Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 12(7): e0180579, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28704421

RESUMO

The complex architecture of adult brain derives from tightly regulated migration and differentiation of precursor cells generated during embryonic neurogenesis. Changes at transcriptional level of genes that regulate migration and differentiation may lead to neurodevelopmental disorders. Androgen receptor (AR) is a transcription factor that is already expressed during early embryonic days. However, AR role in the regulation of gene expression at early embryonic stage is yet to be determinate. Long non-coding RNA (lncRNA) Sox2 overlapping transcript (Sox2OT) plays a crucial role in gene expression control during development but its transcriptional regulation is still to be clearly defined. Here, using Bicalutamide in order to pharmacologically inactivated AR, we investigated whether AR participates in the regulation of the transcription of the lncRNASox2OTat early embryonic stage. We identified a new DNA binding region upstream of Sox2 locus containing three androgen response elements (ARE), and found that AR binds such a sequence in embryonic neural stem cells and in mouse embryonic brain. Our data suggest that through this binding, AR can promote the RNA polymerase II dependent transcription of Sox2OT. Our findings also suggest that AR participates in embryonic neurogenesis through transcriptional control of the long non-coding RNA Sox2OT.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neurogênese , RNA Longo não Codificante/genética , Receptores Androgênicos/metabolismo , Ativação Transcricional , Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Nitrilas/farmacologia , RNA Longo não Codificante/metabolismo , Elementos de Resposta , Compostos de Tosil/farmacologia
3.
Neurorehabil Neural Repair ; 30(5): 461-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26359345

RESUMO

Background Sleep evaluation is increasingly being used as prognostic tool in patients with disorders of consciousness, but, surprisingly, the role of Period3 (Per3) gene polymorphism has never been evaluated. Objective The aim of this study was to investigate the contribution of Per3 genotype on sleep quantity and consciousness recovery level in patients with disorders of consciousness (DOC). Methods In this observational study, we evaluated 71 patients with DOC classified as vegetative state/unresponsive wakefulness syndrome or minimally conscious state. Demographic and clinical data were collected and a standardised diagnostic workup, including a polysomnographic record, was applied. After informed consent provided by proxy, genomic DNA was obtained and Per3 polymorphism was analysed by polymerase chain reaction to identify 5/5, 4/5, or 4/4 genotype. Results Per3(5/5) genotype was found in 12.7% of our DOC patients. The median total Coma Recovery Scale-revised score in Per3(5/5) carriers was significantly higher than 4/4 genotype (10, range 5-16 vs 7, range 4-11; post hoc P = .036). Moreover, total sleep time seemed to be higher in 5/5 genotype (5/5, 221 minutes, range 88-515 minutes; 4/4, 151.5 minutes, range 36-477 minutes; and 4/5, 188 minutes, range 44-422 minutes). Conclusion For the first time we have shown a possible association between Per3 polymorphism and consciousness recovery level in DOC patients. Even though the exact molecular mechanism has not been defined, we speculate that its effect is mediated by higher total sleep time and slow wave sleep, which would improve the preservation of main cerebral connections.


Assuntos
Transtornos da Consciência/genética , Transtornos da Consciência/fisiopatologia , Proteínas Circadianas Period/genética , Polimorfismo de Nucleotídeo Único/genética , Recuperação de Função Fisiológica/genética , Sono/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Distribuição de Qui-Quadrado , Eletroencefalografia , Potenciais Evocados/genética , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Repetições Minissatélites/genética , Polissonografia , Estudos Retrospectivos , Adulto Jovem
4.
PLoS One ; 9(11): e112746, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25392924

RESUMO

Spinal and bulbar muscular atrophy (SBMA) or Kennedy's disease is an X-linked CAG/polyglutamine expansion motoneuron disease, in which an elongated polyglutamine tract (polyQ) in the N-terminal androgen receptor (ARpolyQ) confers toxicity to this protein. Typical markers of SBMA disease are ARpolyQ intranuclear inclusions. These are generated after the ARpolyQ binds to its endogenous ligands, which promotes AR release from chaperones, activation and nuclear translocation, but also cell toxicity. The SBMA mouse models developed so far, and used in preclinical studies, all contain an expanded CAG repeat significantly longer than that of SBMA patients. Here, we propose the use of SBMA patients adipose-derived mesenchymal stem cells (MSCs) as a new human in vitro model to study ARpolyQ toxicity. These cells have the advantage to express only ARpolyQ, and not the wild type AR allele. Therefore, we isolated and characterized adipose-derived MSCs from three SBMA patients (ADSC from Kennedy's patients, ADSCK) and three control volunteers (ADSCs). We found that both ADSCs and ADSCKs express mesenchymal antigens, even if only ADSCs can differentiate into the three typical cell lineages (adipocytes, chondrocytes and osteocytes), whereas ADSCKs, from SBMA patients, showed a lower growth potential and differentiated only into adipocyte. Moreover, analysing AR expression on our mesenchymal cultures we found lower levels in all ADSCKs than ADSCs, possibly related to negative pressures exerted by toxic ARpolyQ in ADSCKs. In addition, with proteasome inhibition the ARpolyQ levels increased specifically in ADSCKs, inducing the formation of HSP70 and ubiquitin positive nuclear ARpolyQ inclusions. Considering all of this evidence, SBMA patients adipose-derived MSCs cultures should be considered an innovative in vitro human model to understand the molecular mechanisms of ARpolyQ toxicity and to test novel therapeutic approaches in SBMA.


Assuntos
Adipócitos/patologia , Tecido Adiposo/patologia , Células-Tronco Mesenquimais/patologia , Modelos Biológicos , Transtornos Musculares Atróficos/patologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Idoso , Estudos de Casos e Controles , Diferenciação Celular , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Cultura Primária de Células , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo
5.
Stem Cell Res Ther ; 5(3): 63, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24887516

RESUMO

INTRODUCTION: Human adipose-derived stromal cells (hASCs), due to their relative feasibility of isolation and ability to secrete large amounts of angiogenic factors, are being evaluated for regenerative medicine. However, their limited culture life span may represent an obstacle for both preclinical investigation and therapeutic use. To overcome this problem, hASCs immortalization was performed in order to obtain cells with in vitro prolonged life span but still maintain their mesenchymal marker expression and ability to secrete angiogenic factors. METHODS: hASCs were transduced with the human telomerase reverse transcriptase (hTERT) gene alone or in combination with either SV-40 or HPV E6/E7 genes. Mesenchymal marker expression on immortalized hASCs lines was confirmed by flow cytometry (FC), differentiation potential was evaluated by immunocytochemistry and ELISA kits were used for evaluation of angiogenic factors. Green fluorescent protein (GFP) gene transduction was used to obtain fluorescent cells. RESULTS: We found that hTERT alone failed to immortalize hASCs (hASCs-T), while hTERT/SV40 (hASCs-TS) or hTERT/HPV E6/E7 (hASCs-TE) co-transductions successfully immortalized cells. Both hASCs-TS and hASCs-TE were cultured for up to one year with a population doubling level (PDL) up to 100. Comparative studies between parental not transduced (hASCs-M) and immortalized cell lines showed that both hASCs-TS and hASCs-TE maintained a mesenchymal phenotypic profile, whereas differentiation properties were reduced particularly in hASCs-TS. Interestingly, hASCs-TS and hASCs-TE showed a capability to secrete significant amount of HGF and VEGF. Furthermore, hASCs-TS and hASCs-TE did not show tumorigenic properties in vitro although some chromosomal aberrations were detected. Finally, hASCs-TS and hASCs-TE lines were stably fluorescent upon transduction with the GFP gene. CONCLUSIONS: Here we demonstrated, for the first time, that hASCs, upon immortalization, maintain a strong capacity to secrete potent angiogenic molecules. By combining hASCs immortalization and their paracrine characteristics, we have developed a "hybridoma-like model" of hASCs that could have potential applications for discovering and producing molecules to use in regenerative medicine (process scale-up).


Assuntos
Tecido Adiposo/citologia , Técnicas de Cultura de Células/métodos , Linhagem Celular/citologia , Linhagem Celular/metabolismo , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/metabolismo , Diferenciação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fator de Crescimento de Hepatócito/biossíntese , Humanos , Imuno-Histoquímica , Imunofenotipagem , Células-Tronco Mesenquimais/metabolismo , Telomerase/genética , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/biossíntese
6.
Stem Cell Res Ther ; 5(1): 7, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24423450

RESUMO

INTRODUCTION: Silk fibroin (SF) scaffolds have been shown to be a suitable substrate for tissue engineering and to improve tissue regeneration when cellularized with mesenchymal stromal cells (MSCs). We here demonstrate, for the first time, that electrospun nanofibrous SF patches cellularized with human adipose-derived MSCs (Ad-MSCs-SF), or decellularized (D-Ad-MSCs-SF), are effective in the treatment of skin wounds, improving skin regeneration in db/db diabetic mice. METHODS: The conformational and structural analyses of SF and D-Ad-MSCs-SF patches were performed by scanning electron microscopy, confocal microscopy, Fourier transform infrared spectroscopy and differential scanning calorimetry. Wounds were performed by a 5 mm punch biopsy tool on the mouse's back. Ad-MSCs-SF and D-Ad-MSCs-SF patches were transplanted and the efficacy of treatments was assessed by measuring the wound closure area, by histological examination and by gene expression profile. We further investigated the in vitro angiogenic properties of Ad-MSCs-SF and D-Ad-MSCs-SF patches by affecting migration of human umbilical vein endothelial cells (HUVECs), keratinocytes (KCs) and dermal fibroblasts (DFs), through the aortic ring assay and, finally, by evaluating the release of angiogenic factors. RESULTS: We found that Ad-MSCs adhere and grow on SF, maintaining their phenotypic mesenchymal profile and differentiation capacity. Conformational and structural analyses on SF and D-Ad-MSCs-SF samples, showed that sterilization, decellularization, freezing and storing did not affect the SF structure. When grafted in wounds of diabetic mice, both Ad-MSCs-SF and D-Ad-MSCs-SF significantly improved tissue regeneration, reducing the wound area respectively by 40% and 35%, within three days, completing the process in around 10 days compared to 15-17 days of controls. RT2 gene profile analysis of the wounds treated with Ad-MSCs-SF and D-Ad-MSCs-SF showed an increment of genes involved in angiogenesis and matrix remodeling. Finally, Ad-MSCs-SF and D-Ad-MSCs-SF co-cultured with HUVECs, DFs and KCs, preferentially enhanced the HUVECs' migration and the release of angiogenic factors stimulating microvessel outgrowth in the aortic ring assay. CONCLUSIONS: Our results highlight for the first time that D-Ad-MSCs-SF patches are almost as effective as Ad-MSCs-SF patches in the treatment of diabetic wounds, acting through a complex mechanism that involves stimulation of angiogenesis. Our data suggest a potential use of D-Ad-MSCs-SF patches in chronic diabetic ulcers in humans.


Assuntos
Fibroínas/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Reepitelização , Alicerces Teciduais/química , Tecido Adiposo/citologia , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Fibroblastos/fisiologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Queratinócitos/fisiologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Obesos , Neovascularização Fisiológica , Ratos , Ratos Sprague-Dawley , Receptores para Leptina/genética
7.
Nat Neurosci ; 12(10): 1248-56, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19734891

RESUMO

Neural stem cells (NSCs) are controlled by diffusible factors. The transcription factor Sox2 is expressed by NSCs and Sox2 mutations in humans cause defects in the brain and, in particular, in the hippocampus. We deleted Sox2 in the mouse embryonic brain. At birth, the mice showed minor brain defects; shortly afterwards, however, NSCs and neurogenesis were completely lost in the hippocampus, leading to dentate gyrus hypoplasia. Deletion of Sox2 in adult mice also caused hippocampal neurogenesis loss. The hippocampal developmental defect resembles that caused by late sonic hedgehog (Shh) loss. In mutant mice, Shh and Wnt3a were absent from the hippocampal primordium. A SHH pharmacological agonist partially rescued the hippocampal defect. Chromatin immunoprecipitation identified Shh as a Sox2 target. Sox2-deleted NSCs did not express Shh in vitro and were rapidly lost. Their replication was partially rescued by the addition of SHH and was almost fully rescued by conditioned medium from normal cells. Thus, NSCs control their status, at least partly, through Sox2-dependent autocrine mechanisms.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog/fisiologia , Hipocampo , Neurônios/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular , Células Cultivadas , Imunoprecipitação da Cromatina/métodos , Meios de Cultivo Condicionados/farmacologia , DNA Nucleotidiltransferases/genética , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida , Proteínas de Fluorescência Verde/genética , Proteínas Hedgehog/antagonistas & inibidores , Hipocampo/citologia , Hipocampo/embriologia , Hipocampo/crescimento & desenvolvimento , Marcação In Situ das Extremidades Cortadas/métodos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas de Filamentos Intermediários/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurogênese/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXB1/deficiência , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt3 , Proteína Wnt3A
8.
Reprod Fertil Dev ; 19(1): 35-42, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17389133

RESUMO

A gradual transition from oocyte-derived mRNA and proteins to full embryonic transcription characterises early embryonic development. Messenger RNAs and proteins of maternal origin are accumulated into the oocyte throughout its growth inthe ovary. Upon fertilisation, sev eral mechanisms ar e activated that controlthe appropriate use of such material and prepare for the synthesis of new products. The present review will describe some of the mechanisms active in early embryos of domestic species. Data will be presented on the control of gene expression by the 3' untranslated regions and their interaction with specialised sequences at the 5' cap end. The process of RNA sorting and localisation, initially described in different cell types and in oocytes of lower species, will also be discussed, particularly in relation to its possible role in regulating early pig development. Finally, specific genes involved in the activation of cattle embryonic transcription will be described. This brief overview will provide some suggestions on how these different mechanisms may be integrated and cooperate to ensure the correct initiation of embryonic development.


Assuntos
Animais Domésticos/embriologia , Animais Domésticos/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Regiões 3' não Traduzidas , Animais , Feminino , Oócitos/fisiologia , Gravidez , Transporte de RNA
9.
Theriogenology ; 67(1): 54-63, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17055567

RESUMO

Embryonic stem cells (ESCs) hold great promise for therapeutic use and represent a unique tool for investigating the process of self-renewal and differentiation. The properties that make ESCs unique are their capacity of unlimited self-renewal coupled with the property of re-entering the developmental process if returned inside a blastocyst. Such plasticity enable ESCs to form all embryonic tissues including germ cells. However, these remarkable properties, at present, have been demonstrated only for mouse ESCs even if cells with somehow more limited capacities have been derived in many different species including humans. The isolation of pluripotent embryonic cells lines from human embryos marked a crucial change of perspective in evaluating the properties defining an embryonic stem cell lines moving the focus from the generation of a germ-line chimera, obviously not feasible nor desirable in human, to the capacity of these cells to differentiate both in vivo and in vitro in fully mature and functional cell types of all kinds. Therefore, ESCs properties in species different from the mouse are being reassessed and re-evaluated, in view of their potential use as experimental models for the development of clinical applications. Among the species that may play a useful role in this field, the pig has a long-standing history as a prime animal model for pre-clinical biomedical applications and therefore, pig ESCs are attracting renewed interest. In this review, we will summarize the current knowledge on this topic and will contrast the relatively limited data available in this species with the much larger wealth of information available for mouse and human ESCs, in an attempt to assess whether or not pig ESCs can actually become a useful tool in the fast growing field of cell therapy.


Assuntos
Embrião de Mamíferos/citologia , Células-Tronco/citologia , Suínos/embriologia , Animais , Diferenciação Celular , Transplante de Células , Embrião de Mamíferos/fisiologia , Humanos , Masculino , Camundongos , Modelos Animais , Especificidade da Espécie , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...