Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Int J Biol Sci ; 17(14): 3862-3874, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34671204

RESUMO

Fibroblast growth factors (FGFs) include a large family of growth factors that play a critical role in maintaining bone homeostasis, but the specific role of its members such as FGF7 does not well understand. Osteoblasts are a kind of major cells essential for bone formation. Osteoblasts interact with one another to create the unique structure of osteons. The well-connected osteons constitute the cortical bone. As an early osteocyte marker that triggers actin cytoskeleton dynamics, E11 is essential for osteoblasts' dendrites formation. However, the upstream which regulates E11 is mainly unknown. The purpose of this study was to examine the influence of FGF7 on the expression and the distribution of E11 in osteoblasts, which mediated osteoblasts' processes formation and gap junctional intercellular communication (GJIC) partly through connexin43 (Cx43). We first demonstrated that FGF7 increased the expression of E11 in osteoblasts. We then showed that FGF7 promoted osteoblasts' dendrites elongation and functional gap junctions formation. Furthermore, E11 interacted directly with Cx43 in primary osteoblasts. MAPK pathway and PI3K-AKT pathway were involved in the effect of FGF7. Our results shed light on the unique role of FGF7 on osteoblasts, which may indicate that FGF7 plays a more significant role in the later stages of bone development and homeostasis.


Assuntos
Comunicação Celular/fisiologia , Conexina 43/metabolismo , Fator 7 de Crescimento de Fibroblastos/fisiologia , Glicoproteínas de Membrana/metabolismo , Células 3T3 , Animais , Ligantes , Sistema de Sinalização das MAP Quinases , Camundongos , Osteoblastos/metabolismo
2.
PLoS Biol ; 17(1): e2006972, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30695023

RESUMO

Insulin provides important information to tissues about feeding behavior and energy status. Defective insulin signaling is associated with ageing, tissue dysfunction, and impaired wound healing. In the liver, insulin resistance leads to chronic damage and fibrosis, but it is unclear how tissue-repair mechanisms integrate insulin signals to coordinate an appropriate injury response or how they are affected by insulin resistance. In this study, we demonstrate that insulin resistance impairs local cellular crosstalk between the fibrotic stroma and bipotent adult liver progenitor cells (LPCs), whose paracrine interactions promote epithelial repair and tissue remodeling. Using insulin-resistant mice deficient for insulin receptor substrate 2 (Irs2), we highlight dramatic impairment of proregenerative fibroblast growth factor 7 (Fgf7) signaling between stromal niche cells and LPCs during chronic injury. We provide a detailed account of the role played by IRS2 in promoting Fgf7 ligand and receptor (Fgfr2-IIIb) expression by the two cell compartments, and we describe an insulin/IRS2-dependent feed-forward loop capable of sustaining hepatic re-epithelialization by driving FGFR2-IIIb expression. Finally, we shed light on the regulation of IRS2 and FGF7 within the fibrotic stroma and show-using a human coculture system-that IRS2 silencing shifts the equilibrium away from paracrine epithelial repair in favor of fibrogenesis. Hence, we offer a compelling insight into the contribution of insulin resistance to the pathogenesis of chronic liver disease and propose IRS2 as a positive regulator of communication between cell types and the transition between phases of stromal to epithelial repair.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Fator 7 de Crescimento de Fibroblastos/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Fator 7 de Crescimento de Fibroblastos/fisiologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/fisiologia , Resistência à Insulina/fisiologia , Fígado/metabolismo , Camundongos , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
3.
Am J Respir Cell Mol Biol ; 57(2): 162-173, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28296468

RESUMO

Repair of the lung epithelium after injury is a critical component for resolution; however, the processes necessary to drive epithelial resolution are not clearly defined. Published data demonstrate that Foxp3+ regulatory T cells (Tregs) enhance alveolar epithelial proliferation after injury, and Tregs in vitro directly promote type II alveolar epithelial cell (AT2) proliferation, in part by a contact-independent mechanism. Therefore, we sought to determine the contribution of Treg-specific expression of a growth factor that is known to be important in lung repair, keratinocyte growth factor (kgf). The data demonstrate that Tregs express kgf and that Treg-specific expression of kgf regulates alveolar epithelial proliferation during the resolution phase of acute lung injury and in a model of regenerative alveologenesis in vivo. In vitro experiments demonstrate that AT2 cells cocultured with Tregs lacking kgf have decreased rates of proliferation compared with AT2 cells cocultured with wild-type Tregs. Moreover, Tregs isolated from lung tissue and grown in culture express higher levels of two growth factors that are important for lung repair (kgf and amphiregulin) compared with Tregs isolated from splenic tissue. Lastly, Tregs isolated from human lung tissue can be stimulated ex vivo to induce kgf expression. This study reveals mechanisms by which Tregs direct tissue-reparative effects during resolution after acute lung injury, further supporting the emerging role of Tregs in tissue repair.


Assuntos
Células Epiteliais Alveolares/citologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Linfócitos T Reguladores/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Transferência Adotiva , Células Epiteliais Alveolares/patologia , Anfirregulina/biossíntese , Anfirregulina/genética , Animais , Divisão Celular , Técnicas de Cocultura , Toxina Diftérica/toxicidade , Fator 7 de Crescimento de Fibroblastos/biossíntese , Fator 7 de Crescimento de Fibroblastos/genética , Fatores de Transcrição Forkhead/análise , Regulação da Expressão Gênica/imunologia , Humanos , Lipopolissacarídeos/toxicidade , Pulmão/citologia , Depleção Linfocítica , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pneumonectomia , Complicações Pós-Operatórias/imunologia , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/patologia , Linfócitos T Reguladores/classificação , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante
4.
Oncotarget ; 7(52): 86011-86025, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-27852068

RESUMO

We have recently demonstrated that, fibroblast growth factor 2 (FGFR2), signalling via ribosomal S6 kinase 2 (RSK2), promotes progression of breast cancer (BCa). Loss of progesterone receptor (PR), whose activity in BCa cells can be stimulated by growth factor receptors (GFRs), is associated with poor patient outcome. Here we showed that FGF7/FGFR2 triggered phosphorylation of PR at Ser294, PR ubiquitination and subsequent receptor`s degradation via the 26S proteasome pathway in BCa cells. We further demonstrated that RSK2 mediated FGF7/FGFR2-induced PR downregulation. In addition, a strong synergistic effect of FGF7 and progesterone (Pg), reflected in the enhanced anchorage-independent growth and cell migration, was observed. Analysis of clinical material demonstrated that expression of PR inversely correlated with activated RSK (RSK-P) (p = 0.016). Patients with RSK-P(+)/PR(-) tumours had 3.629-fold higher risk of recurrence (p = 0.002), when compared with the rest of the cohort. Moreover, RSK-P(+)/PR(-) phenotype was shown as an independent prognostic factor (p = 0.006). These results indicate that the FGF7/FGFR2-RSK2 axis promotes PR turnover and activity, which may sensitize BCa cells to stromal stimuli and contribute to the progression toward steroid hormone negative BCa.


Assuntos
Neoplasias da Mama/metabolismo , Fator 7 de Crescimento de Fibroblastos/fisiologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/fisiologia , Receptores de Progesterona/metabolismo , Transdução de Sinais/fisiologia , Neoplasias da Mama/mortalidade , Linhagem Celular Tumoral , Feminino , Humanos , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia
5.
Dev Biol ; 419(2): 348-356, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27590203

RESUMO

Hypertrophy, hyperplasia and altered mucus secretion from the respiratory submucosal glands (SMG) are characteristics of airway diseases such as cystic fibrosis, asthma and chronic bronchitis. More commonly, hyper-secretion of the nasal SMGs contributes to allergic rhinitis and upper airway infection. Considering the role of these glands in disease states, there is a significant dearth in understanding the molecular signals that regulate SMG development and patterning. Due to the imperative role of FGF signalling during the development of other branched structures, we investigated the role of Fgf10 during initiation and branching morphogenesis of murine nasal SMGs. Fgf10 is expressed in the mesenchyme around developing SMGs while expression of its receptor Fgfr2 is seen within glandular epithelial cells. In the Fgf10 null embryo, Steno's gland and the maxillary sinus gland were completely absent while other neighbouring nasal glands showed normal duct elongation but defective branching. Interestingly, the medial nasal glands were present in Fgf10 homozygotes but missing in Fgfr2b mutants, with expression of Fgf7 specifically expressed around these developing glands, indicating that Fgf7 might compensate for loss of Fgf10 in this group of glands. Intriguingly the lateral nasal glands were only mildly affected by loss of FGF signalling, while these glands were missing in Eda mutant mice, where the Steno's and maxillary sinus gland developed as normal. This analysis reveals that regulation of nasal gland development is complex with different subsets of glands being regulated by different signalling pathways. This analysis helps shed light on the nasal gland defects observed in patients with hypohidrotic ectodermal dysplasia (HED) (defect EDA pathway) and LADD syndrome (defect FGFR2b pathway).


Assuntos
Ectodisplasinas/fisiologia , Glândulas Exócrinas/embriologia , Fator 10 de Crescimento de Fibroblastos/fisiologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/fisiologia , Animais , Ectodisplasinas/deficiência , Ectodisplasinas/genética , Ressecção Endoscópica de Mucosa , Glândulas Exócrinas/metabolismo , Glândulas Exócrinas/ultraestrutura , Feminino , Fator 10 de Crescimento de Fibroblastos/deficiência , Fator 10 de Crescimento de Fibroblastos/genética , Fator 7 de Crescimento de Fibroblastos/fisiologia , Masculino , Seio Maxilar/embriologia , Seio Maxilar/ultraestrutura , Mesoderma/metabolismo , Camundongos , Morfogênese , Mucosa Nasal/embriologia , Mucosa Nasal/ultraestrutura , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/deficiência , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética
6.
Am J Physiol Lung Cell Mol Physiol ; 310(9): L868-79, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26919897

RESUMO

Keratinocyte growth factor (KGF) is an epithelial mitogen that has been reported to protect the lungs from a variety of toxic and infectious insults. In prior studies we found that recombinant human KGF accelerates clearance of bacteria from the murine lung by augmenting the function of alveolar macrophages (AM). In this study we tested the hypothesis that endogenous KGF plays a role in the maintenance of innate pulmonary defense against gram-negative bacterial infections. KGF-deficient mice exhibited delayed clearance of Escherichia coli from the lungs, attenuated phagocytosis by AM, and decreased antimicrobial activity in bronchoalveolar lavage (BAL) fluid, due in part to reductions in levels of surfactant protein A, surfactant protein D, and lysozyme. These immune deficits were accompanied by lower alveolar type II epithelial cell counts and reduced alveolar type II epithelial cell expression of collectin and lysozyme genes on a per cell basis. No significant between-group differences were detected in selected inflammatory cytokines or BAL inflammatory cell populations at baseline or after bacterial challenge in the wild-type and KGF-deficient mice. A single intranasal dose of recombinant human KGF reversed defects in bacterial clearance, AM function, and BAL fluid antimicrobial activity. We conclude that KGF supports alveolar innate immune defense through maintenance of alveolar antimicrobial protein levels and functions of AM. Together these data demonstrate a role for endogenous KGF in maintenance of normal pulmonary innate immune function.


Assuntos
Infecções por Escherichia coli/imunologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Imunidade Inata , Macrófagos Alveolares/imunologia , Pneumonia Bacteriana/imunologia , Animais , Células Cultivadas , Colectinas/genética , Colectinas/metabolismo , Infecções por Escherichia coli/metabolismo , Feminino , Expressão Gênica , Humanos , Macrófagos Alveolares/microbiologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muramidase/genética , Muramidase/metabolismo , Pneumonia Bacteriana/metabolismo , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia
7.
J Endocrinol ; 228(2): R31-43, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26542145

RESUMO

Fibroblast growth factors (FGFs) have been shown to alter growth and differentiation of reproductive tissues in a variety of species. Within the female reproductive tract, the effects of FGFs have been focused on the ovary, and the most studied one is FGF2, which stimulates granulosa cell proliferation and decreases differentiation (decreased steroidogenesis). Other FGFs have also been implicated in ovarian function, and this review summarizes the effects of members of two subfamilies on ovarian function; the FGF7 subfamily that also contains FGF10, and the FGF8 subfamily that also contains FGF18. There are data to suggest that FGF8 and FGF18 have distinct actions on granulosa cells, despite their apparent similar receptor binding properties. Studies of non-reproductive developmental biology also indicate that FGF8 is distinct from FGF18, and that FGF7 is also distinct from FGF10 despite similar receptor binding properties. In this review, the potential mechanisms of differential action of FGF7/FGF10 and FGF8/FGF18 during organogenesis will be reviewed and placed in the context of follicle development. A model is proposed in which FGF8 and FGF18 differentially activate receptors depending on the properties of the extracellular matrix in the follicle.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Folículo Ovariano/fisiologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Células Epiteliais/fisiologia , Feminino , Fator 10 de Crescimento de Fibroblastos/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Fator 8 de Crescimento de Fibroblasto/fisiologia , Células da Granulosa/fisiologia , Humanos , Mesoderma/fisiologia , Organogênese , Folículo Ovariano/crescimento & desenvolvimento
8.
Br J Dermatol ; 174(3): 533-41, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26471375

RESUMO

BACKGROUND: Data indicate that in psoriasis, abnormalities are already present in nonlesional skin. Transforming growth factor-ß and keratinocyte growth factor (KGF), together with fibronectin and α5ß1 integrin, were suggested to play a crucial role in the pathogenesis of psoriasis by influencing inflammation and keratinocyte hyperproliferation. OBJECTIVES: To investigate the expression of KGF, fibroblast growth factor receptor (FGFR)2, fibronectin (FN) and extra domain A (EDA)-positive FN in healthy and nonlesional psoriatic skin, and to study the effect of KGF on the regulation of FN and EDA(+) FN production by fibroblasts. METHODS: Healthy, nonlesional psoriatic skin and lesional psoriatic skin were immunostained for α5 integrin, KGF, FGFR2, EDA(+) FN and signal transducer and activator of transcription (STAT)1. KGF-treated cell cultures were analysed for FN and EDA(+) FN mRNA and protein by real-time reverse-transcriptase polymerase chain reaction and flow cytometry, respectively. The major downstream signalling of KGF was investigated by blocking experiments using inhibitors of mitogen-activated protein kinase (MAPK) kinase (MEK1), AKT1/2, STAT1 and STAT3. RESULTS: The expression of α5 integrin, EDA(+) FN, KGF and its receptor FGFR2 is elevated in psoriatic nonlesional skin compared with healthy skin. KGF mildly induced EDA(+) FN, but not FN expression in healthy fibroblasts through MAPK signalling. Fibroblasts express the FGFR2-IIIc splice variant. STAT1 negatively regulates both FN and EDA(+) FN expression in healthy fibroblasts, and this regulation is compromised in fibroblasts derived from nonlesional psoriatic dermis. We detected active STAT1 in healthy and lesional skin, similarly to a previous report. However, in the nonlesional skin STAT1 activation was absent in tissues far away from lesions. CONCLUSIONS: The production of FN and EDA(+) FN by fibroblasts and the signalling of STAT1 are abnormally regulated in psoriatic nonlesional skin.


Assuntos
Fator 7 de Crescimento de Fibroblastos/fisiologia , Fibroblastos/metabolismo , Fibronectinas/biossíntese , Psoríase/metabolismo , Adolescente , Adulto , Estudos de Casos e Controles , Células Cultivadas , Fibronectinas/metabolismo , Voluntários Saudáveis , Humanos , Queratinócitos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Melanócitos/metabolismo , Pessoa de Meia-Idade , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3 , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Adulto Jovem
9.
J Cell Mol Med ; 18(9): 1895-907, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25088572

RESUMO

One of the most frequent complaints for post-menopausal women is vaginal atrophy, because of reduction in circulating oestrogens. Treatments based on local oestrogen administration have been questioned as topic oestrogens can reach the bloodstream, thus leading to consider their safety as controversial, especially for patients with a history of breast or endometrial cancers. Recently, growth factors have been shown to interact with the oestrogen pathway, but the mechanisms still need to be fully clarified. In this study, we investigated the effect of keratinocyte growth factor (KGF), a known mitogen for epithelial cells, on human vaginal mucosa cells, and its potential crosstalk with oestrogen pathways. We also tested the in vivo efficacy of KGF local administration on vaginal atrophy in a murine model. We demonstrated that KGF is able to induce proliferation of vaginal mucosa, and we gained insight on its mechanism of action by highlighting its contribution to switch ERα signalling towards non-genomic pathway. Moreover, we demonstrated that KGF restores vaginal trophism in vivo similarly to intravaginal oestrogenic preparations, without systemic effects. Therefore, we suggest a possible alternative therapy for vaginal atrophy devoid of the risks related to oestrogen-based treatments, and a patent (no. RM2012A000404) has been applied for this study.


Assuntos
Fator 7 de Crescimento de Fibroblastos/administração & dosagem , Doenças Vaginais/tratamento farmacológico , Administração Tópica , Animais , Proliferação de Células , Epitélio/patologia , Estradiol/fisiologia , Feminino , Fator 7 de Crescimento de Fibroblastos/fisiologia , Humanos , Células MCF-7 , Camundongos , Mucosa/patologia , Ovariectomia , Transdução de Sinais , Vagina/patologia
10.
Autophagy ; 10(5): 803-21, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24577098

RESUMO

Autophagy is a degradative pathway through which cells overcome stressful conditions and rapidly change their phenotype during differentiation. Despite its protective role, when exacerbated, autophagy may lead to cell death. Several growth factors involved in cell survival and in preventing differentiation are able to inhibit autophagy. Here we investigated the autophagic role of FGF7/KGF, an important player in epithelial cell protection and differentiation. Biochemical and quantitative fluorescence approaches showed that FGF7 and its signaling induce autophagy in human keratinocytes and the use of specific inhibitors indicated that this effect is independent of the PI3K-AKT-MTOR pathway. The selective block of autophagosome-to-lysosome fusion clarified that FGF7 induces autophagy stimulating autophagosome formation. However, quantitative fluorescence approaches also indicated that, upon a prolonged autophagic stimulus, FGF7 is able to accelerate autophagosome turnover. Moreover, in differentiating keratinocytes, the use of the autophagic inhibitor 3-MA as well as the depletion of BECN1 and ATG5, 2 essential regulators of the process, counteracted the FGF7-induced increase of the differentiation marker KRT1/K1, suggesting that autophagy is required for the FGF7-mediated early differentiation. These results provide the first evidence of a role of FGF7 in the regulation of sequential steps of the autophagic process and strengthen the hypothesis of a direct interplay between autophagy and differentiation. On the other hand, the ability of FGF7 to accelerate autophagosome turnover, preventing their dangerous accumulation, is consistent with the well-established protective role played by the growth factor in epithelial cells.


Assuntos
Autofagia/genética , Fator 7 de Crescimento de Fibroblastos/fisiologia , Queratinócitos/fisiologia , Fagossomos/genética , Fagossomos/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Humanos , Lisossomos/fisiologia , Fusão de Membrana/genética
11.
J Clin Invest ; 123(8): 3525-38, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23867503

RESUMO

Oncogenic transcription factors drive many human cancers, yet identifying and therapeutically targeting the resulting deregulated pathways has proven difficult. Squamous cell carcinoma (SCC) is a common and lethal human cancer, and relatively little progress has been made in improving outcomes for SCC due to a poor understanding of its underlying molecular pathogenesis. While SCCs typically lack somatic oncogene-activating mutations, they exhibit frequent overexpression of the p53-related transcription factor p63. We developed an in vivo murine tumor model to investigate the function and key transcriptional programs of p63 in SCC. Here, we show that established SCCs are exquisitely dependent on p63, as acute genetic ablation of p63 in advanced, invasive SCC induced rapid and dramatic apoptosis and tumor regression. In vivo genome-wide gene expression analysis identified a tumor-survival program involving p63-regulated FGFR2 signaling that was activated by ligand emanating from abundant tumor-associated stroma. Correspondingly, we demonstrate the therapeutic efficacy of extinguishing this signaling axis in endogenous SCCs using the clinical FGFR2 inhibitor AZD4547. Collectively, these results reveal an unanticipated role for p63-driven paracrine FGFR2 signaling as an addicting pathway in human cancer and suggest a new approach for the treatment of SCC.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Proteínas de Membrana/fisiologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Neoplasias Cutâneas/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose , Benzamidas/farmacologia , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/tratamento farmacológico , Fator 7 de Crescimento de Fibroblastos/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Comunicação Parácrina , Piperazinas/farmacologia , Pirazóis/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Transdução de Sinais , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/tratamento farmacológico , Transcrição Gênica , Transcriptoma , Células Tumorais Cultivadas
12.
Ann Dermatol Venereol ; 139 Suppl 3: S96-101, 2012 Nov.
Artigo em Francês | MEDLINE | ID: mdl-23260525

RESUMO

Hyperpigmentation of the skin is a common dermatologic condition in all skin types but most prominent in brown-skinned population. In skin of color any inflammation or injury can be accompanied by alterations in pigmentation (hyper/hypo-pigmentation). Postinflammatory hyperpigmentation (PIH) can be observed in many skin conditions including acne, eczema, and contact dermatitis. In the control of skin pigmentation, parallel to the cross-talk between keratinocytes and melanocytes, increasing evidence has underlined the crucial role exerted by the interactions between mesenchymal and epithelial cells through the release of fibroblast-derived growth factors. Among these factors, the keratinocyte growth factor (KGF), alone or in combination with interleukin-1α, induces melanin deposition in vitro and hyperpigmented lesions in vivo. Furthermore, a moderate increase of KGF and a high induction of its receptor have been shown in solar lentigo lesions, suggesting the involvement of this growth factor in the onset of the hyperpigmented spots. Several studies highlight the possible contribution of the fibroblast-derived melanogenic growth factors to the hyperpigmentated lesions, in the context of the mesenchymal - epithelial interactions modulating melanocyte functions.


Assuntos
Dermatite/fisiopatologia , Hiperpigmentação/fisiopatologia , Lentigo/fisiopatologia , Transtornos de Fotossensibilidade/fisiopatologia , Células Epiteliais/fisiologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Interleucina-1alfa/fisiologia , Queratinócitos/fisiologia , Melanócitos/fisiologia , Mesoderma/fisiologia , Receptor Cross-Talk/fisiologia , Pele/fisiopatologia
13.
Tissue Eng Part C Methods ; 18(12): 947-57, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22646688

RESUMO

To study human skin pigmentation in a physiological in vitro model, we developed a pigmented reconstructed skin reproducing the three-dimensional architecture of the melanocyte environment and the interactions of melanocyte with its cellular partners, keratinocytes, and fibroblasts. Co-seeding melanocytes and keratinocytes onto a fibroblast-populated collagen matrix led to a correct integration of melanocytes within the epidermal basal layer, but melanocytes remained amelanotic even after supplementation with promelanogenic factors. Interestingly, normalization of keratinocyte differentiation using keratinocyte growth factor instead of epidermal growth factor finally allowed an active pigmentary system to develop, as shown by the expression of key melanogenic markers, the production, and transfer of melanosome-containing melanin into keratinocytes. Various degrees of constitutive pigmentation were reproduced using melanocytes from different skin phenotypes. Furthermore, induction of pigmentation was achieved by treatment with known propigmenting molecules, αMSH and forskolin, thus demonstrating the functionality of the pigmentary system. This pigmented full-thickness skin model therefore represents a highly relevant tool to study the role of cell-cell, cell-matrix, and mesenchymal-epithelial interactions in the control of skin pigmentation.


Assuntos
Colforsina/farmacologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Melanócitos/metabolismo , Pigmentação da Pele , Pele/citologia , alfa-MSH/farmacologia , Animais , Células Cultivadas , Colorimetria , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Pele/metabolismo
14.
Biochem Biophys Res Commun ; 423(4): 775-80, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22713451

RESUMO

Cholesterol 7α-hydroxylase (CYP7A1) is the initial and rate-limiting enzyme for bile acid synthesis. Transcription of the CYP7A1 gene is regulated by bile acids, nuclear receptors and cytokines. Fibroblast growth factor 7 (FGF7) secreted from activated hepatic stellate cells (HSC) during chronic liver fibrosis regulates hepatocyte survival and liver regeneration. In the carbon tetrachloride (CCl(4))-induced fibrotic mouse liver, we demonstrated that the expression of CYP7A1 was largely decreased while the expression of FGF7 was significantly increased. We further demonstrated that FGF7 inhibited CYP7A1 gene expression in hepatocytes. Knockdown study by short interfering RNA, kinase inhibition and phosphorylation assays revealed that the suppression of CYP7A1 expression by FGF7 was mediated by FGFR2 and its downstream JNK signaling cascade. The FGF7 neutralizing antibody restored CYP7A1 expression in Hep3B cells treated with conditioned medium from HSC. In summary, the data suggest that FGF7 is a novel regulator of CYP7A1 expression in hepatocytes and may prevent hepatocytes from accumulating toxic bile acids during liver injury and fibrosis.


Assuntos
Ácidos e Sais Biliares/biossíntese , Colesterol 7-alfa-Hidroxilase/genética , Fator 7 de Crescimento de Fibroblastos/fisiologia , Regulação Enzimológica da Expressão Gênica , Hepatócitos/enzimologia , Cirrose Hepática/enzimologia , Animais , Tetracloreto de Carbono/toxicidade , Linhagem Celular Tumoral , Colesterol 7-alfa-Hidroxilase/antagonistas & inibidores , Modelos Animais de Doenças , Fator 7 de Crescimento de Fibroblastos/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Interferente Pequeno/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo
15.
Ann Dermatol Venereol ; 139 Suppl 4: S148-52, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23522630

RESUMO

Hyperpigmentation of the skin is a common dermatologic condition in all skin types but most prominent in brown-skinned population. In skin of color any inflammation or injury can be accompanied by alterations in pigmentation (hyper/hypo-pigmentation). Postinflammatory hyperpigmentation (PIH) can be observed in many skin conditions including acne, eczema, and contact dermatitis. In the control of skin pigmentation, parallel to the cross-talk between keratinocytes and melanocytes, increasing evidence has underlined the crucial role exerted by the interactions between mesenchymal and epithelial cells through the release of fibroblast-derived growth factors. Among these factors, the keratinocyte growth factor (KGF), alone or in combination with interleukin-1α, induces melanin deposition in vitro and hyperpigmented lesions in vivo. Furthermore, a moderate increase of KGF and a high induction of its receptor have been shown in solar lentigo lesions, suggesting the involvement of this growth factor in the onset of the hyperpigmented spots. Several studies highlight the possible contribution of the fibroblast-derived melanogenic growth factors to the hyperpigmentated lesions, in the context of the mesenchymal - epithelial interactions modulating melanocyte functions.


Assuntos
Hiperpigmentação/etiologia , Inflamação/complicações , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/fisiologia , Técnicas de Cocultura , Colforsina/farmacologia , Citocinas/fisiologia , Epitélio/fisiopatologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Hiperpigmentação/fisiopatologia , Queratinócitos/metabolismo , Lentigo/etiologia , Lentigo/fisiopatologia , Melaninas/metabolismo , Melanócitos/metabolismo , Mesoderma/fisiopatologia , Comunicação Parácrina , Fagocitose , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/fisiologia , Pigmentação da Pele/efeitos dos fármacos , Pigmentação da Pele/fisiologia , Pigmentação da Pele/efeitos da radiação , Fator de Células-Tronco/fisiologia , Luz Solar/efeitos adversos , alfa-MSH/farmacologia
16.
Plast Reconstr Surg ; 128(6): 673e-684e, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22094769

RESUMO

BACKGROUND: The authors have previously demonstrated that normal mice lacking fibroblast growth factor (FGF)-7 can heal cutaneous wounds normally, likely as a result of various compensatory mechanisms. In this study, the authors explored the role of FGF-7 on wound healing in diabetic mice. METHODS: Full-thickness excisional dorsal wounds were created in FGF-7-null diabetic (FGF-7 Lepr, experimental group, n = 8), FGF-7-null (FGF-7 Lepr, FGF-7-null group, n = 8), diabetic (FGF-7 Lepr, diabetic group, n = 11), and wild-type (FGF-7 Lepr, wild-type group, n = 11) mice. Wound closure was followed by digital planimetry. Wound tissues were harvested on day 7 for immunohistochemical staining of cell proliferation (Ki67) and real-time polymerase chain reaction. RESULTS: As expected, the experimental and diabetic groups had significantly slower wound healing than the FGF-7-null or wild-type group. The absence of FGF-7, however, further delayed wound healing in diabetic mice. Curiously, the contraction rate in the experimental group was significantly lower than that in the diabetic group, whereas the epithelialization rate in experimental mice was comparable to that in the diabetic group. Real-time polymerase chain reaction expression of growth factors, including transforming growth factor-ß1, basic fibroblast growth factor, and epidermal growth factor in experimental mice, was also generally lower than that in diabetic mice. CONCLUSIONS: Although the lack of FGF-7 did not appear to affect reepithelialization of cutaneous wounds even in diabetic mice, it significantly reduced the wound contraction rate of healing by further altering the dermal components in diabetic mice. Given the specifically targeted effects of FGF-7 on epithelial cells, the authors' findings suggest that further FGF-7-dependent epithelial-mesenchymal interaction exists that may be important in diabetic wound healing.


Assuntos
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Pele/lesões , Cicatrização/genética , Cicatrização/fisiologia , Animais , Cruzamentos Genéticos , Diabetes Mellitus Experimental/patologia , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real , Pele/patologia
17.
Cell Physiol Biochem ; 27(6): 641-52, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21691082

RESUMO

BACKGROUND/AIM: Previous studies have shown that fibroblast growth factors (FGFs) are involved in the process of liver injury repair. Liver regeneration after partial hepatectomy (PH) is impaired in transgenic mice expressing dominant-negative FGFR2b in hepatocytes. Although FGF7, a ligand specifically bound to FGFR2b, is expressed by activated hepatic stellate cells (HSCs) in fibrotic livers, the expressions and functions of FGF7 and FGFR2b after PH remain unexplored. Therefore, this study sought to examine the potential role of FGF7 signaling during liver regeneration. METHODS: We examined the expression of FGF7 and FGFR2b in normal and regenerating livers. Effects of FGF7 on hepatocytes were examined in vitro using primary hepatocyte culture with FGF7 recombinant protein and in vivo by hydrodynamic-based gene transfer method. RESULTS: We found that FGF7 expression was increased according to the activation status of HSCs after PH. The receptor, FGFR2b, was also increased in hepatocytes during liver regeneration. In vitro treatment with FGF7 protein activated ERK1/2 and promoted proliferation of hepatocytes isolated from regenerating livers. In vivo overexpression of exogenous FGF7 could notably promote hepatic proliferation and activate MAPKs after PH. CONCLUSION: This study suggests a role for activated HSC-expressed FGF7 in stimulating FGF signaling pathways in hepatocytes and regulating liver regeneration.


Assuntos
Fator 7 de Crescimento de Fibroblastos/fisiologia , Regeneração Hepática , Animais , Sequência de Bases , Primers do DNA , Fator 7 de Crescimento de Fibroblastos/genética , Imunofluorescência , Humanos , Masculino , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Clin Cancer Res ; 17(9): 2842-51, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21367751

RESUMO

PURPOSE: Salivary glands are significantly affected when head and neck cancer patients are treated by radiation. We evaluated the effect of human keratinocyte growth factor (hKGF) gene transfer to murine salivary glands on the prevention of radiation-induced salivary hypofunction. EXPERIMENTAL DESIGN: A hybrid serotype 5 adenoviral vector encoding hKGF (AdLTR(2)EF1α-hKGF) was constructed. Female C3H mice, 8 weeks old, were irradiated by single (15 Gy) or fractionated (6 Gy for 5 days) doses to induce salivary hypofunction. AdLTR(2)EF1α-hKGF or AdControl was administered (10(8) - 10(10) particles per gland) to both submandibular glands (SG) by retrograde ductal instillation before irradiation (IR). Salivary flow was measured following pilocarpine stimulation. Human KGF levels were measured by ELISA. SG cell proliferation was measured with bromodeoxyuridine labeling. Endothelial and progenitor or stem cells in SGs were measured by flow cytometry. The effect of SG hKGF production on squamous cell carcinoma (SCC VII) tumor growth was assessed. RESULTS: In 3 separate single-dose IR experiments, salivary flow rates of mice administered the AdLTR(2)EF1α-hKGF vector were not significantly different from nonirradiated control mice (P > 0.05). Similarly, in 3 separate fractionated IR experiments, the hKGF-expressing vector prevented salivary hypofunction dramatically. Transgenic hKGF protein was found at high levels in serum and SG extracts. AdLTR(2)EF1α-hKGF-treated mice showed increased cell proliferation and numbers of endothelial cells, compared with mice treated with AdControl. hKGF gene transfer had no effect on SCC VII tumor growth ± radiation. CONCLUSIONS: hKGF gene transfer prevents salivary hypofunction caused by either single or fractionated radiation dosing in mice. The findings suggest a potential clinical application.


Assuntos
Fator 7 de Crescimento de Fibroblastos/genética , Lesões Experimentais por Radiação/prevenção & controle , Doenças das Glândulas Salivares/prevenção & controle , Glândulas Salivares/fisiopatologia , Glândula Submandibular/metabolismo , Animais , Carcinoma/radioterapia , Feminino , Fator 7 de Crescimento de Fibroblastos/administração & dosagem , Fator 7 de Crescimento de Fibroblastos/fisiologia , Técnicas de Transferência de Genes , Terapia Genética , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Camundongos , Camundongos Endogâmicos C3H , Lesões Experimentais por Radiação/complicações , Radioterapia/efeitos adversos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Doenças das Glândulas Salivares/etiologia , Doenças das Glândulas Salivares/fisiopatologia , Glândulas Salivares/metabolismo , Glândulas Salivares/efeitos da radiação , Glândula Submandibular/patologia , Glândula Submandibular/efeitos da radiação , Células Tumorais Cultivadas
19.
Tumour Biol ; 32(3): 597-602, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21340484

RESUMO

Carcinoma-associated fibroblasts (CAFs) have been confirmed to play an important role in the occurrence and development of many kinds of tumors. Regarding proliferation as one manifestation of malignance, the objective was to observe the effects of oral CAFs on the proliferation of oral squamous carcinoma cells (OSCC) and to explore the role of keratinocyte growth factor (KGF) in this process. The results showed that oral CAFs secreted a higher level of KGF than oral normal fibroblasts (NFs), and the conditioned medium of CAFs could increase the viability of carcinoma cells and promote more of them into G2 and S phase. However, after blocking with KGF antibody, the viability and cell cycle of Tca8113 cultured with CAFs conditioned medium changed to be similar with NFs control groups. It was concluded that CAFs could promote the proliferation of OSCC through secreting high levels of KGF. These findings support the use of carcinoma-associated fibroblasts as a novel target in anticancer therapy.


Assuntos
Carcinoma de Células Escamosas/patologia , Proliferação de Células , Fator 7 de Crescimento de Fibroblastos/fisiologia , Fibroblastos/fisiologia , Neoplasias Bucais/patologia , Comunicação Celular , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Fibroblastos/citologia , Humanos
20.
In Vitro Cell Dev Biol Anim ; 47(2): 173-81, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21082280

RESUMO

A variety of cytokines have been detected in inflamed intestinal mucosal tissues, including the pro-inflammatory cytokine, interleukin-1 (IL-1), along with growth factors involved in wound healing processes such as proliferation and cell migration. However, little is known about how IL-1 and growth factors interact with intestinal epithelial cells to regulate the production of inflammatory cytokines such as interleukin-8 (IL-8). Previously, we have shown that hepatocyte growth factor (HGF) could significantly enhance IL-1-stimulated IL-8 secretion by the Caco-2 colonic epithelial cell line, yet HGF, by itself, did not stimulate IL-8 secretion. In this report, a second growth factor, keratinocyte growth factor (KGF), was also found to significantly enhance IL-1-induced IL-8 secretion by Caco-2 cells, yet KGF, by itself, also had no effect. Simultaneous addition of both IL-1 and KGF was also required for the enhancing effect. Treatment of the Caco-2 cells with wortmannin or triciribine suppressed the enhancing effect of HGF, suggesting that the effect was mediated by signaling through phosphatidylinositol-3-kinase (PI3K) and the kinase AKT. The enhancing effect of KGF was not affected by wortmannin, but was suppressed by triciribine, suggesting that the effect of KGF was through a PI3K-independent activation of AKT. These results suggest that the growth factors HGF and KGF may play a role in enhancing IL-1-stimulated production of IL-8 by epithelial cells during mucosal inflammations. However, the mechanism by which the growth factors enhance the IL-1 response may be through different initial signaling pathways.


Assuntos
Fator 7 de Crescimento de Fibroblastos/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Interleucina-8/metabolismo , Mucosa Intestinal/metabolismo , Androstadienos/farmacologia , Células CACO-2 , Movimento Celular , Fator 7 de Crescimento de Fibroblastos/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Interleucina-1/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/genética , Wortmanina , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...