Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Neurochem Res ; 49(9): 2364-2378, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38837091

RESUMO

As an element of the cellular signaling systems, extracellular vesicles (EVs) exhibit many desirable traits for usage as targeted delivery vehicles. When administered, EVs cause little to no toxic or immune response, stay in circulation for longer periods compared to synthetic carriers, preferentially accumulate in tissues that are the same or similar to their cell-of-origin and can pass through the blood-brain barrier. Combined, these traits make neural EVs a particularly promising tool for delivering drugs to the brain. This study aims to combine tissue and EVs engineering to prepare neural differentiated cells derived EVs that exhibit neural properties, to develop an effective, tissue-homing drug and gene delivery platform for the brain. Early neural differentiated cell-derived EVs were produced with neural characteristics from neural differentiated human neonatal dermal fibroblasts. The EVs carried key neural proteins such as Nestin, Sox2 and Doublecortin. The cellular uptake of early neural differentiated cell-derived EVs was higher compared to non-neural EVs during in vitro uptake assays on neuroblastoma cells. Moreover, eND-EVs were significantly decreased the viability of neuroblastoma cells. In conclusion, this study revealed that early neural differentiated cell-derived EVs have potential as a promising drug carrier for the treatment of various neural disorders.


Assuntos
Vesículas Extracelulares , Células-Tronco Neurais , Neuroblastoma , Vesículas Extracelulares/metabolismo , Humanos , Células-Tronco Neurais/metabolismo , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Fibroblastos/metabolismo , Fatores de Transcrição SOXB1
2.
Cancer Sci ; 111(1): 175-185, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31715070

RESUMO

Neurogenic differentiation factor 1 (NeuroD1) is a transcription factor critical for promoting neuronal differentiation and maturation. NeuroD1 is involved in neuroblastoma and medulloblastoma; however, its molecular mechanism in promoting tumorigenesis remains unclear. Furthermore, the role of NeuroD1 in non-neural malignancies has not been widely characterized. Here, we found that NeuroD1 is highly expressed in colorectal cancer. NeuroD1-silencing induces the expression of p21, a master regulator of the cell cycle, leading to G2 -M phase arrest and suppression of colorectal cancer cell proliferation as well as colony formation potential. Moreover, NeuroD1-mediated regulation of p21 expression occurs in a p53-dependent manner. Through chromatin immunoprecipitation and point mutation analysis in the predicted NeuroD1 binding site of the p53 promoter, we found that NeuroD1 directly binds to the p53 promoter and suppresses its transcription, resulting in increased p53 expression in NeuroD1-silenced colorectal cancer cells. Finally, xenograft experiments demonstrated that NeuroD1-silencing suppresses colorectal cancer cell tumorigenesis potential by modulating p53 expression. These findings reveal NeuroD1 as a novel regulator of the p53/p21 axis, underscoring its importance in promoting non-neural malignancies. Furthermore, this study provides insight into the transcriptional regulation of p53.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinogênese/genética , Neoplasias Colorretais/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteína Supressora de Tumor p53/genética , Carcinogênese/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica/genética , Células HCT116 , Humanos , Neuroblastoma/genética , Neuroblastoma/patologia , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética
3.
Acta Biochim Biophys Sin (Shanghai) ; 52(9): 1016-1029, 2020 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-32845287

RESUMO

Recent studies have reported an increasing incidence of ischemic stroke, particularly in younger age groups. Dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) are the most common stem cells acquired from the teeth of adults, even elderly people. However, there are no detailed reports on whether DPSCs or PDLSCs are suitable for the treatment of ischemic stroke. In this study, the in vitro differentiation of DPSCs and PDLSCs into neuron-like cells was evaluated. Then, we established a rat model of cerebral ischemia. DPSCs or PDLSCs were administered to animals, and the therapeutic effects of these two types of cells were investigated. The results showed that PDLSCs had a higher differentiation rate than DPSCs. Immunofluorescence studies showed that the expression of the neuronal differentiation marker Thy-1 was higher in PDLSCs than in DPSCs, and other gene markers of neuronal differentiation showed corresponding trends, which were confirmed by western blot analysis. In this process, the Notch and Wnt signaling pathways were inhibited and activated, respectively. Finally, rats with transient occlusion of the right middle cerebral artery were used as a model to assess the therapeutic effect of PDLSCs and DPSCs on ischemia. The results showed that rats in the PDLSC-treated group emitted significantly greater red fluorescence signal than the DPSC-treated group. PDLSC transplantation promoted the recovery of neurological function more effectively than DPSC transplantation. Hence, PDLSCs represent an autogenous source of adult mesenchymal stem cells with desirable biological properties and may be an ideal candidate for clinical applications.


Assuntos
Isquemia Encefálica , Diferenciação Celular , Polpa Dentária/metabolismo , Neurônios/metabolismo , Ligamento Periodontal/metabolismo , Transplante de Células-Tronco , Células-Tronco/metabolismo , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/terapia , Polpa Dentária/patologia , Modelos Animais de Doenças , Xenoenxertos , Humanos , Masculino , Neurônios/patologia , Ligamento Periodontal/patologia , Ratos , Ratos Sprague-Dawley , Células-Tronco/patologia
4.
J Oral Rehabil ; 47 Suppl 1: 55-65, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31291686

RESUMO

OBJECTIVES: In dental tissue engineering, niche is important for maintaining stem cell function and regenerating the dental tissues. However, there is limited knowledge for the growth factors in niche to maintain the function of stem cells. In this study, we investigated the effect of IGF2, a growth factor in stem cells from apical papilla (SCAPs) niche, on differentiation and proliferation potentials of SCAPs. MATERIALS AND METHODS: Recombinant human IGF2 protein (rhIGF2) was used. Cell counting kit-8 assay, Carboxyfluorescein succinimidyl ester assay, alkaline phosphatase (ALP) activity, Alizarin Red staining, quantitative calcium analysis, immunofluorescence staining and real-time RT-PCR were performed to investigate the cell proliferation and differentiation potentials of SCAPs. And proteomic analysis was used to identify the differential secreted proteins. RESULTS: By ALP activity assay, we found that 5 ng/mL rhIGF2 might be the optimal concentration for treatment. Then, Alizarin Red staining, quantitative calcium analysis and osteogenesis-related gene expression results showed that 5 ng/mL rhIGF2 could enhance the osteo-/dentinogenic differentiation potentials in SCAPs. Immunofluorescence staining and real-time RT-PCR results showed that neurogenic markers were significantly induced by 5 ng/mL rhIGF2 in SCAPs. Then, CCK-8 assay and CFSE assay results showed that 5 ng/mL rhIGF2 could enhance the cell proliferation in SCAPs. Furthermore, proteomic analysis showed that IGF2 could induce some secreted proteins which function related to the osteogenesis, neurogenesis and cell proliferation. CONCLUSIONS: Our results identified that IGF2 might be the potential mediator in niche to promote SCAP function and dental tissue regeneration.


Assuntos
Papila Dentária , Proteômica , Diferenciação Celular , Células Cultivadas , Humanos , Fator de Crescimento Insulin-Like II , Neurogênese , Células-Tronco
5.
Molecules ; 25(3)2020 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-32019204

RESUMO

It was shown that AEDG peptide (Ala-Glu-Asp-Gly, Epitalon) regulates the function of the pineal gland, the retina, and the brain. AEDG peptide increases longevity in animals and decreases experimental cancerogenesis. AEDG peptide induces neuronal cell differentiation in retinal and human periodontal ligament stem cells. The aim of the study was to investigate the influence of AEDG peptide on neurogenic differentiation gene expression and protein synthesis in human gingival mesenchymal stem cells, and to suggest the basis for the epigenetic mechanism of this process. AEDG peptide increased the synthesis of neurogenic differentiation markers: Nestin, GAP43, ß Tubulin III, Doublecortin in hGMSCs. AEDG peptide increased Nestin, GAP43, ß Tubulin III and Doublecortin mRNA expression by 1.6-1.8 times in hGMSCs. Molecular modelling method showed, that AEDG peptide preferably binds with H1/6 and H1/3 histones in His-Pro-Ser-Tyr-Met-Ala-His-Pro-Ala-Arg-Lys and Tyr-Arg-Lys-Thr-Gln sites, which interact with DNA. These results correspond to previous experimental data. AEDG peptide and histones H1/3, H1/6 binding may be one of the mechanisms which provides an increase of Nestin, GAP43, ß Tubulin III, and Doublecortin neuronal differentiation gene transcription. AEDG peptide can epigenetically regulate neuronal differentiation gene expression and protein synthesis in human stem cells.


Assuntos
Epigênese Genética , Gengiva/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neurogênese , Neurônios/metabolismo , Oligopeptídeos/farmacologia , Biossíntese de Proteínas , Regulação da Expressão Gênica , Gengiva/citologia , Gengiva/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos
6.
Dev Growth Differ ; 61(9): 457-465, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31599466

RESUMO

Dental stem cells for dental pulp regeneration have become a new strategy for pulpitis treatment. Angiogenesis and neurogenesis play a vital role in the pulp-dentin complex regeneration, and appropriate growth factors will promote the process of angiogenesis and neurogenesis. Insulin-like growth factor-binding protein 5 (IGFBP5) is involved in the regulation of tooth growth and development. A previous study showed that IGFBP5 enhanced osteo/odontogenic differentiation of dental stem cells. Our research intends to reveal the function of IGFBP5 in the angiogenic and neurogenic differentiation of human dental stem cells. Human dental pulp stem cells (DPSCs) were used in the present study. Lentiviral IGFBP5 shRNA was used to silence the IGFBP5. Retroviruses expressing Wild-type IGFBP5 were used to over-express IGFBP5. Angiogenic and neurogenic differentiation were carried out by in vitro study. Real-time RT-PCR and western blot results showed that over-expression of IGFBP5 upregulated the expressions of angiogenic markers, including VEGF, PDGFA and ANG-1, and neurogenic markers, including NCAM, TH, Nestin, ßIII-tubulin, and TH, in DPSCs. Moreover, microscope observation confirmed that over-expression of IGFBP5 enhanced neurosphere formation in DPSCs in size and amount. Immunofluorescence staining results showed that over-expression of IGFBP5 also prompted the percentage of Nestin and ßIII-tubulin positive neurospheres in DPSCs. While depletion of IGFBP5 downregulated the expressions of VEGF, PDGFA, ANG-1, NCAM, TH, Nestin, ßIII-tubulin, and TH, it decreased the neurosphere formation and percentage of Nestin and ßIII-tubulin positive neurospheres in DPSCs. In conclusion, our results revealed that IGFBP5 promoted angiogenic and neurogenic differentiation potential of DPSCs in vitro and provided the possible potential target for enhancing directed differentiation of dental stem cells and dental pulp-dentin functional regeneration.


Assuntos
Polpa Dentária/metabolismo , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Neovascularização Fisiológica , Células-Tronco/metabolismo , Diferenciação Celular , Células Cultivadas , Polpa Dentária/citologia , Humanos , Células-Tronco/citologia
7.
Cell Physiol Biochem ; 50(6): 2097-2107, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30415244

RESUMO

BACKGROUND/AIMS: MiRNAs may regulate neurogenic differentiation of adipose-derived stem cells (ADSCs). In this study, we hypothesized that the miR-142-5p can repress the expression of RhoA/ROCK1 pathway on the neurogenesis of ADSCs. METHODS: Deregulated miRNA during neurogenic differentiation of ADSCs were identified. The expression of neuron-specific enolase (NSE) and ß III tubulin (Neuron-specific class III beta-tubulin) were detected as the markers of neurogenic differentiation by immunostaining and western blot. The targeting of miR-142-5p on RhoA and ROCK1 was verified by dual luciferase assay, qRT-PCR and western blot. The roles of miR-142-5p and the RhoA/ROCK1 signaling pathway were explored by using functional experiments including cell viability and colony formation assays. RESULTS: MiR-142-5p is significantly upregulated during neurogenic differentiation of ADSCs. Knockdown of endogenous miR-142-5p hampered neurogenic differentiation. MiR-142-5p could directly target RhoA and ROCK1 mRNA and repress their expressions, through which it increased the proportion of differentiated cells with positive NSE and ß III tubulin. RhoA/ROCK1 signaling pathway is involved in miR-142-5p effect on the process of neurogenic differentiation of ADSCs. CONCLUSION: Our results demonstrate that miR-142-5p functions as a growth promotive miRNA and plays an important role in neurogenic differentiation by targeting RhoA/ROCK1 in ADSCs.


Assuntos
Diferenciação Celular , MicroRNAs/metabolismo , Neurônios/citologia , Tecido Adiposo/citologia , Antagomirs/metabolismo , Linhagem Celular , Proliferação de Células , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Neurônios/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Tubulina (Proteína)/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Cell Biol Int ; 41(5): 534-543, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28244619

RESUMO

Dental tissue-derived mesenchymal stem cells (MSCs) are easily obtained and considered as a favorable cell source for tissue engineering, but the regulation of direct differentiation is unknown, which restricts their application. The present study investigated the effect of SFRP2, a Wnt signaling modulator, on MSC differentiation using stem cells from apical papilla (SCAPs). The cells were cultured in specific inducing medium for adipogenic, neurogenic, or chondrogenic differentiation. Over-expression of SFRP2 via retroviral infection enhanced the adipogenic and neurogenic differentiation of SCAPs. While inhibit of Wnt pathway by IWR1-endo could enhance the neurogenic differentiation potentials of SCAPs, similar with the function of SFRP2. In addition, over-expression of SFRP2 up-regulated the expression of stemness-related genes SOX2 and OCT4. Furthermore, SOX2 and OCT4 expression was significantly inhibited after lentiviral silencing of SFRP2 in SCAPs. Therefore, our results suggest that SFRP2 enhances the adipogenic and neurogenic differentiation potentials of SCAPs by up-regulating SOX2 and OCT4. Moreover, the effect of SFRP2 in neurogenic differentiation of SCAPs maybe also associated with Wnt inhibition. Our results provided useful information about the molecular mechanism underlying directed differentiation in dental tissue-derived MSCs.


Assuntos
Adipogenia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Papila Dentária/citologia , Proteínas de Membrana/farmacologia , Neurônios/citologia , Células-Tronco/citologia , Adipogenia/genética , Adolescente , Diferenciação Celular/genética , Condrogênese/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células-Tronco/efeitos dos fármacos , Adulto Jovem
9.
Cytotherapy ; 18(10): 1245-55, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27426933

RESUMO

Stem cell therapy for incurable central nervous system disorders has long been viewed as a promising therapeutic option. In this review, we discuss the existing data and approaches on cell transplantation in the context of the neural differentiation potential of adult autologous stem cells, focusing on those of mesenchymal origin as easily accessible and well studied. Mesenchymal stromal cells (MSCs) are a heterogeneous cell population with a remarkable therapeutic plasticity, demonstrated by their ability to dampen inflammation, inhibit pathogenic immune responses and secrete neuroprotective factors. To demonstrate and discuss the broad therapeutic potential of MSCs, this review focuses on two examples of neurological conditions: amyotrophic lateral sclerosis and epilepsy. We review the lessons from animal models and clinical trials, and consider encouraging newly published clinical data on therapeutic applications of neurally induced MSCs.


Assuntos
Esclerose Lateral Amiotrófica/terapia , Epilepsia/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Adulto , Células-Tronco Adultas/patologia , Células-Tronco Adultas/fisiologia , Células-Tronco Adultas/transplante , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Células Cultivadas , Epilepsia/fisiopatologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Neurais/transplante , Transplante Autólogo
10.
Bull Exp Biol Med ; 159(4): 576-81, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26395626

RESUMO

We studied a new method of treatment of amyotrophic lateral sclerosis with autologous mesenchymal stem cells. Autologous mesenchymal stem cells were injected intravenously (intact cells) or via lumbar puncture (cells committed to neuronal differentiation). Evaluation of the results of cell therapy after 12-month follow-up revealed slowing down of the disease progression in 10 patients in comparison with the control group consisting of 15 patients. The cell therapy was safe for the patients.


Assuntos
Esclerose Lateral Amiotrófica/terapia , Transplante de Células-Tronco Mesenquimais , Adulto , Idoso , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/enzimologia , Pessoa de Meia-Idade , Nestina/metabolismo , Fosfopiruvato Hidratase/metabolismo , República de Belarus , Transplante Autólogo , Resultado do Tratamento
11.
Inflamm Regen ; 44(1): 33, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39014391

RESUMO

BACKGROUND: Neonatal hypoxic-ischemic brain injury (HIBI) is a significant contributor to neonatal mortality and long-term neurodevelopmental disability, characterized by massive neuronal loss and reactive astrogliosis. Current therapeutic approaches for neonatal HIBI have been limited to general supportive therapy because of the lack of methods to compensate for irreversible neuronal loss. This study aimed to establish a feasible regenerative therapy for neonatal HIBI utilizing in vivo direct neuronal reprogramming technology. METHODS: Neonatal HIBI was induced in ICR mice at postnatal day 7 by permanent right common carotid artery occlusion and exposure to hypoxia with 8% oxygen and 92% nitrogen for 90 min. Three days after the injury, NeuroD1 was delivered to reactive astrocytes of the injury site using the astrocyte-tropic adeno-associated viral (AAV) vector AAVShH19. AAVShH19 was engineered with the Cre-FLEX system for long-term tracking of infected cells. RESULTS: AAVShH19-mediated ectopic NeuroD1 expression effectively converted astrocytes into GABAergic neurons, and the converted cells exhibited electrophysiological properties and synaptic transmitters. Additionally, we found that NeuroD1-mediated in vivo direct neuronal reprogramming protected injured host neurons and altered the host environment, i.e., decreased the numbers of activated microglia, reactive astrocytes, and toxic A1-type astrocytes, and decreased the expression of pro-inflammatory factors. Furthermore, NeuroD1-treated mice exhibited significantly improved motor functions. CONCLUSIONS: This study demonstrates that NeuroD1-mediated in vivo direct neuronal reprogramming technology through AAV gene delivery can be a novel regenerative therapy for neonatal HIBI.

12.
Microsc Res Tech ; 85(4): 1433-1443, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34859937

RESUMO

In recent years with regard to the development of nanotechnology and neural stem cell discovery, the combinatorial therapeutic strategies of neural progenitor cells and appropriate biomaterials have raised the hope for brain regeneration following neurological disorders. This study aimed to explore the proliferation and neurogenic effect of PLGA and PLGA-PEG nanofibers on human SH-SY5Y cells in in vitro condition. Nanofibers of PLGA and PLGA-PEG biomaterials were synthesized and fabricated using electrospinning method. Physicochemical features were examined using HNMR, FT-IR, and water contact angle assays. Ultrastructural morphology, the orientation of nanofibers, cell distribution and attachment were visualized by SEM imaging. Cell survival and proliferation rate were measured. Differentiation capacity was monitored by immunofluorescence staining of Map-2. HNMR, FT-IR assays confirmed the integration of PEG to PLGA backbone. Water contact angel assay showed increasing surface hydrophilicity in PLGA-PEG biomaterial compared to the PLGA substrate. SEM analysis revealed the reduction of PLGA-PEG nanofibers' diameter compared to the PLGA group. Cell attachment was observed in both groups while PLGA-PEG had a superior effect in the promotion of survival rate compared to other groups (p < .05). Compared to the PLGA group, PLGA-PEG increased the number of Ki67+ cells (p < .01). PLGA-PEG biomaterial induced neural maturation by increasing protein Map-2 compared to the PLGA scaffold in a three-dimensional culture system. According to our data, structural modification of PLGA with PEG could enhance orientated differentiation and the dynamic growth of neural cells.


Assuntos
Materiais Biocompatíveis , Nanofibras , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Proliferação de Células , Humanos , Nanofibras/química , Neurogênese , Espectroscopia de Infravermelho com Transformada de Fourier , Alicerces Teciduais/química
13.
Hum Cell ; 35(6): 1708-1721, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36040643

RESUMO

Among all the adult stem cells, odontogenic stem cells inherit the characterization of neurogenic potential of their precursor ones-the cranial crest cells. Dental follicle cells (DFCs), one of the special kind of odontogenic stem cells, are raising interest in applying to regenerative medicine for they possess multi-differentiation potential, relatively free access and ethic-friendly characteristic. Parkinson's disease (PD), as one of the common neurodegenerative disorders, affects about 0.3% of the general population. Stem cell therapies are thought to be effective to treat it. Aiming at tackling ethical-concernings, confined sources and practically applicational limits, we made use of dopaminergic neurongenic differentiation potential of the DFCs and dedicated every effort to applying them as promising cell source for treating PD. Dental follicle cells were cultured from human dental follicle tissues collected from 12 to 18-year-old teenagers' completely impacted third molars. Our data demonstrated that hDFCs were expressing mesenchymal stem cell-associated surface markers, and possessed the ability of osteogenic, adipogenic and neurogenic differentiation in vitro. Additionally, hDFCs formed neuron-like cells in vitro and in vivo, as well as expressing dopaminergic-neuronogenic marker-TH. Moreover, hDFCs survived in the transplanted areas of the Parkinson's disease model of mouse over six weeks post-surgery, and the number of TH-positive DFCs in the DFCs-Grafted group surpassed its counterpart of the MPTP group with statistically significant difference. This study indicated that hDFCs might be a promising source of dopaminergic neurons for functional transplantation, and encouraged further detailed studies on the potential of hDFCs for treating PD.


Assuntos
Doença de Parkinson , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Adolescente , Adulto , Animais , Diferenciação Celular , Células Cultivadas , Criança , Saco Dentário , Humanos , Camundongos , Osteogênese , Doença de Parkinson/terapia
14.
Nanomaterials (Basel) ; 12(18)2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36144905

RESUMO

BACKGROUND: Dental stem cells, which originate from the neural crest, due to their easy accessibility might be good candidates in neuro-regenerative procedures, along with graphene-based nanomaterials shown to promote neurogenesis in vitro. We aimed to explore the potential of liquid-phase exfoliated graphene (LPEG) film to stimulate the neuro-differentiation of stem cells from apical papilla (SCAP). METHODS: The experimental procedure was structured as follows: (1) fabrication of graphene film; (2) isolation, cultivation and SCAP stemness characterization by flowcytometry, multilineage differentiation (osteo, chondro and adipo) and quantitative PCR (qPCR); (3) SCAP neuro-induction by cultivation on polyethylene terephthalate (PET) coated with graphene film; (4) evaluation of neural differentiation by means of several microscopy techniques (light, confocal, atomic force and scanning electron microscopy), followed by neural marker gene expression analysis using qPCR. RESULTS: SCAP demonstrated exceptional stemness, as judged by mesenchymal markers' expression (CD73, CD90 and CD105), and by multilineage differentiation capacity (osteo, chondro and adipo-differentiation). Neuro-induction of SCAP grown on PET coated with graphene film resulted in neuron-like cellular phenotype observed under different microscopes. This was corroborated by the high gene expression of all examined key neuronal markers (Ngn2, NF-M, Nestin, MAP2, MASH1). CONCLUSIONS: The ability of SCAPs to differentiate toward neural lineages was markedly enhanced by graphene film.

15.
ACS Appl Bio Mater ; 5(10): 4741-4759, 2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-36102324

RESUMO

Stem cells play a critical role in peripheral nerve regeneration. Nerve scaffolds fabricated by specific materials can help induce the neurogenic differentiation of stem cells. Therefore, it is a potential strategy to enhance therapeutic efficiency. Graphene family nanomaterials are widely applied in repairing peripheral nerves. However, the mechanism underlying the pro-regeneration effects remains elusive. In this review, we first discuss the properties of graphene family nanomaterials, including monolayer and multilayer graphene, few-layer graphene, graphene oxide, reduced graphene oxide, and graphene quantum dots. We also introduce their applications in regulating stem cell differentiation. Then, we review the potential mechanisms of the neurogenic differentiation of stem cells facilitated by the materials. Finally, we discuss the existing challenges in this field to advance the development of nerve biomaterials.


Assuntos
Grafite , Nanoestruturas , Materiais Biocompatíveis , Diferenciação Celular , Grafite/farmacologia , Nanoestruturas/uso terapêutico , Regeneração Nervosa , Nervos Periféricos , Células-Tronco , Engenharia Tecidual
16.
Diagn Pathol ; 17(1): 72, 2022 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-36153506

RESUMO

BACKGROUND: DICER1-associated central nervous system sarcoma (DCS) without evidence of other cancer-related syndromes is rare. Though the morphology of DCS was highly variable, the immunophenotype was predominant myogenic phenotype. Other lineage markers were consistently negative. CASE PRESENTATION: We report a case of DCS with neurogenic differentiation proved by immunohistochemical staining and whole-exome sequencing (WES). An 8-year-old female patient presented with 8-day history of headache, nausea and vomiting. Magnetic resonance imaging (MRI) revealed a heterogeneous mass in the left parietal lobe. The patient underwent the craniotomy via left parietal approach to resect the tumor completely. Histologically, the tumor predominately showed fibrosarcoma-like spindle cells with obvious cytoplasmic eosinophilic globules. Immunohistochemically, the tumor stained positively for DICER1, Desmin, and several neurogenic markers. DICER1 somatic hotspot mutation was confirmed by WES, as well as TP53 and RAF1 mutations which were commonly found in DCS, and other sarcoma-associated genes including AR, AXL and ETV5 mutations. Subsequently, the result of Gene Ontology (GO) analysis showed that the mutated genes in this case were involved in neuron development. All of these findings indicated the diagnosis of DCS with neurogenic differentiation. Postoperatively, the patient received high-dose radiotherapy (60 Gy) and chemotherapy. There was no MRI evidence of tumor recurrence at the 21-month postoperative follow-up. CONCLUSIONS: This unusual DCS case with neuronal differentiation is an important addition to the immuno-phenotypic spectrum of DCS. Although the prognosis for DCS is poor, gross tumor resection with high dose radiotherapy and chemotherapy may assist in prolonging survival.


Assuntos
Neoplasias do Sistema Nervoso Central , Sarcoma , Neoplasias de Tecidos Moles , Sistema Nervoso Central/patologia , RNA Helicases DEAD-box/genética , Desmina/genética , Feminino , Humanos , Mutação , Recidiva Local de Neoplasia , Ribonuclease III/genética , Sarcoma/patologia
17.
Front Bioeng Biotechnol ; 10: 1074990, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36524054

RESUMO

The technologies used to generate human induced pluripotent stem cell (iPSC) from somatic cells potentially enable the wide application of iPSC-derived differentiated cells in industrial research fields as a replacement for animals. However, as highly trained individuals are required to obtain reproducible results, this approach has limited social implementation. In the research field of iPSC, it is believed that documentable information is not enough for reproducing the quality of the differentiated cells. Therefore, automated culture machines for cell processing should make the starting of iPSC-using researches easier. We developed a programmable all-in-one automated culture machine, with dense and compact constitution that fits within a normal biosafety cabinet (200 mm wide, 233 mm height, and 110 mm depth). This instrument was fabricated using novel x-y-z-axes-rail-system, such as an overhead traveling crane, in a factory, which served as the main handling machinery. This machine enabled stable and efficient expansion of human iPSC under the feeder-free condition, without karyotype alterations, and simultaneously differentiated the cells into various cell types, including cardiomyocytes, hepatocytes, neural progenitors, and keratinocytes. Overall, this machine would facilitate the social implementation of human pluripotent stem cells and contribute to the accumulation of sharable knowledge for the standardization of the entire handling processes of iPSC in pharmaceutical, food, and cosmetic research.

18.
J Biomed Mater Res A ; 110(3): 623-634, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34590393

RESUMO

The limited neurogenic potential of adult stem cells and their non-specific lineage differentiation pose major challenges in cell-replacement therapy for neurological disorders. In our previous study, we demonstrated that the neurogenic potential of stem cells from apical papilla (SCAPs) was significantly improved upon induction with a small molecule cocktail. This study attempted to investigate whether neuronal differentiation of SCAPs induced by a small molecule cocktail can be further enhanced in a three-dimensional gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor (BDNF-GelMA). The physiological properties and neural differentiation of SCAPs treated with a combination of small molecules and BDNF-GelMA were evaluated by CCK8, Live/Dead assay, quantitative reverse transcription-polymerase chain reaction, western blot and immunocytochemistry. SCAPs embedded in BDNF-GelMA displayed superior morphological characteristics when induced by a small molecule cocktail, similar to neuronal phenotypes as compared to pure GelMA. There was significant upregulation of neural markers including Tuj1 and MAP2 by SCAPs embedded in BDNF-GelMA, as compared to pure GelMA. Hence, GelMA hydrogel loaded with a potent neurotrophic factor (BDNF) provides a conducive scaffold that can further enhance the differentiation of small molecule-treated SCAPs into neuronal-like cells, which may provide a therapeutic platform for the management of neurological disorders.


Assuntos
Gelatina , Hidrogéis , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Diferenciação Celular , Células Cultivadas , Gelatina/farmacologia , Hidrogéis/farmacologia , Metacrilatos/farmacologia , Osteogênese , Células-Tronco
19.
Front Bioeng Biotechnol ; 9: 623886, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33692988

RESUMO

Human amniotic fluid stem cells (AFSC) are an exciting and very promising source of stem cells for therapeutic applications. In this study we investigated the effects of short-term treatments of small molecules to improve stem cell properties and differentiation capability. For this purpose, we used epigenetically active compounds, such as histone deacetylase inhibitors Trichostatin A (TSA) and sodium butyrate (NaBut), as well as multifunctional molecules of natural origin, such as retinoic acid (RA) and vitamin C (vitC). We observed that combinations of these compounds triggered upregulation of genes involved in pluripotency (KLF4, OCT4, NOTCH1, SOX2, NANOG, LIN28a, CMYC), but expression changes of these proteins were mild with only significant downregulation of Notch1. Also, some alterations in cell surface marker expression was established by flow cytometry with the most explicit changes in the expression of CD105 and CD117. Analysis of cellular energetics performed using Seahorse analyzer and assessment of gene expression related to cell metabolism and respiration (NRF1, HIF1α, PPARGC1A, ERRα, PKM, PDK1, LDHA, NFKB1, NFKB2, RELA, RELB, REL) revealed that small molecule treatments stimulate AFSCs toward a more energetically active phenotype. To induce cells to differentiate toward neurogenic lineage several different protocols including commercial supplements N2 and B27 together with RA were used and compared to the same differentiation protocols with the addition of a pre-induction step consisting of a combination of small molecules (vitC, TSA and RA). During differentiation the expression of several neural marker genes was analyzed (Nestin, MAP2, TUBB3, ALDH1L1, GFAP, CACNA1D, KCNJ12, KCNJ2, KCNH2) and the beneficial effect of small molecule treatment on differentiation potential was observed with upregulated gene expression. Differentiation was also confirmed by staining TUBB3, NCAM1, and Vimentin and assessed by secretion of BDNF. The results of this study provide valuable insights for the potential use of short-term small molecule treatments to improve stem cell characteristics and boost differentiation potential of AFSCs.

20.
Clin Exp Dent Res ; 7(5): 739-753, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33605088

RESUMO

OBJECTIVES: Peri-implantitis (PI) is an inflammatory disease associated with peri-implant bone loss and impaired healing potential. There is limited evidence about the presence of mesenchymal stromal cells (MSCs) and their regenerative properties within the granulation tissue (GT) of infrabony peri-implantitis defects. The aim of the present study was to characterize the cells derived from the GT of infrabony PI lesions (peri-implantitis derived mesenchymal stromal cells-PIMSCs). MATERIAL AND METHODS: PIMSC cultures were established from GT harvested from PI lesions with a pocket probing depth ≥6 mm, bleeding on probing/suppuration, and radiographic evidence of an infrabony component from four systemically healthy individuals. Cultures were analyzed for embryonic (SSEA4, NANOG, SOX2, OCT4A), mesenchymal (CD90, CD73, CD105, CD146, STRO1) and hematopoietic (CD34, CD45) stem cell markers using flow cytometry. PIMSC cultures were induced for neurogenic, angiogenic and osteogenic differentiation by respective media. Cultures were analyzed for morphological changes and mineralization potential (Alizarin Red S method). Gene expression of neurogenic (NEFL, NCAM1, TUBB3, ENO2), angiogenic (VEGFR1, VEGFR2, PECAM1) and osteogenic (ALPL, BGLAP, BMP2, RUNX2) markers was determined by quantitative RT-PCR. RESULTS: PIMSC cultures demonstrated high expression of embryonic and mesenchymal stem cell markers with inter-individual variability. After exposure to neurogenic, angiogenic and osteogenic conditions, PIMSCs showed pronounced tri-lineage differentiation potential, as evidenced by their morphology and expression of respective markers. High mineralization potential was observed. CONCLUSIONS: This study provides evidence that MSC-like populations reside within the GT of PI lesions and exhibit a multilineage differentiation potential. Further studies are needed to specify the biological role of these cells in the healing processes of inflamed PI tissues and to provide indications for their potential use in regenerative therapies.


Assuntos
Células-Tronco Mesenquimais , Peri-Implantite , Diferenciação Celular , Tecido de Granulação , Humanos , Osteogênese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA