Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
PLoS Biol ; 17(1): e2006972, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30695023

RESUMEN

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.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Animales , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Factor 7 de Crecimiento de Fibroblastos/fisiología , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/fisiología , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Ratones , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/fisiología
2.
Am J Respir Cell Mol Biol ; 57(2): 162-173, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28296468

RESUMEN

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.


Asunto(s)
Células Epiteliales Alveolares/citología , Factor 7 de Crecimiento de Fibroblastos/fisiología , Linfocitos T Reguladores/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Traslado Adoptivo , Células Epiteliales Alveolares/patología , Anfirregulina/biosíntesis , Anfirregulina/genética , Animales , División Celular , Técnicas de Cocultivo , Toxina Diftérica/toxicidad , Factor 7 de Crecimiento de Fibroblastos/biosíntesis , Factor 7 de Crecimiento de Fibroblastos/genética , Factores de Transcripción Forkhead/análisis , Regulación de la Expresión Génica/inmunología , Humanos , Lipopolisacáridos/toxicidad , Pulmón/citología , Depleción Linfocítica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Neumonectomía , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/metabolismo , Complicaciones Posoperatorias/patología , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante
3.
Dev Biol ; 419(2): 348-356, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27590203

RESUMEN

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).


Asunto(s)
Ectodisplasinas/fisiología , Glándulas Exocrinas/embriología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Animales , Ectodisplasinas/deficiencia , Ectodisplasinas/genética , Resección Endoscópica de la Mucosa , Glándulas Exocrinas/metabolismo , Glándulas Exocrinas/ultraestructura , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/fisiología , Masculino , Seno Maxilar/embriología , Seno Maxilar/ultraestructura , Mesodermo/metabolismo , Ratones , Morfogénesis , Mucosa Nasal/embriología , Mucosa Nasal/ultraestructura , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética
4.
Am J Physiol Lung Cell Mol Physiol ; 310(9): L868-79, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26919897

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Factor 7 de Crecimiento de Fibroblastos/fisiología , Inmunidad Innata , Macrófagos Alveolares/inmunología , Neumonía Bacteriana/inmunología , Animales , Células Cultivadas , Colectinas/genética , Colectinas/metabolismo , Infecciones por Escherichia coli/metabolismo , Femenino , Expresión Génica , Humanos , Macrófagos Alveolares/microbiología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Muramidasa/genética , Muramidasa/metabolismo , Neumonía Bacteriana/metabolismo , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/microbiología
5.
Br J Dermatol ; 174(3): 533-41, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26471375

RESUMEN

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.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/fisiología , Fibroblastos/metabolismo , Fibronectinas/biosíntesis , Psoriasis/metabolismo , Adolescente , Adulto , Estudios de Casos y Controles , Células Cultivadas , Fibronectinas/metabolismo , Voluntarios Sanos , Humanos , Queratinocitos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Melanocitos/metabolismo , Persona de Mediana Edad , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3 , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
6.
J Cell Mol Med ; 18(9): 1895-907, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25088572

RESUMEN

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.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/administración & dosificación , Enfermedades Vaginales/tratamiento farmacológico , Administración Tópica , Animales , Proliferación Celular , Epitelio/patología , Estradiol/fisiología , Femenino , Factor 7 de Crecimiento de Fibroblastos/fisiología , Humanos , Células MCF-7 , Ratones , Membrana Mucosa/patología , Ovariectomía , Transducción de Señal , Vagina/patología
7.
Biochem Biophys Res Commun ; 423(4): 775-80, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22713451

RESUMEN

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.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Factor 7 de Crecimiento de Fibroblastos/fisiología , Regulación Enzimológica de la Expresión Génica , Hepatocitos/enzimología , Cirrosis Hepática/enzimología , Animales , Tetracloruro de Carbono/toxicidad , Línea Celular Tumoral , Colesterol 7-alfa-Hidroxilasa/antagonistas & inhibidores , Modelos Animales de Enfermedad , Factor 7 de Crecimiento de Fibroblastos/farmacología , Hepatocitos/efectos de los fármacos , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , ARN Interferente Pequeño/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo
8.
Ann Dermatol Venereol ; 139 Suppl 3: S96-101, 2012 Nov.
Artículo en Francés | MEDLINE | ID: mdl-23260525

RESUMEN

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.


Asunto(s)
Dermatitis/fisiopatología , Hiperpigmentación/fisiopatología , Lentigo/fisiopatología , Trastornos por Fotosensibilidad/fisiopatología , Células Epiteliales/fisiología , Factor 7 de Crecimiento de Fibroblastos/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Interleucina-1alfa/fisiología , Queratinocitos/fisiología , Melanocitos/fisiología , Mesodermo/fisiología , Receptor Cross-Talk/fisiología , Piel/fisiopatología
9.
Ann Dermatol Venereol ; 139 Suppl 4: S148-52, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23522630

RESUMEN

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.


Asunto(s)
Hiperpigmentación/etiología , Inflamación/complicaciones , Células Cultivadas/efectos de los fármacos , Células Cultivadas/fisiología , Técnicas de Cocultivo , Colforsina/farmacología , Citocinas/fisiología , Epitelio/fisiopatología , Factor 7 de Crecimiento de Fibroblastos/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Hiperpigmentación/fisiopatología , Queratinocitos/metabolismo , Lentigo/etiología , Lentigo/fisiopatología , Melaninas/metabolismo , Melanocitos/metabolismo , Mesodermo/fisiopatología , Comunicación Paracrina , Fagocitosis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Pigmentación de la Piel/efectos de los fármacos , Pigmentación de la Piel/fisiología , Pigmentación de la Piel/efectos de la radiación , Factor de Células Madre/fisiología , Luz Solar/efectos adversos , alfa-MSH/farmacología
10.
Cell Physiol Biochem ; 27(6): 641-52, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21691082

RESUMEN

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.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/fisiología , Regeneración Hepática , Animales , Secuencia de Bases , Cartilla de ADN , Factor 7 de Crecimiento de Fibroblastos/genética , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Tumour Biol ; 32(3): 597-602, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21340484

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas/patología , Proliferación Celular , Factor 7 de Crecimiento de Fibroblastos/fisiología , Fibroblastos/fisiología , Neoplasias de la Boca/patología , Comunicación Celular , Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular , Fibroblastos/citología , Humanos
12.
Int J Biol Sci ; 17(14): 3862-3874, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671204

RESUMEN

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.


Asunto(s)
Comunicación Celular/fisiología , Conexina 43/metabolismo , Factor 7 de Crecimiento de Fibroblastos/fisiología , Glicoproteínas de Membrana/metabolismo , Células 3T3 , Animales , Ligandos , Sistema de Señalización de MAP Quinasas , Ratones , Osteoblastos/metabolismo
13.
J Cell Biochem ; 109(4): 737-46, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20069574

RESUMEN

Keratinocyte growth factor (KGF; also known as FGF-7) is a well-characterized paracrine growth factor for tissue growth and regeneration. However, its role in adipose tissue, which is known to undergo tremendous expansion in obesity, is virtually unknown. Given that we previously identified KGF as one of the up-regulated growth factors in adipose tissue of an early-life programmed rat model of visceral obesity, the present study was undertaken to examine the hypothesis that KGF promotes adipogenesis. Using 3T3-L1 and rat primary preadipocytes as in vitro model systems, we demonstrated that (1) KGF stimulated preadipocyte proliferation in a concentration-dependent manner with a maximal effect at 2.5 ng/ml (approximately 2-fold increase); (2) KGF mRNA was highly expressed in rat adipocytes and preadipocytes as well as 3T3-L1 cells; (3) treatment of preadipocytes with a neutralizing antibody against KGF and siRNA-mediated knockdown of KGF led to a 50% reduction in their proliferative capacity; (4) KGF activated the protein kinase Akt, and the PI3 kinase inhibitor LY294002 blocked KGF stimulation of preadipocyte proliferation; and (5) KGF did not promote differentiation of preadipocytes to mature adipocytes. Together, these results reveal adipocytes and their precursor cells as novel sites of KGF production. Importantly, they also demonstrate that KGF promotes preadipocyte proliferation by an autocrine mechanism that involves activation of the PI3K/Akt signaling pathway. Aberrant KGF expression may have consequences not only for normal adipose tissue growth but also for the pathogenesis of obesity.


Asunto(s)
Adipocitos/citología , Adipogénesis , Comunicación Autocrina , Proliferación Celular , Factor 7 de Crecimiento de Fibroblastos/fisiología , Células 3T3-L1 , Adipocitos/química , Adipocitos/metabolismo , Animales , Células Cultivadas , Factor 7 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/farmacología , Ratones , Obesidad Abdominal/etiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero , Ratas , Células Madre/citología
14.
Am J Physiol Renal Physiol ; 299(3): F479-86, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20591940

RESUMEN

Identification of renal cell progenitors and recognition of the events contributing to cell regeneration following ischemia-reperfusion injury (IRI) are a major challenge. In a mouse model of unilateral renal IRI, we demonstrated that the first cells to proliferate within injured kidneys were urothelial cells expressing the progenitor cell marker cytokeratin 14. A systematic cutting of the injured kidney revealed that these urothelial cells were located in the deep cortex at the corticomedullary junction in the vicinity of lobar vessels. Contrary to multilayered bladder urothelium, these intrarenal urothelial cells located in the upper part of the medulla constitute a monolayered barrier and express among uroplakins only uroplakin III. However, like bladder progenitors, intrarenal urothelial cells proliferated through a FGF receptor-2 (FGFR2)-mediated process. They strongly expressed FGFR2 and proliferated in vivo after recombinant FGF7 administration to control mice. In addition, IRI led to FGFR phosphorylation together with the selective upregulation of FGF7 and FGF2. Conversely, by day 2 following IRI, renal urothelial cell proliferation was significantly inhibited by FGFR2 antisense oligonucleotide administration into an intrarenal urinary space. Of notice, no significant migration of these early dividing urothelial cells was detected in the cortex within 7 days following IRI. Thus our data show that following IRI, proliferation of urothelial cells is mediated by the FGFR2 pathway and precedes tubular cell proliferation, indicating a particular sensitivity of this structure to changes caused by the ischemic process.


Asunto(s)
Proliferación Celular , Corteza Renal/patología , Daño por Reperfusión/patología , Animales , Modelos Animales de Enfermedad , Femenino , Factor 7 de Crecimiento de Fibroblastos/fisiología , Ratones , Ratones Endogámicos C57BL , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Daño por Reperfusión/fisiopatología , Transducción de Señal/fisiología , Urotelio/patología
15.
Curr Opin Rheumatol ; 21(6): 649-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19667993

RESUMEN

PURPOSE OF REVIEW: The lung in systemic sclerosis (scleroderma) is susceptible to fibrosis and the ensuing respiratory insufficiency contributes to significant morbidity and mortality in this disease. The lack of effective therapies for pulmonary fibrosis has spurred a re-evaluation of pathobiological paradigms and therapeutic strategies in scleroderma-associated interstitial lung disease and in idiopathic pulmonary fibrosis. The purpose of this review is to examine emerging new therapeutic targets that modulate pro-fibrotic phenotypes of tissue-resident cells and the associated aberrant tissue remodeling responses in fibrotic disorders. RECENT FINDINGS: Progressive forms of tissue fibrosis, including scleroderma, are characterized by an accumulation of activated mesenchymal cells and their secreted extracellular matrix proteins in association with dysrepair of epithelial and endothelial cells. Recent studies suggest that emergence of cellular phenotypes that perpetuate loss of cellular homeostasis is characteristic of many fibrosis-related clinical syndromes. SUMMARY: Therapeutic strategies that modulate the fate/phenotype of reparative structural cells, including epithelial, endothelial, and mesenchymal cells, offer new opportunities for the development of more effective drugs for the treatment of fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/terapia , Esclerodermia Sistémica/complicaciones , Factor 4E Eucariótico de Iniciación/fisiología , Factor 7 de Crecimiento de Fibroblastos/fisiología , Fibrosis , Factor de Crecimiento de Hepatocito/fisiología , Homeostasis , Humanos , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/fisiopatología , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , Óxido Nítrico/fisiología , PPAR gamma/agonistas , Fenotipo , Inhibidores de Proteínas Quinasas/uso terapéutico , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/fisiopatología , Esclerodermia Sistémica/terapia , Transducción de Señal , Trasplante de Células Madre
16.
Ann Surg ; 249(5): 824-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19387319

RESUMEN

OBJECTIVE: To determine precisely the role of parathyroid hormone (PTH) and of phosphatonins in the genesis of posthepatectomy hypophosphatemia. BACKGROUND: Posthepatectomy hypophosphatemia has recently been related to increased renal fractional excretion of phosphate (FE P). To address the cause of hypophosphatemia, we measured serum concentrations of PTH, various phosphatonins, and the number of removed hepatic segment in patients with this disorder. METHODS: Serum phosphate (PO4), ionized calcium (Ca++), HCO3-, pH and FE P, intact PTH (I-PTH), carboxyl-terminal fibroblast growth factor 23 (C-FGF-23) and intact fibroblast growth factor 23 (I-FGF-23), FGF-7, and secreted frizzled related-protein-4 (sFRP-4) were measured before and on postoperative (po) days 1, 2, 3, 5, and 7, in 18 patients undergoing liver resection. The number of removed hepatic segments was also assessed. RESULTS: Serum PO4 concentrations decreased within 24 hours, were lowest (0.66 +/- 0.03 mmol/L; P < 0.001) at 48 hours, and returned to normal within 5 days of the procedure. FE P peaked at 25.07% +/- 2.26% on po day 1 (P < 0.05). Decreased ionized calcium concentrations (1.10 +/- 0.01 mmol/L; P < 0.01) were observed on po day 1 and were negatively correlated with increased I-PTH concentrations (8.8 +/- 0.9 pmol/L; P < 0.01; correlation: r = -0.062, P = 0.016). FE P was positively related to I-PTH levels on po day 1 (r = 0.52, P = 0.047) and negatively related to PO4 concentrations (r = -0.56, P = 0.024). Severe hypophosphatemia and increased urinary phosphate excretion persisted for 72 hours even when I-PTH concentrations had returned to normal. I-FGF-23 decreased to its nadir of 7.8 +/- 6.9 pg/mL (P < 0.001) on po day 3 and was correlated with PO4 levels on po days 0, 3, 5, and 7 (P < 0.001). C-FGF-23, FGF-7 and sFRP-4 levels could not be related to either PO4 concentrations or FE P. CONCLUSION: Posthepatectomy hypophosphatemia is associated with increased FE P unrelated to I-FGF-23 or C-FGF-23, FGF-7, or sFRP-4. I-PTH contributes to excessive FE P partially on po day 1 but not thereafter. Other yet defined factors should explain post hepatectomy hypophosphatemia.


Asunto(s)
Hepatectomía/efectos adversos , Hipofosfatemia/fisiopatología , Enfermedades Renales/fisiopatología , Riñón/fisiopatología , Adulto , Anciano , Femenino , Factor 7 de Crecimiento de Fibroblastos/sangre , Factor 7 de Crecimiento de Fibroblastos/fisiología , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Hipofosfatemia/etiología , Enfermedades Renales/etiología , Masculino , Persona de Mediana Edad , Hormona Paratiroidea/sangre , Hormona Paratiroidea/fisiología , Fosfatos/sangre , Fosfatos/fisiología , Fosfatos/orina , Proteínas Proto-Oncogénicas/sangre , Proteínas Proto-Oncogénicas/fisiología
17.
Eur J Dermatol ; 19(5): 469-73, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19505863

RESUMEN

Two patients with a generalized, progressive dyschromatosis disorder are described and investigated as a model to study the role of fibroblast-derived mediators on skin pigmentation. The patients (father and daughter) had had a widespread hyperpigmentation since early life which then progressively worsened with the appearance of hyperpigmented macules, café-au-lait macules and freckles, also involving the lips, palms and soles, intermixed with small hypopigmented spots. These features resembled those of familial progressive hyperpigmentation (FPH). Histology revealed a normal epidermis with pronounced keratinocyte hyperpigmentation and the presence of dermal melanophages. Ultrastructural analysis showed basal and suprabasal keratinocytes enriched in melanosome complexes. Immunohistochemical staining displayed an increased expression of hepatocyte growth factor (HGF), stem cell factor (SCF) and keratinocyte growth factor (KGF) in fibroblast-like cells of the upper dermis in hyperpigmented lesions of both patients, compared to control healthy skin. Our data suggest that a persistent activation of fibroblasts abnormally stimulating melanocyte functions is involved in hyperpigmentation disorders.


Asunto(s)
Factor 7 de Crecimiento de Fibroblastos/fisiología , Fibroblastos/química , Factor de Crecimiento de Hepatocito/fisiología , Hiperpigmentación/genética , Factor de Células Madre/fisiología , Adulto , Femenino , Factor 7 de Crecimiento de Fibroblastos/análisis , Factor de Crecimiento de Hepatocito/análisis , Humanos , Hiperpigmentación/etiología , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Factor de Células Madre/análisis
18.
Am J Respir Cell Mol Biol ; 38(2): 239-46, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17872496

RESUMEN

Rat alveolar epithelial cells (AEC) in primary culture transdifferentiate from a type II (AT2) toward a type I (AT1) cell-like phenotype, a process that can be both prevented and reversed by keratinocyte growth factor (KGF). Microarray analysis revealed that these effects of KGF are associated with up-regulation of key molecules in the mitogen-activated protein kinase (MAPK) pathway. To further explore the role of three key MAPK (i.e., extracellular signal-related kinase [ERK] 1/2, c-Jun N-terminal kinase [JNK] and p38) in mediating effects of KGF on AEC phenotype, primary rat AEC cultivated in minimal defined serum-free medium (MDSF) were treated with KGF (10 ng/ml) from Day 4 for intervals up to 48 hours. Exposure to KGF activated all three MAPK, JNK, ERK1/2, and p38. Inhibition of JNK, but not of ERK1/2 or p38, abrogated the ability of KGF to maintain the AT2 cell phenotype, as evidenced by loss of expression of lamellar membrane protein (p180) and increased reactivity with the AT1 cell-specific monoclonal antibody VIIIB2 by Day 6 in culture. Overexpression of JNKK2, upstream kinase of JNK, increased activation of endogenous c-Jun in association with increased expression of p180 and abrogation of AQP5, suggesting that activation of c-Jun promotes retention of the AT2 cell phenotype. These results indicate that retention of the AT2 cell phenotype by KGF involves c-Jun and suggest that activation of c-Jun kinase may be an important determinant of maintenance of AT2 cell phenotype.


Asunto(s)
Diferenciación Celular/fisiología , Factor 7 de Crecimiento de Fibroblastos/fisiología , MAP Quinasa Quinasa 4/metabolismo , Alveolos Pulmonares/citología , Transducción de Señal , Animales , Western Blotting , Transdiferenciación Celular , Células Epiteliales/citología , Masculino , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Sprague-Dawley
19.
PLoS Med ; 5(1): e19, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18232728

RESUMEN

BACKGROUND: Important support functions, including promotion of tumor growth, angiogenesis, and invasion, have been attributed to the different cell types populating the tumor stroma, i.e., endothelial cells, cancer-associated fibroblasts, pericytes, and infiltrating inflammatory cells. Fibroblasts have long been recognized inside carcinomas and are increasingly implicated as functional participants. The stroma is prominent in cervical carcinoma, and distinguishable from nonmalignant tissue, suggestive of altered (tumor-promoting) functions. We postulated that pharmacological targeting of putative stromal support functions, in particular those of cancer-associated fibroblasts, could have therapeutic utility, and sought to assess the possibility in a pre-clinical setting. METHODS AND FINDINGS: We used a genetically engineered mouse model of cervical carcinogenesis to investigate platelet-derived growth factor (PDGF) receptor signaling in cancer-associated fibroblasts and pericytes. Pharmacological blockade of PDGF receptor signaling with the clinically approved kinase inhibitor imatinib slowed progression of premalignant cervical lesions in this model, and impaired the growth of preexisting invasive carcinomas. Inhibition of stromal PDGF receptors reduced proliferation and angiogenesis in cervical lesions through a mechanism involving suppression of expression of the angiogenic factor fibroblast growth factor 2 (FGF-2) and the epithelial cell growth factor FGF-7 by cancer-associated fibroblasts. Treatment with neutralizing antibodies to the PDGF receptors recapitulated these effects. A ligand trap for the FGFs impaired the angiogenic phenotype similarly to imatinib. Thus PDGF ligands expressed by cancerous epithelia evidently stimulate PDGFR-expressing stroma to up-regulate FGFs, promoting angiogenesis and epithelial proliferation, elements of a multicellular signaling network that elicits functional capabilities in the tumor microenvironment. CONCLUSIONS: This study illustrates the therapeutic benefits in a mouse model of human cervical cancer of mechanism-based targeting of the stroma, in particular cancer-associated fibroblasts. Drugs aimed at stromal fibroblast signals and effector functions may prove complementary to conventional treatments targeting the overt cancer cells for a range of solid tumors, possibly including cervical carcinoma, the second most common lethal malignancy in women worldwide, for which management remains poor.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma/irrigación sanguínea , Proteínas de Neoplasias/fisiología , Neovascularización Patológica/tratamiento farmacológico , Comunicación Paracrina/fisiología , Piperazinas/uso terapéutico , Factor de Crecimiento Derivado de Plaquetas/fisiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Neoplasias del Cuello Uterino/irrigación sanguínea , Animales , Antineoplásicos/farmacología , Benzamidas , Carcinoma/tratamiento farmacológico , Carcinoma/fisiopatología , Carcinoma/virología , Células Epiteliales/metabolismo , Estradiol/administración & dosificación , Estradiol/toxicidad , Femenino , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/fisiología , Factor 7 de Crecimiento de Fibroblastos/biosíntesis , Factor 7 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/fisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/metabolismo , Papillomavirus Humano 16 , Humanos , Mesilato de Imatinib , Ratones , Ratones Endogámicos , Ratones Transgénicos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neovascularización Patológica/fisiopatología , Comunicación Paracrina/efectos de los fármacos , Pericitos/metabolismo , Piperazinas/farmacología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Células del Estroma/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/fisiopatología , Neoplasias del Cuello Uterino/virología
20.
Clin Cancer Res ; 13(16): 4713-20, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699848

RESUMEN

PURPOSE: Fibroblast growth factor (FGF) signals play fundamental roles in development and tumorigenesis. Thyroid cancer is an example of a tumor with nonoverlapping genetic mutations that up-regulate mitogen-activated protein kinase. We reported recently that FGF receptor 2 (FGFR2) is down-regulated through extensive DNA promoter methylation in thyroid cancer. Reexpression of the FGFR2-IIIb isoform impedes signaling upstream of the BRAF/mitogen-activated protein kinase pathway to interrupt tumor progression. In this analysis, we examined a novel target of FGFR2-IIIb signaling, melanoma-associated antigen-A3 and A6 (MAGE-A3/6). EXPERIMENTAL DESIGN: cDNA microarray analysis was done on human WRO thyroid cancer cells transfected with FGFR2-IIIb or empty vector. Identified gene target was confirmed by reverse transcription-PCR and Western blotting. Gene regulation was examined by treatment of WRO cells with the methylation inhibitor 5'-azacytidine followed by methylation-specific PCR and reverse transcription-PCR and by chromatin immunoprecipitation. RESULTS: Gene expression profiling identified the cancer/testis antigen MAGE-A3/6 as a novel target of FGFR2-IIIb signaling. MAGE-A3/6 regulation was mediated through DNA methylation and chromatin modifications. In particular, FGF7/FGFR2-IIIb activation resulted in histone 3 methylation and deacetylation associated with the MAGE-A3/6 promoter to down-regulate gene expression. CONCLUSIONS: These data unmask a complex repertoire of epigenetically controlled signals that govern FGFR2-IIIb and MAGE-A3/6 expression. Our findings provide insights into the interrelationship between novel tumor markers that may also represent overlapping therapeutic targets.


Asunto(s)
Antígenos de Neoplasias/genética , Histonas/metabolismo , Proteínas de Neoplasias/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Neoplasias de la Tiroides/metabolismo , Línea Celular Tumoral , Metilación de ADN , Epigénesis Genética , Factor 7 de Crecimiento de Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Isoformas de Proteínas/fisiología , Transducción de Señal , Neoplasias de la Tiroides/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA