Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Stem Cells ; 42(3): 278-289, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38134938

RESUMO

ß-thalassemia is an inherited blood disease caused by reduced or inadequate ß-globin synthesis due to ß-globin gene mutation. Our previous study developed a gene-edited mice model (ß654-ER mice) by CRISPR/Cas9-mediated genome editing, targeting both the ßIVS2-654 (C > T) mutation site and the 3' splicing acceptor site at 579 and corrected abnormal ß-globin mRNA splicing in the ß654-thalassemia mice. Herein, we further explored the therapeutic effect of the hematopoietic stem cells (HSCs) from ß654-ER mice on ß-thalassemia by consecutive HSC transplantation. The results indicated that HSC transplantation derived from gene-edited mice can significantly improve the survival rate of mice after lethal radiation doses and effectively achieve hematopoietic reconstruction and long-term hematopoiesis. Clinical symptoms, including hematologic parameters and tissue pathology of transplanted recipients, were significantly improved compared to the non-transplanted ß654 mice. The therapeutic effect of gene-edited HSC transplantation demonstrated no significant difference in hematological parameters and tissue pathology compared with wild-type mouse-derived HSCs. Our data revealed that HSC transplantation from gene-edited mice completely recovered the ß-thalassemia phenotype. Our study systematically investigated the therapeutic effect of HSCs derived from ß654-ER mice on ß-thalassemia and further confirmed the efficacy of our gene-editing approach. Altogether, it provided a reference and primary experimental data for the clinical usage of such gene-edited HSCs in the future.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Talassemia , Talassemia beta , Camundongos , Animais , Talassemia beta/genética , Talassemia beta/terapia , Edição de Genes , Células-Tronco Hematopoéticas , Globinas beta/genética
2.
Clin Genet ; 103(6): 663-671, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36999564

RESUMO

Limb-girdle muscular dystrophy recessive 1 (LGMDR1), previously known as LGMD2A, is a specific LGMD caused by a gene mutation encoding the calcium-dependent neutral cysteine protease calpain-3 (CAPN3). In our study, the compound heterozygosity with two missense variants c.635 T > C (p.Leu212Pro) and c.2120A > G (p.Asp707Gly) was identified in patients with LGMDR1. However, the pathogenicity of c.635 T > C has not been investigated. To evaluate the effects of this novel likely pathogenic variant to the motor system, the mouse model with c.635 T > C variant was prepared by CRISPR/Cas9 gene editing technique. The pathological results revealed that a limited number of inflammatory cells infiltrated the endomyocytes of certain c.635 T > C homozygous mice at 10 months of age. Compared with wild-type mice, motor function was not significantly impaired in Capn3 c. 635 T > C homozygous mice. Western blot and immunofluorescence assays further indicated that the expression levels of the Capn3 protein in muscle tissues of homozygous mice were similar to those of wild-type mice. However, the arrangement and ultrastructural alterations of the mitochondria in the muscular tissues of homozygous mice were confirmed by electron microscopy. Subsequently, muscle regeneration of LGMDR1 was simulated using cardiotoxin (CTX) to induce muscle necrosis and regeneration to trigger the injury modification process. The repair of the homozygous mice was significantly worse than that of the control mice at day 15 and day 21 following treatment, the c.635 T > C variant of Capn3 exhibited a significant effect on muscle regeneration of homozygous mice and induced mitochondrial damage. RNA-sequencing results demonstrated that the expression levels of the mitochondrial-related functional genes were significantly downregulated in the mutant mice. Taken together, the results of the present study strongly suggested that the LGMDR1 mouse model with a novel c.635 T > C variant in the Capn3 gene was significantly dysfunctional in muscle injury repair via impairment of the mitochondrial function.


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Mutação de Sentido Incorreto , Humanos , Animais , Camundongos , Proteínas Musculares/genética , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Calpaína/genética , Modelos Animais de Doenças
3.
Invest New Drugs ; 37(5): 865-875, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30488243

RESUMO

Purpose Transdifferentiation exists within stromal cells in the tumour microenvironment. Transforming growth factor-ß (TGF-ß) secreted by tumour-associated fibroblasts (TAFs) affects the differentiation states of epithelial cells, including epithelial-mesenchymal transition (EMT). Evodiamine, a natural drug, can regulate differentiation. However, the specific effects and relative mechanisms of evodiamine remain unknown. Design We used four models to observe the influence of TAF-like CCD-18Co cells on the colon epithelial cell line HCoEpiC: the 3D- and 2D-mono-culture system, Transwell and direct co-culture model. Additionally, we established conditioned medium from CCD-18Co cells. The TGF-ß pathway inhibitor LY364947 and evodiamine were added. Morphological changes and classical EMT markers were observed and detected using phase contrast microscopy and immunofluorescence. Cell migration was measured by the wound-healing assay. Western blotting was performed to detect the TGF-ß/Smad signalling pathway. Results CCD-18Co cells induced EMT-like changes in the 2D- and 3D-cultured epithelial cell line HCoEpiC, accompanied by high expression of ZEB1 and Snail and the enhancement of migration. Moreover, CCD-18Co-derived conditioned medium caused dysfunction of TGF-ß/Smad signalling in EMT. Evodiamine inhibited these EMT-like HCoEpiC and their migration. Additionally, evodiamine down-regulated the expression of ZEB1/Snail and up-regulated the expression of phosphorylated Smad2/3 (pSmad2/3). Evodiamine also increased the ratios of pSmad2/Smad2 and pSmad3/Smad3. Conclusion Based on our observations, evodiamine can reverse the TAF-induced EMT-like phenotype in colon epithelial cells, which may be associated with its mediation of phosphorylated Smad2 and Smad3 expression.


Assuntos
Fibroblastos Associados a Câncer/patologia , Colo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Quinazolinas/farmacologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Apoptose , Movimento Celular , Proliferação de Células , Colo/metabolismo , Colo/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Fosforilação , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas
4.
Genes (Basel) ; 9(8)2018 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-30060537

RESUMO

Inflammation and fibrosis in human liver are often precursors to hepatocellular carcinoma (HCC), yet none of them is easily modeled in animals. We previously generated transgenic mice with hepatocyte-specific expressed herpes simplex virus thymidine kinase (HSV-tk). These mice would develop hepatitis with the administration of ganciclovir (GCV)(Zhang, 2005 #1). However, our HSV-tk transgenic mice developed hepatitis and HCC tumor as early as six months of age even without GCV administration. We analyzed the transcriptome of the HSV-tk HCC tumor and hepatitis tissue using microarray analysis to investigate the possible causes of HCC. Gene Ontology (GO) enrichment analysis showed that the up-regulated genes in the HCC tissue mainly include the immune-inflammatory and cell cycle genes. The down-regulated genes in HCC tumors are mainly concentrated in the regions related to lipid metabolism. Gene set enrichment analysis (GSEA) showed that immune-inflammatory-related signals in the HSV-tk mice are up-regulated compared to those in Notch mice. Our study suggests that the immune system and inflammation play an important role in HCC development in HSV-tk mice. Specifically, increased expression of immune-inflammatory-related genes is characteristic of HSV-tk mice and that inflammation-induced cell cycle activation maybe a precursory step to cancer. The HSV-tk mouse provides a suitable model for the study of the relationship between immune-inflammation and HCC, and their underlying mechanism for the development of therapeutic application in the future.

5.
Oncol Rep ; 37(3): 1637-1645, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28098901

RESUMO

The tissue microenvironment functions as a crucial player in carcinogenesis, and transforming growth factor-ß1 (TGF-ß1) within the microenvironment stimulates the formation of neoplasms. Using an in vitro model of malignancy induced by TGF-ß1, we assessed the effect of evodiamine and berberine on the interaction between DNA methyltransferases (DNMTs) and target microRNAs (miRNAs) in the model. Colon tissues from neonatal rats 7 days of age were cultured and malignancy was induced by TGF-ß1 in vitro for 48 h, and then the tissues were respectively treated with evodiamine and berberine for 24 h. Morphological alteration of tissues was observed by an inverted microscope, histological structures were observed using hematoxylin and eosin staining, and the expression levels of DNMTs and targeted miRNAs screened by bioinformatics software combined with Gene chip analysis in our previous study were detected by immunohistochemistry and quantified by real-time PCR. Twenty-four hours after treatment with TGF-ß1, expression levels of DNMT1, DNMT3A, DNMT3B and miR-152 (target DNMT1), miR-429 (target DNMT3A) and miR-29a (target DNMT3A/3B) were markedly decreased; however, after 48 h, the expression levels of DNMT1 and DNMT3A were significantly increased, but their target miRNAs were still decreased. After treatment with a DNMT inhibitor (5-Aza-dC), expression levels of the miRNAs were increased to a larger extent, but did not reach normal levels. After treatment with berberine and evodiamine for 24 h, respectively, increased expression of DNMT1, DNMT3A, DNMT3B and miR-152, miR-429, miR-29a was noted. In conclusion, the results of the present study suggest that miRNAs can also be post-transcriptionally regulated by their corresponding DNMTs and that berberine and evodiamine regulate the expression of these genes, which provides early epigenetic evidence for the prevention and therapy of colorectal cancer.


Assuntos
Berberina/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Colo/metabolismo , Neoplasias do Colo/tratamento farmacológico , DNA (Citosina-5-)-Metiltransferases/metabolismo , MicroRNAs/genética , Quinazolinas/farmacologia , Fator de Crescimento Transformador beta1/toxicidade , Animais , Animais Recém-Nascidos , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Colo/patologia , Neoplasias do Colo/etiologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , DNA (Citosina-5-)-Metiltransferases/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas Imunoenzimáticas , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Mol Med Rep ; 14(6): 5429, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27779680

RESUMO

Owing to an oversight during the proof checking stage, the above article has been published with the incorrect author listed for correspondence. The first author, Chao Huang, is listed as the corresponding author, although he was only intended to have been temporarily assigned to handle queries during the pre-press stages of the publication. The correct corresponding author should have been listed as Professor Bin Wen (also at the Spleen­Stomach Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China). The email address for Professor Wen is: wenbin@gzucm.edu.cn. We sincerely apologize for this mistake, and regret any inconvenience this mistake has caused [the original article was published in the Molecular Medicine Reports 14: 2555-2565, 2016; DOI: 10.3892/mmr.2016.5584].

7.
Oncol Rep ; 36(5): 2705-2714, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27666771

RESUMO

Tissue microenvironment functions as a pivotal mediator in colorectal carcinogenesis, and its alteration can cause some important cellular responses including epigenetic events. The present study examined histologically altered tissue structure, DNA methyltransferases (DNMTs) and their corresponding expression of target microRNAs (miRNA). Tissues resected by surgery were from primary colorectal carcinoma. These samples were from three locations: and were ≥10, 5 and ≤2 cm away from the proximal lesion of colon cancer, and marked as no. 1, no. 2 and no. 3, respectively. Histological alteration was assessed by H&E staining, expression of DNMT1, DNMT3A, and DNMT3B was detected by immunohistochemistry and western blotting, microarray chip was used to screen distinguishable miRNAs and miRNAs targeting DNMTs whose validation assay was performed by quantitative real-time polymerase chain reaction (qRT-PCR). Our results revealed that normal crypt structure was shown in no. 1, while many aberrant crypt foci appeared in no. 3. Significant upregulation of DNMT1, DNMT3A, and DNMT3B expression was found in para-carcinoma tissues, compared with the histopathologically unchanged tissues (P<0.05), furthermore, distinguishable expression profiling was observed of target miRNAs in tissues with different distance. Our results provide additional insights for future research of colorectal carcinogenesis by introducing the tissue microenvironment.


Assuntos
Neoplasias Colorretais/genética , DNA (Citosina-5-)-Metiltransferases/biossíntese , Carcinogênese/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , DNA Metiltransferase 3A , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/biossíntese , Microambiente Tumoral/genética , DNA Metiltransferase 3B
8.
Mol Med Rep ; 14(3): 2555-65, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27484148

RESUMO

The present study aimed to observe the varying expression of biomarkers in the microenvironment adjacent to colorectal cancer lesions to provide additional insight into the functions of microenvironment components in carcinogenesis and present a novel or improved indicator for early diagnosis of cancer. A total of 144 human samples from three different locations in 48 patients were collected, these locations were 10, 5 and 2 cm from the colorectal cancer lesion, respectively. The biomarkers analyzed included E­cadherin, cytokeratin 18 (CK18), hyaluronidase­1 (Hyal­1), collagen type I (Col­I), Crumbs3 (CRB3), vimentin, proteinase activated receptor 3 (PAR­3), α­smooth muscle actin (α­SMA), cyclin D1 (CD1) and cluster of differentiation (CD)133. In addition, crypt architecture was observed. Related functional analysis of proteins was performed using hierarchical index cluster analysis. More severe destroyed crypt architecture closer to the cancer lesions was observed compared with the 10 cm sites, with certain crypts degraded entirely. Expression levels of E­cadherin, CK18, CRB3 and PAR­3 were lower in 2 cm sites compared with the 10 cm sites (all P<0.001), while the expression levels of the other biomarkers in the 2 cm sites were increased compared with 10 cm sites (all P<0.0001). Notably, the expression of CK18 in 2 cm sites was higher than in the 5 cm site (P<0.0001), which was different from the expression of E­cadherin, CRB3 and PAR­3. The expression levels of Hyal­1 and Col­I at the 2 cm sites were lower than that of the 5 cm sites (P>0.05 and P=0.0001, respectively), while the expression of vimentin, α­SMA, CD1 and CD133 were not. Hyal­1 and Col­I may be independently important in cancer initiation in the tumor microenvironment. The results of the present study suggest that the biomarkers in the tissue microenvironment are associated with early tumorigenesis and may contribute to the development of carcinomas. These observations may be useful for early diagnosis of colorectal cancer.

9.
Cell Biol Int ; 37(5): 420-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23504762

RESUMO

Human amniotic fluid derived progenitor cells (hAFPCs) may be multipotent and can be considered a potential tool in the field of cell therapy for haemophilia B. Their capacity to express human coagulation factor IX (hFIX) after transduction and their fate after in utero transplantation is unknown. hAFPCs isolated from second trimester pregnancies were assessed for their phenotypic markers, multilineage capacity, and expression of hFIX after transduction. Their engraftment potential was analysed in a mouse model after in utero transplantation at embryonic day 12.5. Immunohistochemistry, fluorescence in situ, ELISA and PCR were used to assess post-transplant chimeras. hAFPCs expressed several pluripotent markers, including NANOG, SOX2, SSEA4 and TRA-1-60, and could differentiate into adipocytes and osteocytes. In vitro, after transduction with hFIX and EGFP cDNAs, constitutive hFIX protein expression and clotting activity were found. Engraftment was achieved in various foetal tissues after in utero transplantation. Safe engraftment without oncogenesis was confirmed, with low donor cell levels, but persistent engraftment, into different organs (liver, heart and lung) through to 12 weeks of age. Transgenic expression of circulating hFIX was detected in recipient mice for up to 12 weeks. hAFPCs can be engrafted long-term in immunocompetent mice after in utero transplantation. Thus, cell transplantation approaches using genetically engineered hAFPCs may prove valuable for the prenatal treatment for haemophilia B.


Assuntos
Líquido Amniótico/citologia , Fator IX/metabolismo , Células-Tronco/metabolismo , Adulto , Animais , Diferenciação Celular , Células Cultivadas , Fator IX/genética , Feminino , Feto/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hemofilia B/terapia , Humanos , Hospedeiro Imunocomprometido , Camundongos , Gravidez , Segundo Trimestre da Gravidez , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transplante de Células-Tronco , Células-Tronco/citologia , Fatores de Transcrição/metabolismo
10.
J Genet Genomics ; 40(12): 617-28, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24377868

RESUMO

Hemophilia A (HA) is an inherited X-linked recessive bleeding disorder caused by coagulant factor VIII (FVIII) deficiency. Previous studies showed that introduction of mesenchymal stem cells (MSCs) modified by FVIII-expressing retrovirus may result in phenotypic correction of HA animals. This study aimed at the investigation of an alternative gene therapy strategy that may lead to sustained FVIII transgene expression in HA mice. B-domain-deleted human FVIII (hFVIIIBD) vector was microinjected into single-cell embryos of wild-type mice to generate a transgenic mouse line, from which hFVIIIBD-MSCs were isolated, followed by transplantation into HA mice. RT-PCR and real-time PCR analysis demonstrated the expression of hFVIIIBD in multi-organs of recipient HA mice. Immunohistochemistry showed the presence of hFVIIIBD positive staining in multi-organs of recipient HA mice. ELISA indicated that plasma hFVIIIBD level in recipient mice reached its peak (77 ng/mL) at the 3rd week after implantation, and achieved sustained expression during the 5-week observation period. Plasma FVIII activities of recipient HA mice increased from 0% to 32% after hFVIIIBD-MSCs transplantation. APTT (activated partial thromboplastin time) value decreased in hFVIIIBD-MSCs transplanted HA mice compared with untreated HA mice (45.5 s vs. 91.3 s). Our study demonstrated an effective phenotypic correction in HA mice using genetically modified MSCs from hFVIIIBD transgenic mice.


Assuntos
Terapia Genética/métodos , Hemofilia A/genética , Hemofilia A/terapia , Células-Tronco Mesenquimais/metabolismo , Fenótipo , Tromboplastina/genética , Tromboplastina/metabolismo , Animais , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Transgênicos , Estrutura Terciária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
Int J Epidemiol ; 39 Suppl 2: ii29-37, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21113034

RESUMO

China's methadone maintenance treatment program was initiated in 2004 as a small pilot project in just eight sites. It has since expanded into a nationwide program encompassing more than 680 clinics covering 27 provinces and serving some 242 000 heroin users by the end of 2009. The agencies that were tasked with the program's expansion have been confronted with many challenges, including high drop-out rates, poor cooperation between local governing authorities and poor service quality at the counter. In spite of these difficulties, ongoing evaluation has suggested reductions in heroin use, risky injection practices and, importantly, criminal behaviours among clients, which has thus provided the impetus for further expansion. Clinic services have been extended to offer clients a range of ancillary services, including HIV, syphilis and hepatitis C testing, information, education and communication, psychosocial support services and referrals for treatment of HIV, tuberculosis and sexually transmitted diseases. Cooperation between health and public security officials has improved through regular meetings and dialogue. However, institutional capacity building is still needed to deliver sustainable and standardized services that will ultimately improve retention rates. This article documents the steps China made in overcoming the many barriers to success of its methadone program. These lessons might be useful for other countries in the region that are scaling-up their methadone programs.


Assuntos
Educação em Saúde/organização & administração , Dependência de Heroína/tratamento farmacológico , Metadona/uso terapêutico , Programas Nacionais de Saúde/organização & administração , China/epidemiologia , Crime/estatística & dados numéricos , Infecções por HIV/complicações , Infecções por HIV/diagnóstico , Infecções por HIV/epidemiologia , Hepatite C/complicações , Hepatite C/diagnóstico , Dependência de Heroína/complicações , Humanos , Metadona/administração & dosagem , Prevalência , Qualidade da Assistência à Saúde/organização & administração , Apoio Social , Sífilis/complicações , Sífilis/diagnóstico
12.
Exp Mol Pathol ; 87(3): 173-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19666020

RESUMO

A novel intrauterine transplantation (IUT) approach was developed to improve the efficiency of engraftment of hematopoietic stem cells (HSCs). HSCs with a green fluorescent protein (GFP) reporter gene were transplanted in utero on days 12.5, 13.5 and 14.5 post coitum (p.c.). The degree of chimerism of donor cells in recipient newborn mice was examined using fluorescent microscopy, polymerase chain reaction (PCR), fluorescence-activated cell sorting (FACS), and fluorescence in situ hybridization (FISH) analyses. Microscopic examination revealed the presence of green fluorescent signal in the peripheral blood of the chimeric mice. The highest survival rate (47%) as well as the highest chimerism rate (73%) were achieved by our new approach in the newborn mice that were subjected to in utero transplantation (IUT) on day 12.5 p.c. (E12.5) compared to the conventional IUT method. FACS analysis indicated that 1.55+/-1.10% of peripheral blood cells from the newborn mice were GFP-positive donor cells. FISH showed that cells containing the donor-specific GFP sequence were present in the bone marrow (BM) of the chimeric mice. Thus, the efficiency of chimera production with this new method of IUT was significantly improved over the existing IUT techniques and instruments.


Assuntos
Quimerismo , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Útero , Animais , Separação Celular , Sobrevivência Celular , Quimera , Feminino , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde/sangue , Proteínas de Fluorescência Verde/genética , Transplante de Células-Tronco Hematopoéticas/instrumentação , Células-Tronco Hematopoéticas/citologia , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos , Microscopia de Fluorescência , Modelos Animais
13.
Int J Hematol ; 89(4): 414-421, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19387772

RESUMO

Expression of human locus control region (LCR) and beta-globin promoter has been recognized as an important factor in time- and tissue-specific expression event. DNA methylation can affect the transcriptional activity of specific genes. To investigate the methylation mechanism in the regulation of LCR and promote expression, this study used a transgenic mouse strain generated previously, in which the hematopoietic-specific expression of the EGFP was driven by human beta-globin promoter and under the control of LCR, to examine the CpG methylation pattern in various tissues. The results showed the inverse correlation between the methylated extent and the levels of gene expression in all tested tissues. We also found that the methylated extent of the 10 examined CpG sites was biased along their positions and is more efficient near the transcription start site. Real-time quantitative RT-PCR analysis of DNA methyltransferases (DNMTs) transcripts showed that Dnmt3a and Dnmt3b expressed with a very low level in the hematopoietic tissues that was coincident with the relative higher EGFP expression in these tissues, indicating that the differential expression of DNMTs contributed to the tissue-specific methylated patterns which caused the diverse gene expression in various tissues. These findings provide significant clues to elucidate the mechanism of the regulation on tissue-specific expression of genes.


Assuntos
Ilhas de CpG/genética , Metilação de DNA , Metilases de Modificação do DNA/metabolismo , Regulação da Expressão Gênica , Globinas beta/metabolismo , Animais , Células Sanguíneas/metabolismo , Metilases de Modificação do DNA/genética , Genes Reporter/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Plasmídeos/genética , Regiões Promotoras Genéticas/genética , Globinas beta/genética
14.
Haematologica ; 93(3): 356-62, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18268280

RESUMO

BACKGROUND: beta-thalassemia is one of the most common genetic diseases in the world and requires extensive therapy. Lentiviral-mediated gene therapy has been successfully exploited in the treatment of beta-thalassemia and showed promise in clinical application. Using a human beta-globin transgenic mouse line in a beta-thalassemia diseased model generated with a lentiviral-mediated approach, we investigate the stable therapeutic effect on a common thalassemia syndrome. DESIGN AND METHODS: Human beta-globin gene lentiviral vector was constr ucted, followed by subzonal microinjection into single-cell embryos of beta(IVS-2-654)-thalassemia mice to generate a transgenic line. Human beta-globin gene expression was examined with RT-PCR, Western-blotting and ELISA. The hematologic parameters and tissue pathology were investigated over time in founder mice and their off-spring. RESULTS: Transgenic mice with stable expression of the lentivirus carrying human beta-globin gene were obtained. A marked improvement in red blood cell indices and a dramatic reduction in red blood cell anisocytosis, poikilocytosis and target cells were observed. Nucleated cell proportion was greatly decreased in bone marrow, and splenomegaly with extramedullary hematopoiesis was ameliorated. Iron deposition in liver was also reduced. There was a two-fold increase in the survival rate of the beta(IVS-2-654) mice carrying human beta-globin transgene. Significantly, the germline integration of the lentiviral construct was obtained and stable hematologic phenotype correction was observed over the next two generations of the transgenic mice. CONCLUSIONS: The generation of human beta-globin transgenic mice in a beta(IVS-2-654)-thalassemia mouse mediated with lentiviral vectors provides a useful model and offers an attractive means to investigate the transgenic stable therapeutic effect in beta-thalassemia.


Assuntos
Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/uso terapêutico , Globinas/genética , Lentivirus/genética , Camundongos Transgênicos/genética , Integração Viral , Talassemia beta/genética , Animais , Medula Óssea/patologia , Feminino , Vetores Genéticos/genética , Mutação em Linhagem Germinativa , Humanos , Fígado/patologia , Masculino , Camundongos , Camundongos Mutantes , Microinjeções , Mutagênese Insercional , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Baço/patologia , Talassemia beta/patologia , Talassemia beta/terapia
15.
Hum Mol Genet ; 16(21): 2616-25, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17716993

RESUMO

The beta-thalassemia is associated with abnormality in beta-globin gene, leading to imbalanced synthesis of alpha-/beta-globin chains. Consequently, the excessive free alpha-globin chains precipitate to the erythrocyte membrane, resulting in hemolytic anemia. We have explored post-transcriptional strategies aiming at alpha-globin reduction and beta-globin enrichment on beta(654) (Hbb(th-4)/Hbb(+)) mouse, carrying a human splicing-deficient beta-globin allele (Hbb(th-4)). Lentiviral vectors of short hairpin RNA (shRNA) targeting alpha-globin and/or antisense RNA facilitating beta-globin correct splicing were microinjected into beta(654) single-cell embryos. Three transgenic strains were generated, as alpha(i)-Hbb(th-4)/Hbb(+)(shRNA), beta(a)-Hbb(th-4)/Hbb(+)(antisense) and alpha(i)beta(a)-Hbb(th-4)/Hbb(+)(both shRNA and antisense). Without notable abnormalities, all the founders and their offsprings showed sustained amelioration of hematologic parameters, ineffective erythropoiesis and extramedullary hematopoiesis. Augmented effects appeared in alpha(i)beta(a)-Hbb(th-4)/Hbb(+), which correlated with a better-balanced alpha-/beta-globin mRNA level. Among the transgenic mice integrated with shRNA and antisense RNA, one homozygous mouse (Hbb(th-4)/Hbb(th-4)) had been viable, and the 3-week survival rate for heterozygotes (Hbb(th-4)/Hbb(+)) was 97%, compared with 45.4% for untreated. Our data have demonstrated the feasibility of techniques for beta-thalassemia therapy by balancing the synthesis of alpha-/beta-globin chains.


Assuntos
Terapia Genética/métodos , Globinas/antagonistas & inibidores , RNA Antissenso/genética , RNA Interferente Pequeno/genética , Talassemia beta/terapia , Anemia/terapia , Animais , Medula Óssea/patologia , Eritropoese/genética , Expressão Gênica , Vetores Genéticos/genética , Globinas/genética , Hemoglobinas/genética , Humanos , Lentivirus/genética , Camundongos , Camundongos Transgênicos , Fenótipo , Splicing de RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA