imunoprecipitação da Cromatina , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Imunoprecipitação , Recém-Nascido , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Nascimento Prematuro , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
20.
Mech Dev ; 127(3-4): 220-34, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19969077

RESUMO

Fgf10 is a critical component of mesenchymal-to-epithelial signaling during endodermal development. In the Fgf10 null pancreas, the embryonic progenitor population fails to expand, while ectopic Fgf10 expression forces progenitor arrest and organ hyperplasia. Using a conditional Fgf10 gain-of-function model, we observed that the timing of Fgf10 expression affected the cellular competence of the arrested pancreatic progenitors. We present evidence that the Fgf10-arrested progenitor state is reversible and that terminal differentiation resumes upon cessation of Fgf10 production. However, competence towards the individual pancreatic cell lineages depended upon the gestational time of when Fgf10 expression was attenuated. This revealed a competence window of endocrine and ductal cell formation that coincided with the pancreatic secondary transition between E13.5 and E15.5. We demonstrate that maintaining the Fgf10-arrested state during this period leads to permanent loss of competence for the endocrine and ductal cell fates. However, competence of the arrested progenitors towards the exocrine cell fate was retained throughout the secondary transition. Sustained Fgf10 expression caused irreversible loss of Ngn3 expression, which may underlie the loss of endocrine competence. Maintenance of exocrine competence may be attributable to continuous Ptf1a expression in the Fgf10-arrested progenitors. This may explain the rapid induction of Bhlhb8, a normally distalized cell intrinsic marker, following loss of ectopic Fgf10 expression. We conclude that the window for endocrine and ductal cell competence ceases during the secondary transition in pancreatic development.


Assuntos
Diferenciação Celular/fisiologia , Fator 10 de Crescimento de Fibroblastos/fisiologia , Pâncreas/citologia , Animais , Linhagem da Célula , Doxiciclina/farmacologia , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA