Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 638253, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33936051

RESUMO

High resolution typing of the HLA-DPB1 locus for patient who requested for hematopoietic stem cell transplantation (HSCT) workup has recently become mandatory by the National Marrow Donor Program (NMDP) in order to facilitate matching between donors and recipients for better outcomes. The likelihood of identifying HLA matched donors in Hong Kong, on top of the existing HLA-A, -B, -C, and -DRB1 loci, is revisited in this study. HLA-A, -B, -C, -DRB1 and -DPB1 genotypes of 5,266 volunteer unrelated Chinese donors from the Hong Kong Bone Marrow Donor Registry (HKBMDR), were included in this study. Matching models were employed to determine the matching probabilities for 10/10(DPB1) and 9/10(DPB1) HLA match. The matching probabilities are 20% at 10/10(DPB1) HLA match and 55% at 9/10(DPB1) match, based on the existing 130,000 donors in the HKBMDR. The likelihoods of match become 27% and 65% respectively, by increasing the registry to 250,000. However, if DPB T-cell-epitope (TCE) model is considered in the matching, the probability will increase to 46% at 10/10 DPB1 permissive mismatching. Our findings provide vital information about the future planning on the targeted recruitment size, HLA typing and search strategies of the donor registry and arose the transplant physicians' acceptability to 9/10(DBP1) or 10/10(DBP1) HLA match. Nevertheless, the marrow donor registry has planned for increasing the registry size and bringing down the age of recruited donors which will ultimately enhance patient outcome.


Assuntos
Transplante de Medula Óssea , Epitopos de Linfócito T/genética , Genótipo , Cadeias beta de HLA-DP/genética , Doação Dirigida de Tecido , Frequência do Gene , Histocompatibilidade , Teste de Histocompatibilidade , Hong Kong , Humanos , Polimorfismo Genético , Sistema de Registros , Doadores de Tecidos
2.
Int J Cardiol ; 298: 85-92, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31668660

RESUMO

AIMS: To recapitulate progressive human dilated cardiomyopathy (DCM) and heart block in the Lmna R225X mutant mice model and investigate the molecular basis of LMNA mutation induced cardiac conduction disorders (CD); To investigate the potential interventional impact of exercise endurance. METHODS AND RESULTS: A Lmna R225X knock-in mice model in either heterozygous or homozygous genotype was generated. Electrical remodeling was observed with higher occurrence of AV block from neonatal and aged mutant mice as measured by surface electrocardiogram and atrio-ventricular Wenckebach point detection. Histological and molecular profiles revealed an increase in apoptotic cells and activation of caspase-3 activities in heart tissue. Upon aging, extracellular cellular matrix (ECM) remodeling appeared with accumulation of collagen in Lmna R225X mutant hearts as visualized by Masson's trichrome stain. This could be explained by the upregulated ECM gene expression, such as Fibronectin: Fn1, collagen: Col12a1, intergrin: Itgb2 and 3, as detected by microarray gene chip. Also, endurance exercise for 3 month improved the ventricular ejection fraction, attenuated fibrosis and cardiomyocytes apoptosis in the aged mutant mice. CONCLUSIONS: The mechanism of LMNA nonsense mutation induced cardiac conduction defects through AV node fibrosis is due to upregulated ECM gene expression upon activation of cardiac apoptosis. Lmna R225X mutant mice hold the potential for serving as in vivo models to explore the mechanism and therapeutic methods for AV block or myopathy associated with the aging process.


Assuntos
Doença do Sistema de Condução Cardíaco/genética , Cardiomiopatia Dilatada/genética , Códon sem Sentido/genética , Lamina Tipo A/genética , Condicionamento Físico Animal/fisiologia , Animais , Animais Recém-Nascidos , Doença do Sistema de Condução Cardíaco/metabolismo , Doença do Sistema de Condução Cardíaco/terapia , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/terapia , Expressão Gênica , Técnicas de Introdução de Genes/métodos , Frequência Cardíaca/fisiologia , Lamina Tipo A/biossíntese , Camundongos , Condicionamento Físico Animal/métodos
3.
Physiol Rep ; 2(2): e00237, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24744906

RESUMO

Mesenchymal stem cells (MSCs) derived from human-induced pluripotent stem cells (iPSCs) show superior proliferative capacity and therapeutic potential than those derived from bone marrow (BM). Ectopic expression of myocardin further improved the therapeutic potential of BM-MSCs in a mouse model of myocardial infarction. The aim was of this study was to assess whether forced myocardin expression in iPSC-MSCs could further enhance their transdifferentiation to cardiomyocytes and improve their electrophysiological properties for cardiac regeneration. Myocardin was overexpressed in iPSC-MSCs using viral vectors (adenovirus or lentivirus). The expression of smooth muscle cell and cardiomyocyte markers, and ion channel genes was examined by reverse transcription-polymerase chain reaction (RT-PCR), immunofluorescence staining and patch clamp. The conduction velocity of the neonatal rat ventricular cardiomyocytes cocultured with iPSC-MSC monolayer was measured by multielectrode arrays recording plate. Myocardin induced the expression of α-MHC, GATA4, α-actinin, cardiac MHC, MYH11, calponin, and SM α-actin, but not cTnT, ß-MHC, and MLC2v in iPSC-MSCs. Overexpression of myocardin in iPSC-MSC enhanced the expression of SCN9A and CACNA1C, but reduced that of KCa3.1 and Kir2.2 in iPSC-MSCs. Moreover, BKCa, IKir, ICl, Ito and INa.TTX were detected in iPSC-MSC with myocardin overexpression; while only BKCa, IKir, ICl, IKDR, and IKCa were noted in iPSC-MSC transfected with green florescence protein. Furthermore, the conduction velocity of iPSC-MSC was significantly increased after myocardin overexpression. Overexpression of myocardin in iPSC-MSCs resulted in partial transdifferentiation into cardiomyocytes phenotype and improved the electrical conduction during integration with mature cardiomyocytes.

4.
PLoS One ; 9(1): e87273, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475260

RESUMO

Hutchinson-Gillford Progeria Syndrome (HGPS) is a fatal genetic disorder characterized by premature aging in multiple organs including the skin, musculoskeletal and cardiovascular systems. It is believed that an increased mechanosensitivity of HGPS cells is a causative factor for vascular cell death and vascular diseases in HGPS patients. However, the exact mechanism is unknown. Transient receptor potential (TRP) channels are cationic channels that can act as cellular sensors for mechanical stimuli. The aim of this present study was to examine the expression and functional role of TRP channels in human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) from the patients with HGPS. The mRNA and protein expression of TRP channels in HGPS and control (IMR90) iPSC-ECs were examined by semi-quantitative RT-PCRs and immunoblots, respectively. Hypotonicity-induced cytosolic Ca²âº ([Ca²âº](i)) rise in iPSC-ECs was measured by confocal microscopy. RT-PCRs and immunoblots showed higher expressional levels of TRPV2 in iPSC-ECs from HGPS patients than those from normal individuals. In functional studies, hypotonicity induced a transient [Ca²âº](i) rise in iPSC-ECs from normal individuals but a sustained [Ca²âº](i) elevation in iPSC-ECs from HGPS patients. A nonselective TRPV inhibitor, ruthenium red (RuR, 20 µM), and a specific TRPV2 channel inhibitor, tranilast (100 µM), abolished the sustained phase of hypotonicity-induced [Ca²âº](i) rise in iPSC-ECs from HGPS patients, and also markedly attenuated the transient phase of the [Ca²âº](i) rise in these cells. Importantly, a short 10 min hypotonicity treatment caused a substantial increase in caspase 8 activity in iPSC-ECs from HGPS patients but not in cells from normal individuals. Tranilast could also inhibit the hypotonicity-induced increase in caspase 8 activity. Taken together, our data suggest that an up-regulation in TRPV2 expression causes a sustained [Ca²âº](i) elevation in HGPS-iPSC-ECs under hypotonicity, consequently resulting in apoptotic cell death. This mechanism may contribute to the pathogenesis of vascular diseases in HGPS patients.


Assuntos
Cálcio/metabolismo , Citosol/metabolismo , Regulação da Expressão Gênica/fisiologia , Células-Tronco Pluripotentes/metabolismo , Progéria/metabolismo , Canais de Cátion TRPV/metabolismo , Humanos , Immunoblotting , Microscopia Confocal , Pressão Osmótica/fisiologia , Progéria/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Am J Physiol Cell Physiol ; 303(2): C115-25, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22357737

RESUMO

The successful generation of a high yield of mesenchymal stem cells (MSCs) from human induced pluripotent stem cells (iPSCs) may represent an unlimited cell source with superior therapeutic benefits for tissue regeneration to bone marrow (BM)-derived MSCs. We investigated whether the differential expression of ion channels in iPSC-MSCs was responsible for their higher proliferation capacity than BM-MSCs. The expression of ion channels for K(+), Na(+), Ca(2+), and Cl(-) was examined by RT-PCR. The electrophysiological properties of iPSC-MSCs and BM-MSCs were then compared by patch-clamp experiments to verify their functional roles. Significant mRNA expression of ion channel genes including KCa1.1, KCa3.1, KCNH1, Kir2.1, SCN9A, CACNA1C, and Clcn3 was observed in both human iPSC-MSCs and BM-MSCs, whereas Kir2.2 and Kir2.3 were only detected in human iPSC-MSCs. Five types of currents [big-conductance Ca(2+)-activated K(+) current (BK(Ca)), delayed rectifier K(+) current (IK(DR)), inwardly rectifying K(+) current (I(Kir)), Ca(2+)-activated K(+) current (IK(Ca)), and chloride current (I(Cl))] were found in iPSC-MSCs (83%, 47%, 11%, 5%, and 4%, respectively) but only four of them (BK(Ca), IK(DR), I(Kir), and IK(Ca)) were identified in BM-MSCs (76%, 25%, 22%, and 11%, respectively). Cell proliferation was examined with MTT or bromodeoxyuridine assay, and doubling times were 2.66 and 3.72 days for iPSC-MSCs and BM-MSCs, respectively, showing a 1.4-fold discrepancy. Blockade of IK(DR) with short hairpin RNA or human ether-à-go-go 1 (hEAG1) channel blockers, 4-AP and astemizole, significantly reduced the rate of proliferation of human iPSC-MSCs. These treatments also decreased the rate of proliferation of human BM-MSCs albeit to a lesser extent. These findings demonstrate that the hEAG1 channel plays a crucial role in controlling the proliferation rate of human iPSC-MSCs and to a lesser extent in BM-MSCs.


Assuntos
Proliferação de Células , Canais de Potássio Éter-A-Go-Go/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Mesenquimais/fisiologia , Células Cultivadas , Corantes , Humanos , Potenciais da Membrana , Sais de Tetrazólio , Tiazóis
6.
Aging (Albany NY) ; 4(11): 803-822, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23362510

RESUMO

AIMS: We identified an autosomal dominant non­sense mutation (R225X) in exon 4 of the lamin A/C (LMNA) gene in a Chinese family spanning 3 generations with familial dilated cardiomyopathy (DCM). In present study, we aim to generate induced pluripotent stem cells derived cardiomyocytes (iPSC­CMs) from an affected patient with R225X and another patient bearing LMNA frame­shift mutation for drug screening. METHODS AND RESULTS: Higher prevalence of nuclear bleb formation and micronucleation was present in LMNA(R225X/WT) and LMNA(Framshift/WT) iPSC­CMs. Under field electrical stimulation, percentage of LMNA­mutated iPSC­CMs exhibiting nuclear senescence and cellular apoptosis markedly increased. shRNA knockdown of LMNA replicated those phenotypes of the mutated LMNA field electrical stress. Pharmacological blockade of ERK1/2 pathway with MEK1/2 inhibitors, U0126 and selumetinib (AZD6244) significantly attenuated the pro­apoptotic effects of field electric stimulation on the mutated LMNA iPSC­CMs. CONCLUSION: LMNA­related DCM was modeled in­vitro using patient­specific iPSC­CMs. Our results demonstrated that haploinsufficiency due to R225X LMNA non­sense mutation was associated with accelerated nuclear senescence and apoptosis of iPSC­ CMs under electrical stimulation, which can be significantly attenuated by therapeutic blockade of stress­related ERK1/2 pathway.


Assuntos
Envelhecimento/fisiologia , Cardiomiopatia Dilatada , Células-Tronco Pluripotentes Induzidas/citologia , Lamina Tipo A/genética , Modelos Biológicos , Miócitos Cardíacos/citologia , Western Blotting , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Diferenciação Celular/fisiologia , Feminino , Fibroblastos/citologia , Imunofluorescência , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Linhagem
7.
PLoS One ; 6(12): e28246, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22194816

RESUMO

BACKGROUND AND AIMS: Increasing evidence has suggested that hepatocellular carcinoma (HCC) might originate from a distinct subpopulation called cancer stem cells (CSCs), which are responsible for the limited efficacy of conventional therapies. We have previously demonstrated that granulin-epithelin precursor (GEP), a pluripotent growth factor, is upregulated in HCC but not in the adjacent non-tumor, and that GEP is a potential therapeutic target for HCC. Here, we characterized its expression pattern and stem cell properties in fetal and cancerous livers. METHODS: Protein expression of GEP in fetal and adult livers was examined in human and mouse models by immunohistochemical staining and flow cytometry. Liver cancer cell lines, isolated based on their GEP and/or ATP-dependent binding cassette (ABC) drug transporter ABCB5 expression, were evaluated for hepatic CSC properties in terms of colony formation, chemoresistance and tumorigenicity. RESULTS: We demonstrated that GEP was a hepatic oncofetal protein that expressed in the fetal livers, but not in the normal adult livers. Importantly, GEP+ fetal liver cells co-expressed the embryonic stem (ES) cell-related signaling molecules including ß-catenin, Oct4, Nanog, Sox2 and DLK1, and also hepatic CSC-markers CD133, EpCAM and ABCB5. Phenotypic characterization in HCC clinical specimens and cell lines revealed that GEP+ cancer cells co-expressed these stem cell markers similarly as the GEP+ fetal liver cells. Furthermore, GEP was shown to regulate the expression of ES cell-related signaling molecules ß-catenin, Oct4, Nanog, and Sox2. Isolated GEP(high) cancer cells showed enhanced colony formation ability and chemoresistance when compared with the GEP(low) counterparts. Co-expression of GEP and ABCB5 better defined the CSC populations with enhanced tumorigenic ability in immunocompromised mice. CONCLUSIONS: Our findings demonstrate that GEP is a hepatic oncofetal protein regulating ES cell-related signaling molecules. Co-expression of GEP and ABCB5 further enriches a subpopulation with enhanced CSC properties. The current data provide new insight into the therapeutic strategy.


Assuntos
Antígenos de Neoplasias/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Separação Celular , Transformação Celular Neoplásica , Resistencia a Medicamentos Antineoplásicos , Granulinas , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Fenótipo , Progranulinas , Ensaio Tumoral de Célula-Tronco
8.
Cell Reprogram ; 12(6): 641-53, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20858051

RESUMO

Human-induced pluripotent stem cells (iPSCs) generated from human adult somatic cells through reprogramming hold great promises for future regenerative medicine. However, exposure of human iPSCs to animal feeder and serum in the process of their generation and maintenance imposes risk of transmitting animal pathogens to human subjects, thus hindering the potential therapeutic applications. Here, we report the successful generation of human iPSCs in a feeder-independent culture system with defined factors. Two stable human iPSC lines were established from primary human dermal fibroblasts of two healthy volunteers. These human iPSCs expressed a panel of pluripotency markers including stage-specific embryonic antigen (SSEA)-4, tumor-rejection antigen (TRA)-1-60, TRA-1-81, and alkaline phosphatase, while maintaining normal karyotypes and the exogenous reprogramming factors being silenced. In addition, these human iPSCs can differentiate along lineages representative of the three embryonic germ layers upon formation of embryoid bodies, indicating their pluripotency. Furthermore, subcutaneous transplantation of these cells into immunodeficient mice resulted in teratoma formation in 6 to 8 weeks. Our findings are an important step toward generating patient-specific iPSCs in a more clinically compliant manner by eliminating the need of animal feeder cells and animal serum.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura Livres de Soro , Células-Tronco Pluripotentes Induzidas/fisiologia , Quinases Associadas a rho/antagonistas & inibidores , Adulto , Animais , Diferenciação Celular , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Cariotipagem , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...