Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Dis Model Mech ; 15(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35833364

RESUMO

Fibroblast growth factor 8 (FGF8), acting through the fibroblast growth factor receptor 1 (FGFR1), has an important role in the development of gonadotropin-releasing hormone-expressing neurons (GnRH neurons). We hypothesized that FGF8 regulates differentiation of human GnRH neurons in a time- and dose-dependent manner via FGFR1. To investigate this further, human pluripotent stem cells were differentiated during 10 days of dual-SMAD inhibition into neural progenitor cells, followed either by treatment with FGF8 at different concentrations (25 ng/ml, 50 ng/ml or 100 ng/ml) for 10 days or by treatment with 100 ng/ml FGF8 for different durations (2, 4, 6 or 10 days); cells were then matured through DAPT-induced inhibition of Notch signaling for 5 days into GnRH neurons. FGF8 induced expression of GNRH1 in a dose-dependent fashion and the duration of FGF8 exposure correlated positively with gene expression of GNRH1 (P<0.05, Rs=0.49). However, cells treated with 100 ng/ml FGF8 for 2 days induced the expression of genes, such as FOXG1, ETV5 and SPRY2, and continued FGF8 treatment induced the dynamic expression of several other genes. Moreover, during exposure to FGF8, FGFR1 localized to the cell surface and its specific inhibition with the FGFR1 inhibitor PD166866 reduced expression of GNRH1 (P<0.05). In neurons, FGFR1 also localized to the nucleus. Our results suggest that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny. This article has an associated First Person interview with the first author of the paper.


Assuntos
Hormônio Liberador de Gonadotropina , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Fator 8 de Crescimento de Fibroblasto/metabolismo , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Neurônios/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo
2.
Endocrinology ; 162(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34125902

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus play a key role in the regulation of reproductive function. In this study, we sought an efficient method for generating GnRH neurons from human embryonic and induced pluripotent stem cells (hESC and hiPSC, respectively). First, we found that exposure of primitive neuroepithelial cells, rather than neuroprogenitor cells, to fibroblast growth factor 8 (FGF8), was more effective in generating GnRH neurons. Second, addition of kisspeptin to FGF8 further increased the efficiency rates of GnRH neurogeneration. Third, we generated a fluorescent marker mCherry labeled human embryonic GnRH cell line (mCh-hESC) using a CRISPR-Cas9 targeting approach. Fourth, we examined physiological characteristics of GnRH (mCh-hESC) neurons: similar to GnRH neurons in vivo, they released the GnRH peptide in a pulsatile manner at ~60 min intervals; GnRH release increased in response to high potassium, kisspeptin, estradiol, and neurokinin B challenges; and injection of depolarizing current induced action potentials. Finally, we characterized developmental changes in transcriptomes of GnRH neurons using hESC, hiPSC, and mCh-hESC. The developmental pattern of transcriptomes was remarkably similar among the 3 cell lines. Collectively, human stem cell-derived GnRH neurons will be an important tool for establishing disease models to understand diseases, such as idiopathic hypothalamic hypogonadism, and testing contraceptive drugs.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Células-Tronco Embrionárias Humanas/fisiologia , Neurogênese/genética , Neurônios/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Fator 8 de Crescimento de Fibroblasto/farmacologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/fisiologia , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transcriptoma/efeitos dos fármacos
3.
Dis Model Mech ; 13(3)2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31996360

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons provide a fundamental signal for the onset of puberty and subsequent reproductive functions by secretion of gonadotropin-releasing hormone. Their disrupted development or function leads to congenital hypogonadotropic hypogonadism (CHH). To model the development of human GnRH neurons, we generated a stable GNRH1-TdTomato reporter cell line in human pluripotent stem cells (hPSCs) using CRISPR-Cas9 genome editing. RNA-sequencing of the reporter clone, differentiated into GnRH neurons by dual SMAD inhibition and FGF8 treatment, revealed 6461 differentially expressed genes between progenitors and GnRH neurons. Expression of the transcription factor ISL1, one of the top 50 most upregulated genes in the TdTomato-expressing GnRH neurons, was confirmed in 10.5 gestational week-old human fetal GnRH neurons. Among the differentially expressed genes, we detected 15 genes that are implicated in CHH and several genes that are implicated in human puberty timing. Finally, FGF8 treatment in the neuronal progenitor pool led to upregulation of 37 genes expressed both in progenitors and in TdTomato-expressing GnRH neurons, which suggests upstream regulation of these genes by FGF8 signaling during GnRH neuron differentiation. These results illustrate how hPSC-derived human GnRH neuron transcriptomic analysis can be utilized to dissect signaling pathways and gene regulatory networks involved in human GnRH neuron development.This article has an associated First Person interview with the first author of the paper.


Assuntos
Genes Reporter , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transcriptoma/genética , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Linhagem Celular , Feto/citologia , Fator 8 de Crescimento de Fibroblasto/farmacologia , Humanos , Hipogonadismo/genética , Proteínas com Homeodomínio LIM/metabolismo , Neurônios/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
4.
J Proteomics ; 183: 14-24, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29758290

RESUMO

FGF8 specifies early tooth development by directing the migration of the early tooth founder cells to the site of tooth emergence. To date the effect of the FGF8 in adult dental pulp has not been studied. We have assessed the regenerative potential of FGF8 by evaluating changes in the proteome landscape of dental pulp following short- and long-term exposure to recombinant FGF8 protein. In addition, we carried out qRT PCR analysis to determine extracellular/adhesion gene marker expression and assessed cell proliferation and mineralization in response to FGF8 treatment. 2D and mass spectrometry data showed differential expression of proteins implicated in cytoskeleton/ECM remodeling and migration, cell proliferation and odontogenic differentiation as evidenced by the upregulation of gelsolin, moesin, LMNA, WDR1, PLOD2, COPS5 and downregulation of P4HB. qRT PCR showed downregulation of proteins involved in cell-matrix adhesion such as ADAMTS8, LAMB3 and ANOS1 and increased expression of the angiogenesis marker PECAM1. We have observed that, FGF8 treatment was able to boost dental pulp cell proliferation and to enhance dental pulp mineralization. Collectively, our data suggest that, FGF8 treatment could promote endogenous healing of the dental pulp via recruitment of dental pulp progenitors as well as by promoting their angiogenic and odontogenic differentiation. SIGNIFICANCE: Dental pulp cells (DP) have been studied extensively for the purposes of mineralized tissue repair, particularly for the reconstruction of hard and soft tissue maxillofacial defects. Canonical FGF signaling has been implicated throughout multiple stages of tooth development by regulating cell proliferation, differentiation, survival as well as cellular migration. FGF8 expression is indispensible for normal tooth development and particularly for the migration of early tooth progenitors to the sites of tooth emergence. The present study provides proteome and qRT PCR data with regard to the future application and biological relevance of FGF8 in dental regenerative medicine. AUTHORS WITH ORCID: Rozaliya Tsikandelova - 0000-0003-0178-3767 Zornitsa Mihaylova - 0000-0003-1748-4489 Sébastien Planchon - 0000-0002-0455-0574 Nikolay Ishkitiev - 0000-0002-4351-5579.


Assuntos
Polpa Dentária/citologia , Fator 8 de Crescimento de Fibroblasto/farmacologia , Proteoma/metabolismo , Regeneração/efeitos dos fármacos , Adulto , Diferenciação Celular , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Polpa Dentária/efeitos dos fármacos , Polpa Dentária/fisiologia , Regulação da Expressão Gênica , Humanos , Minerais/metabolismo , Reação em Cadeia da Polimerase , Proteoma/efeitos dos fármacos , Proteoma/fisiologia
5.
Exp Eye Res ; 173: 129-137, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29753729

RESUMO

Emerging therapies have begun to evaluate the abilities of Müller glial cells (MGCs) to protect and/or regenerate neurons following retina injury. The migration of donor cells is central to many reparative strategies, where cells must achieve appropriate positioning to facilitate localized repair. Although chemical cues have been implicated in the MGC migratory responses of numerous retinopathies, MGC-based therapies have yet to explore the extent to which external biochemical stimuli can direct MGC behavior. The current study uses a microfluidics-based assay to evaluate the migration of cultured rMC-1 cells (as model MGC) in response to quantitatively-controlled microenvironments of signaling factors implicated in retinal regeneration: basic Fibroblast Growth factor (bFGF or FGF2); Fibroblast Growth factor 8 (FGF8); Vascular Endothelial Growth Factor (VEGF); and Epidermal Growth Factor (EGF). Findings indicate that rMC-1 cells exhibited minimal motility in response to FGF2, FGF8 and VEGF, but highly-directional migration in response to EGF. Further, the responses were blocked by inhibitors of EGF-R and of the MAPK signaling pathway. Significantly, microfluidics data demonstrate that changes in the EGF gradient (i.e. change in EGF concentration over distance) resulted in the directional chemotactic migration of the cells. By contrast, small increases in EGF concentration, alone, resulted in non-directional cell motility, or chemokinesis. This microfluidics-enhanced approach, incorporating the ability both to modulate and asses the responses of motile donor cells to a range of potential chemotactic stimuli, can be applied to potential donor cell populations obtained directly from human specimens, and readily expanded to incorporate drug-eluting biomaterials and combinations of desired ligands.


Assuntos
Quimiotaxia/fisiologia , Células Ependimogliais/fisiologia , Animais , Proliferação de Células/fisiologia , Células Cultivadas , Microambiente Celular , Células Ependimogliais/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Cloridrato de Erlotinib/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator 8 de Crescimento de Fibroblasto/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Técnicas Analíticas Microfluídicas , Nestina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Fatores de Crescimento/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
Tumour Biol ; 39(3): 1010428317695969, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28345462

RESUMO

Fibroblast growth factor 8b and androgen play important roles in cell proliferation of prostate cancer. We investigated the effects of fibroblast growth factor 8b and androgen on the proliferation of prostate cell lines and the corresponding intracellular mechanisms. It is found that dihydrotestosterone and fibroblast growth factor 8b stimulated Lncap cell mitosis in a concentration-responsive manner, with 30 ng/mL as the most suitable concentration, respectively. Dihydrotestosterone treatment alone did not enhance the expression and phosphorylation level of fibroblast growth factor receptor but significantly enhanced the level of fibroblast growth factor receptor phosphorylation elicited by fibroblast growth factor 8b. Phosphorylations of extracellular signal-regulated kinase, p38, and c-Jun NH2-terminal kinase were stimulated by dihydrotestosterone or fibroblast growth factor 8b. Among these major downstream pathways for mitogen-activated protein kinase, c-Jun NH2-terminal kinase signaling was most significantly enhanced. Protein kinase C phosphorylation was higher than AKT by the combined stimulation of dihydrotestosterone and fibroblast growth factor 8b. The phosphorylation of CDC2 was significantly induced by dihydrotestosterone and fibroblast growth factor 8b synergetically, and Smad underwent the same induction as CDC2. So the promoting effect of fibroblast growth factor 8b on cell cycle might contribute to the G2/M transition. This study indicated that the functional interaction between fibroblast growth factor 8b and androgen was essential for the prostate cancer cell proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Di-Hidrotestosterona/farmacologia , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fase G2/efeitos dos fármacos , Neoplasias da Próstata/patologia , Proteína Quinase CDC2 , Linhagem Celular Tumoral , Quinases Ciclina-Dependentes/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 8 de Crescimento de Fibroblasto/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Fosforilação/efeitos dos fármacos , Proteínas Smad/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Mol Cell Biochem ; 425(1-2): 77-84, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27804049

RESUMO

Fibroblast growth factors (FGFs) comprise a large family of signaling molecules that involve cell patterning, mobilization, differentiation, and proliferation. Various FGFs, including FGF-1, FGF-2, and FGF-5, have been shown to play a role in cytoprotection during adverse cardiac events; however, whether FGF-8 is a cytoprotective remains unclear. The current study was designed to evaluate the effect of FGF-8 treatment on oxidative stress-induced apoptosis in H9c2 cells. Cells were divided into three groups: control, H2O2 (400 µm H2O2), and H2O2 + FGF-8 (4 ng/ml FGF-8). Our results suggest apoptosis was significantly (p < 0.05) enhanced in the H2O2 group relative to control. Moreover, a significant (p < 0.05) decline in apoptosis was observed in the H2O2 + FGF-8 group compared to H2O2-treated cells as evidenced by TUNEL staining, a cell death detection ELISA, and cell viability. Levels of downstream apoptotic mediators, caspase-3 and caspase-9, were significantly (p < 0.05) upregulated following H2O2 treatment but were abrogated following FGF-8 application. Expression levels of Forkhead box protein O1 (FoxO-1), MnSOD, catalase, pAKT, and p-mTOR were significantly (p < 0.05) reduced in the H2O2 group (p < 0.05). Notably, these levels were significantly (p < 0.05) reversed following FGF-8 treatment. Our data, for the first time, suggest FGF-8 is an anti-apoptotic mediator in oxidative-stressed H9c2 cells. Furthermore, our data demonstrate that apoptotic inhibition by FGF-8 is consequent to FoxO-1 oxidative detoxification as well as augmentation to the PI3K/AKT cell survival pathway.


Assuntos
Apoptose/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
8.
Stem Cell Reports ; 7(2): 149-57, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27426041

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons regulate human puberty and reproduction. Modeling their development and function in vitro would be of interest for both basic research and clinical translation. Here, we report a three-step protocol to differentiate human pluripotent stem cells (hPSCs) into GnRH-secreting neurons. Firstly, hPSCs were differentiated to FOXG1, EMX2, and PAX6 expressing anterior neural progenitor cells (NPCs) by dual SMAD inhibition. Secondly, NPCs were treated for 10 days with FGF8, which is a key ligand implicated in GnRH neuron ontogeny, and finally, the cells were matured with Notch inhibitor to bipolar TUJ1-positive neurons that robustly expressed GNRH1 and secreted GnRH decapeptide into the culture medium. The protocol was reproducible both in human embryonic stem cells and induced pluripotent stem cells, and thus provides a translational tool for investigating the mechanisms of human puberty and its disorders.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco Pluripotentes/citologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Humanos , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Nariz/citologia , Células-Tronco Pluripotentes/metabolismo , Receptores Notch/metabolismo , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo
9.
Appl Microbiol Biotechnol ; 100(2): 625-35, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26411459

RESUMO

Human fibroblast growth factor 8b (FGF8b) was expressed based on a baculovirus expression vector system (BEVS) and identified as having a protective effect on Parkinson's disease. Immunoblotting demonstrated that rhFGF8b proteins were recognized by a human anti-FGF8b antibody. The multiplicity of infection and timing of harvest had a significant effect on protein yield and protein quality. Our results indicated that the rhFGF8b was first detectable at 36 h postinfection and reached a maximum at 60 h. A multiplicity of infection (MOI) of 8 pfu/mL was suitable for harvest. The target protein was purified by heparin-affinity chromatography. In vitro methylthiazol tetrazolium (MTT) assays demonstrated that the purified rhFGF8b could significantly stimulate proliferation of NIH3T3 cells. Furthermore, to elucidate the effect of rhFGF8b on Parkinson's disease, we used FGF8b pretreatment on a cell model of Parkinson's disease. The results indicated that rhFGF8b prevented necrosis and apoptosis of 1-METHYL-4-phenyl pyridine (MPP(+)) treated PC12 cells. Moreover, the effect of FGF8b on messenger RNA (mRNA) levels of apoptosis and ERS genes was investigated to clarify the molecular mechanisms of FGF8b. The results suggest that FGF8b exerts neuroprotective effects by alleviating endoplasmic reticulum (ER) stress during PD. These results suggest that FGF8b may be a promising candidate therapeutic drug for neurodegenerative diseases related to ER stress.


Assuntos
Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Baculoviridae/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromatografia de Afinidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fator 8 de Crescimento de Fibroblasto/isolamento & purificação , Humanos , Camundongos , Células NIH 3T3 , Fármacos Neuroprotetores/isolamento & purificação , Células PC12 , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Sais de Tetrazólio , Tiazóis
10.
Peptides ; 73: 88-94, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26409788

RESUMO

BMP induces osteoblast differentiation, whereas a key proinflammatory cytokine, TNF-α, causes inflammatory bone damage shown in rheumatoid arthritis. FGF molecules are known to be involved in bone homeostasis. However, the effects of FGF-8 on osteoblast differentiation and the interaction between FGF-8, BMPs and TNF-α have yet to be clarified. Here we investigated the effects of FGF-8 in relation to TNF-α actions on BMP-2-induced osteoblast marker expression using myoblast cell line C2C12, osteoblast precursor cell line MC3T3-E1 and rat calvarial osteoblasts. It was revealed that FGF-8 inhibited BMP-2-induced expression of osteoblast differentiation markers, including Runx2, osteocalcin, alkaline phosphatase, type-1 collagen and osterix, in a concentration-dependent manner. The inhibitory effects of FGF-8 on BMP-induced osteoblast differentiation and Smad1/5/8 activation were enhanced in the presence of TNF-α action. FGF-8 also inhibited BMP-2-induced expression of Wnt5a, which activates a non-canonical Wnt signaling pathway. FGF-8 had no significant influence on the expression levels of TNF receptors, while FGF-8 suppressed the expression of inhibitory Smad6 and Smad7, suggesting a possible feedback activity through FGF to BMP receptor (BMPR) signaling. Of note, inhibition of ERK activity and FGF receptor (FGFR)-dependent protein kinase, but not JNK or NFκB pathway, suppressed the FGF-8 actions on BMP-induced osteoblast differentiation. FGF-8 was revealed to suppress BMP-induced osteoblast differentiation through the ERK pathway and the effects were enhanced by TNF-α. Given the finding that FGF-8 expression was increased in synovial tissues of rheumatoid arthritis, the functional interaction between FGFR and BMPR signaling may be involved in the development process of inflammatory bone damage.


Assuntos
Antígenos de Diferenciação/biossíntese , Proteína Morfogenética Óssea 2/metabolismo , Fator 8 de Crescimento de Fibroblasto/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Osteoblastos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Fator 8 de Crescimento de Fibroblasto/farmacologia , Humanos , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , NF-kappa B/metabolismo , Osteoblastos/citologia , Ratos , Proteínas Smad/metabolismo
11.
Int J Mol Med ; 36(1): 255-62, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25976705

RESUMO

Spermatogonial stem cells (SSCs) are adult male germ cells that develop after birth. Throughout the lifetime of an organism, SSCs sustain spermatogenesis through self-renewal and produce daughter cells that differentiate into spermatozoa. Several studies have demonstrated that SSCs can acquire pluripotency under appropriate culture conditions, thus becoming multipotent germline stem cells (mGSCs) that express markers of pluripotency in culture and form teratomas following transplantation into immunodeficient mice. In the present study, we generated neural precursor cells expressing CD24, a neural precursor marker, from pluripotent stem cell lines and demonstrated that these cells effectively differentiated along a neural lineage in vitro. In addition, we found that paracrine factors promoted CD24 expression during the neural differentiation of mGSCs. Our results indicated that the expression of CD24, enhanced by a combination of retinoic acid (RA), noggin and fibroblast growth factor 8 (FGF8) under serum-free conditions promoted neural precursor differentiation. Using a simple cell sorting method, we were able to collect neural precursor cells with the potential to differentiate from mGSCs into mature neurons and astrocytes in vitro.


Assuntos
Células-Tronco Adultas/citologia , Antígeno CD24/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Neurogênese/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Animais , Astrócitos/citologia , Proteína Morfogenética Óssea 4/farmacologia , Proteínas de Transporte/farmacologia , Células Cultivadas , Fator 8 de Crescimento de Fibroblasto/farmacologia , Fibroblastos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Proteínas Hedgehog/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Células-Tronco Pluripotentes/metabolismo , Espermatogônias/citologia , Tretinoína/farmacologia
12.
Oncotarget ; 6(2): 935-52, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25473897

RESUMO

Colorectal cancer (CRC) is a major cause of cancer-related death worldwide. The poor prognosis of CRC is mainly due to uncontrolled tumor growth and distant metastases. In this study, we found that the level of FGF8 was elevated in the great majority of CRC cases and high FGF8 expression was significantly correlated with lymph nodes metastasis and worse overall survival. Functional studies showed that FGF8 can induce a more aggressive phenotype displaying epithelial-to-mesenchymal transition (EMT) and enhanced invasion and growth in CRC cells. Consistent with this, FGF8 can also promote tumor growth and metastasis in mouse models. Bioinformatics and pathological analysis suggested that YAP1 is a potential downstream target of FGF8 in CRC cells. Molecular validation demonstrated that FGF8 fully induced nuclear localization of YAP1 and enhanced transcriptional outcomes such as the expression of CTGF and CYR61, while decreasing YAP1 expression impeded FGF-8-induced cell growth, EMT, migration and invasion, revealing that YAP1 is required for FGF8-mediated CRC growth and metastasis. Taken together, these results demonstrate that FGF8 contributes to the proliferative and metastatic capacity of CRC cells and may represent a novel candidate for intervention in tumor growth and metastasis formation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proliferação de Células/genética , Neoplasias Colorretais/genética , Fator 8 de Crescimento de Fibroblasto/genética , Regulação Neoplásica da Expressão Gênica , Fosfoproteínas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Idoso , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/genética , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Fator 8 de Crescimento de Fibroblasto/farmacologia , Células HCT116 , Humanos , Imuno-Histoquímica , Metástase Linfática , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Fosfoproteínas/metabolismo , Prognóstico , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Análise de Sobrevida , Fatores de Transcrição , Transplante Heterólogo , Carga Tumoral/genética , Proteínas de Sinalização YAP
13.
PLoS One ; 9(9): e108241, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25259688

RESUMO

Fibroblast growth factor 8 (FGF8) is a key molecular signal that is necessary for early embryonic development of the central nervous system, quickly disappearing past this point. It is known to be one of the primary morphogenetic signals required for cell fate and survival processes in structures such as the cerebellum, telencephalic and isthmic organizers, while its absence causes severe abnormalities in the nervous system and the embryo usually dies in early stages of development. In this work, we have observed a new possible therapeutic role for this factor in demyelinating disorders, such as leukodystrophy or multiple sclerosis. In vitro, oligodendrocyte progenitor cells were cultured with differentiating medium and in the presence of FGF8. Differentiation and proliferation studies were performed by immunocytochemistry and PCR. Also, migration studies were performed in matrigel cultures, where oligodendrocyte progenitor cells were placed at a certain distance of a FGF8-soaked heparin bead. The results showed that both migration and proliferation was induced by FGF8. Furthermore, a similar effect was observed in an in vivo demyelinating mouse model, where oligodendrocyte progenitor cells were observed migrating towards the FGF8-soaked heparin beads where they were grafted. In conclusion, the results shown here demonstrate that FGF8 is a novel factor to induce oligodendrocyte progenitor cell activation, migration and proliferation in vitro, which can be extrapolated in vivo in demyelinated animal models.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Oligodendroglia/citologia , Animais , Animais Recém-Nascidos , Biomarcadores , Diferenciação Celular/genética , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Doenças Desmielinizantes/terapia , Camundongos , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Transplante de Células-Tronco
14.
J Neurosci Methods ; 227: 100-6, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24583076

RESUMO

BACKGROUND: P19 mouse embryonic carcinoma cells are conventionally induced to differentiate into neural cells by suspension culture in the presence of retinoic acid to form cell aggregates, followed by adhesion culture in a poly-l-lysine-coated dish. Drawbacks of this procedure include it taking more than 10 days to obtain mature neurons, and non-neuronal proliferating cells occupying the majority of the cell population with time. NEW METHOD: Here, we show a novel method for the rapid and efficient neurogenesis of P19 cells, without aggregate formation in a suspension culture. The new approach is based on an adherent serum-free culture in a laminin-coated dish in the presence of FGF8, a γ-secretase inhibitor, and cytosine arabinoside. RESULTS: The new method efficiently induced P19 cells to differentiate into neurons within 4 days, and subsequently into mature neurons that were responsive to several neurotransmitters, giving spontaneous neuronal network activity within 6 days. COMPARISON WITH EXISTING METHOD: The novel method accelerated neuritogenesis and enhanced population of neuron selectively compared to the conventional method. Proliferating non-neuronal cells were eliminated by adding cytosine arabinoside during neuronal maturation. CONCLUSIONS: The method is useful for studying neuronal differentiation or activities.


Assuntos
Diferenciação Celular/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Cálcio/metabolismo , Carcinoma Embrionário/patologia , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Meios de Cultura Livres de Soro/farmacologia , Fator 8 de Crescimento de Fibroblasto/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Laminina/metabolismo , Laminina/farmacologia , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Neurotransmissores/farmacologia , Cloreto de Potássio/farmacologia , Fatores de Tempo
15.
Transl Res ; 162(5): 317-23, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24001408

RESUMO

Recently, using the chemical genetics approach for cell reprogramming, via the combination of small molecule modulators of chromatin modifying enzymes, specific SMAD signaling pathways, and cyclic adenosine monophosphate levels, we have been able to generate neuronallike cells predominantly positive to mature neuronal and dopaminergic markers. This study aimed to characterize further the dopaminergic properties of neurally induced (NI) human bone marrow-derived mesenchymal stem cells (hMSCs) and to determine whether addition of sonic hedgehog (SHH)/fibroblast growth factor 8 (FGF8) to NI medium could promote further dopaminergic maturation. Dopaminergic differentiation was evaluated by immunocytochemistry, reverse transcription-polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay. Results demonstrated that release of dopamine by NI-hMSCs differentiated with SMAD inhibitor supplementation significantly increased from picogram to nanogram levels, with a tendency of further increase when supplemented by SHH/FGF8. Direct generation of dopaminergic cells from adult hMSCs by using this reprogramming approach may have significant implications for understanding the mechanism underlying cell plasticity and may open new potentialities for cell replacement therapies.


Assuntos
AMP Cíclico/metabolismo , Dopamina/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Meios de Cultura , Fator 8 de Crescimento de Fibroblasto/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/farmacologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Smad/metabolismo
16.
Cancer Lett ; 339(2): 226-36, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23774400

RESUMO

Previous studies have demonstrated that fibroblast growth factor 8b (FGF8b) is up-regulated in a large proportion of prostate cancer patients and that it plays a key role in prostate carcinogenesis. In this study, we designed and synthesized a gN helix domain derived short peptide (termed 8b-13) based on the analysis of the FGF8b-FGFR structure. The synthetic peptides inhibited the proliferation of prostate cancer cell lines, including PC-3 and DU-145 cells. Further investigations indicated that 8b-13 arrested the cell cycle at the G0/G1 phase, reduced the activation of the Erk1/2, P38, and Akt cascades, and down-regulated the expression of G1/S-specific cyclinD1. The suppression of DNA synthesis and the G1 to S phase transition due to the expression of proteins related to proliferation and cell cycle progression may contribute to the inhibitory effect of 8b-13 peptides on cellular proliferation. Our results not only suggest that 8b-13 exerts an antitumor effect in prostate cancer but also confirm the essential role of the gN helix domain in mediating the activity of FGF8b.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Peptídeos/farmacologia , Neoplasias da Próstata/metabolismo , Domínios e Motivos de Interação entre Proteínas , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Sequência de Aminoácidos , Carboxilesterase/genética , Carboxilesterase/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Fator 8 de Crescimento de Fibroblasto/química , Fator 8 de Crescimento de Fibroblasto/farmacologia , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Peptídeos/química , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Neoplasias da Próstata/genética , Proteômica/métodos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos
17.
Mol Cell Endocrinol ; 358(1): 104-15, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22465097

RESUMO

Fibroblast growth factor-8 (FGF-8) is implicated in the development and progression of breast cancer and its levels are frequently elevated in breast tumors. The mechanisms driving FGF-8-mediated tumorigenesis are not well understood. Herein we aimed to identify target genes associated with FGF-8b-mediated breast cancer cell proliferation by carrying out a cDNA microarray analysis of genes expressed in estrogen receptor negative S115 breast cancer cells treated with FGF-8b for various time periods in comparison with those expressed in non-treated cells. Gene and protein expression was validated for selected genes by qPCR and western blotting respectively. Furthermore, using TRANSBIG data, the expression of human orthologs of FGF-8-regulated genes was correlated to the Nottingham prognostic index and estrogen receptor status. The analysis revealed a number of significantly up- and down-regulated genes in response to FGF-8b at all treatment times. The most differentially expressed genes were genes related to cell cycle regulation, mitosis, cancer, and cell death. Several key regulators of early cell cycle progression such as Btg2 and cyclin D1, as well as regulators of mitosis, including cyclin B, Plk1, survivin, and aurora kinase A, were identified as novel targets for FGF-8b, some of which were additionally shown to correlate with prognosis and ER status in human breast cancer. The results suggest that in stimulation of proliferation FGF-8b not only promotes cell cycle progression through the G1 restriction point but also regulates key proteins involved in chromosomal segregation during mitosis and cytokinesis of breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Ciclo Celular/genética , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Neoplasias Mamárias Animais/genética , Mitose/genética , Animais , Apoptose/genética , Aurora Quinase A , Aurora Quinases , Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , Ciclina B/genética , Ciclina B/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Feminino , Fator 8 de Crescimento de Fibroblasto/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Mamárias Animais/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de Estrogênio/biossíntese , Transdução de Sinais , Survivina , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Quinase 1 Polo-Like
18.
Neurosci Lett ; 506(1): 22-7, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22037229

RESUMO

Amniotic epithelial cells (AECs) have been the subject of interest due to their stem cells characteristics. Pluripotency, lack of teratoma formation, low immunogenicity and no ethical issues make them an alternative source of stem cells for generation of dopaminergic neurons in vitro. AECs may differentiate into midbrain dopaminergic neurons in the presence of induction factors which trigger pathway naturally occurring in the mesencephalon during embryogenesis. In this study, we examined whether FGF8 and Sonic hedgehog (Shh) can induce AECs to express permanently midbrain dopaminergic neurons traits. We found that fresh isolated AECs temporarily expressed some neural markers which decreased 7 days after culture. Co-treatment of cells with FGF8 and Shh had synergistic effect on expression of TH, DAT, Pax2, Lmx1b and En1. Shh ceased differentiation in dopaminergic state and decreased DßH. HPLC analysis revealed that dopamine was released into the medium upon depolarization, indicating that functional dopaminergic neurons were present in the culture. These results demonstrate that AECs have capability to differentiate into dopaminergic neurons and can be a candidate for cell replacement therapy of Parkinson's disease.


Assuntos
Diferenciação Celular/fisiologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Células Epiteliais/fisiologia , Placenta/citologia , Análise de Variância , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Dopamina beta-Hidroxilase/metabolismo , Células Epiteliais/efeitos dos fármacos , Feminino , Fator 8 de Crescimento de Fibroblasto/farmacologia , Proteínas Hedgehog/farmacologia , Humanos , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Fatores de Transcrição/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
19.
Mol Cancer Ther ; 11(2): 267-76, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22135230

RESUMO

Signaling from the human hematopoietic stem cell (HSC) niche formed by osteoblastic cells regulates hematopoiesis. We previously found that retinoic acid receptor alpha (RARα), a transcription factor activated by retinoic acid (RA), mediates both granulocytic and osteoblastic differentiation. This effect depends on decreased phosphorylation of serine 77 of RARα (RARαS77) by the cyclin-dependent kinase-activating kinase (CAK) complex, a key cell-cycle regulator. In this article, we report that, by suppressing CAK phosphorylation of RARα, RA induces FGF8f to mediate osteosarcoma U2OS cell differentiation in an autocrine manner. By contrast, paracrine FGF8f secreted into osteoblast-conditioned medium by U2OS cells transduced with FGF8f or a phosphorylation-defective RARαS77 mutant, RARαS77A, bypasses RA stimuli to cross-mediate granulocytic differentiation of different types of human leukemic myeloblasts and normal primitive hematopoietic CD34(+) cells, possibly through modulating mitogen-activated protein kinase (MAPK) pathways. Further experiments using recombinant human FGF8f (rFGF8f) stimuli, antibody neutralization, and peptide blocking showed that paracrine FGF8f is required for mediating terminal leukemic myeloblast differentiation. These studies indicate a novel regulatory mechanism of granulocytic differentiation instigated by RA from the HSC niche, which links loss of CAK phosphorylation of RARα with paracrine FGF8f-mediated MAPK signaling to mediate leukemic myeloblast differentiation in the absence of RA. Therefore, these findings provide a compelling molecular rationale for further investigation of paracrine FGF8f regulation, with the intent of devising HSC niche-based FGF8f therapeutics for myeloid leukemia, with or without RA-resistance.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/metabolismo , Osteoblastos/efeitos dos fármacos , Retinoides/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Quinases Ciclina-Dependentes/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/farmacologia , Granulócitos/efeitos dos fármacos , Granulócitos/metabolismo , Granulócitos/patologia , Células HL-60 , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Osteoblastos/metabolismo , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Nicho de Células-Tronco/efeitos dos fármacos , Quinase Ativadora de Quinase Dependente de Ciclina
20.
Mol Cell Endocrinol ; 343(1-2): 7-17, 2011 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-21664418

RESUMO

Estrogen is involved in the development and progression of breast cancer. Here we investigated the effect of fibroblast growth factor (FGF)-8 on breast cancer cell proliferation caused by estrogen using human breast cancer MCF-7 cells. MCF-7 cells express estrogen receptor (ER)α, ERß, FGF receptors, and Smad signaling molecules. Estradiol stimulated MCF-7 cell proliferation in a concentration-responsive manner, whereas BSA-bound estradiol had a weak effect on MCF-7 cell mitosis compared with the effect of free estradiol. It is notable that estrogen-induced cell proliferation was enhanced in the presence of FGF-8 and that the combined effects were reversed in the presence of an FGF-receptor kinase inhibitor or an ER antagonist. It was also revealed that FGF-8 increased the expression levels of ERα, ERß and aromatase mRNAs, while estradiol reduced the expression levels of ERs, aromatase and steroid sulfatase in MCF-7 cells. FGF-8-induced phosphorylation of FGF receptors was augmented by estradiol, which was reversed by an ER antagonist. FGF-8-induced activation of MAPKs and AKT signaling was also upregulated in the presence of estrogen. On the other hand, FGF-8 suppressed BMP-7 actions that are linked to mitotic inhibition by activating the cell cycle regulator cdc2. FGF-8 was revealed to inhibit BMP receptor actions including Id-1 promoter activity and Smad1/5/8 phosphorylation by suppressing expression of BMP type-II receptors and by increasing expression of inhibitory Smads. Collectively, the results indicate that FGF-8 acts to facilitate cell proliferation by upregulating endogenous estrogenic actions as well as by suppressing BMP receptor signaling in ER-expressing breast cancer cells.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células , Fator 8 de Crescimento de Fibroblasto/farmacologia , Receptores de Estrogênio/metabolismo , Aromatase/genética , Aromatase/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Neoplasias da Mama/genética , Estradiol/farmacologia , Feminino , Humanos , Receptores de Estrogênio/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Esteril-Sulfatase/genética , Esteril-Sulfatase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA