Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-33975958

RESUMO

Genetic editing of induced pluripotent stem (iPS) cells represents a promising avenue for an HIV cure. However, certain challenges remain before bringing this approach to the clinic. Among them, in vivo engraftment of cells genetically edited in vitro needs to be achieved. In this study, CD34+ cells derived in vitro from iPS cells genetically modified to carry the CCR5Δ32 mutant alleles did not engraft in humanized immunodeficient mice. However, the CD34+ cells isolated from teratomas generated in vivo from these genetically edited iPS cells engrafted in all experiments. These CD34+ cells also gave rise to peripheral blood mononuclear cells in the mice that, when inoculated with HIV in cell culture, were resistant to HIV R5-tropic isolates. This study indicates that teratomas can provide an environment that can help evaluate the engraftment potential of CD34+ cells derived from the genetically modified iPS cells in vitro. The results further confirm the possibility of using genetically engineered iPS cells to derive engraftable hematopoietic stem cells resistant to HIV as an approach toward an HIV cure.


Assuntos
Engenharia Genética , Infecções por HIV/terapia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Diferenciação Celular , Células Cultivadas , Feminino , Edição de Genes , Humanos , Masculino , Camundongos
2.
Proc Natl Acad Sci U S A ; 115(9): 2180-2185, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29386396

RESUMO

Derivation of human hematopoietic stem cells (HSCs) from induced pluripotent stem cells (iPSCs) offers considerable promise for cell therapy, disease modeling, and drug screening. However, efficient derivation of functional iPSC-derived HSCs with in vivo engraftability and multilineage potential remains challenging. Here, we demonstrate a tractable approach for respecifying iPSC-derived blood cells into highly engraftable hematopoietic stem and progenitor cells (HSPCs) through transient expression of a single transcription factor, MLL-AF4 These induced HSPCs (iHSPCs) derived from iPSCs are able to fully reconstitute the human hematopoietic system in the recipient mice without myeloid bias. iHSPCs are long-term engraftable, but they are also prone to leukemic transformation during the long-term engraftment period. On the contrary, primary HSPCs with the same induction sustain the long-term engraftment without leukemic transformation. These findings demonstrate the feasibility of activating the HSC network in human iPSC-derived blood cells through expression of a single factor and suggest iHSPCs are more genomically instable than primary HSPCs, which merits further attention.


Assuntos
Células Eritroides , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Animais , Diferenciação Celular , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Teratoma
3.
Proc Natl Acad Sci U S A ; 113(38): 10661-5, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27601644

RESUMO

Hereditary persistence of fetal hemoglobin (HPFH) is a condition in some individuals who have a high level of fetal hemoglobin throughout life. Individuals with compound heterozygous ß-thalassemia or sickle cell disease (SCD) and HPFH have milder clinical manifestations. Using RNA-guided clustered regularly interspaced short palindromic repeats-associated Cas9 (CRISPR-Cas9) genome-editing technology, we deleted, in normal hematopoietic stem and progenitor cells (HSPCs), 13 kb of the ß-globin locus to mimic the naturally occurring Sicilian HPFH mutation. The efficiency of targeting deletion reached 31% in cells with the delivery of both upstream and downstream breakpoint guide RNA (gRNA)-guided Staphylococcus aureus Cas9 nuclease (SaCas9). The erythroid colonies differentiated from HSPCs with HPFH deletion showed significantly higher γ-globin gene expression compared with the colonies without deletion. By T7 endonuclease 1 assay, we did not detect any off-target effects in the colonies with deletion. We propose that this strategy of using nonhomologous end joining (NHEJ) to modify the genome may provide an efficient approach toward the development of a safe autologous transplantation for patients with homozygous ß-thalassemia and SCD.


Assuntos
Anemia Falciforme/genética , Hemoglobina Fetal/genética , Edição de Genes/métodos , Terapia Genética , Talassemia beta/genética , Anemia Falciforme/patologia , Anemia Falciforme/terapia , Sistemas CRISPR-Cas , Reparo do DNA por Junção de Extremidades/genética , Hemoglobina Fetal/uso terapêutico , Genoma Humano , Genótipo , Células-Tronco Hematopoéticas/metabolismo , Heterozigoto , Humanos , Deleção de Sequência , Talassemia beta/patologia , Talassemia beta/terapia
4.
Genome Res ; 24(9): 1526-33, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25096406

RESUMO

ß-thalassemia, one of the most common genetic diseases worldwide, is caused by mutations in the human hemoglobin beta (HBB) gene. Creation of human induced pluripotent stem cells (iPSCs) from ß-thalassemia patients could offer an approach to cure this disease. Correction of the disease-causing mutations in iPSCs could restore normal function and provide a rich source of cells for transplantation. In this study, we used the latest gene-editing tool, CRISPR/Cas9 technology, combined with the piggyBac transposon to efficiently correct the HBB mutations in patient-derived iPSCs without leaving any residual footprint. No off-target effects were detected in the corrected iPSCs, and the cells retain full pluripotency and exhibit normal karyotypes. When differentiated into erythroblasts using a monolayer culture, gene-corrected iPSCs restored expression of HBB compared to the parental iPSCs line. Our study provides an effective approach to correct HBB mutations without leaving any genetic footprint in patient-derived iPSCs, thereby demonstrating a critical step toward the future application of stem cell-based gene therapy to monogenic diseases.


Assuntos
Sistemas CRISPR-Cas , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação , Reparo Gênico Alvo-Dirigido/métodos , Globinas beta/genética , Animais , Elementos de DNA Transponíveis/genética , Eritroblastos/citologia , Eritroblastos/metabolismo , Eritropoese , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Talassemia beta/genética
5.
Proc Natl Acad Sci U S A ; 111(26): 9591-6, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24927590

RESUMO

Individuals homozygous for the C-C chemokine receptor type 5 gene with 32-bp deletions (CCR5Δ32) are resistant to HIV-1 infection. In this study, we generated induced pluripotent stem cells (iPSCs) homozygous for the naturally occurring CCR5Δ32 mutation through genome editing of wild-type iPSCs using a combination of transcription activator-like effector nucleases (TALENs) or RNA-guided clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 together with the piggyBac technology. Remarkably, TALENs or CRISPR-Cas9-mediated double-strand DNA breaks resulted in up to 100% targeting of the colonies on one allele of which biallelic targeting occurred at an average of 14% with TALENs and 33% with CRISPR. Excision of the piggyBac using transposase seamlessly reproduced exactly the naturally occurring CCR5Δ32 mutation without detectable exogenous sequences. We differentiated these modified iPSCs into monocytes/macrophages and demonstrated their resistance to HIV-1 challenge. We propose that this strategy may provide an approach toward a functional cure of HIV-1 infection.


Assuntos
Diferenciação Celular/imunologia , Resistência à Doença/genética , Engenharia Genética/métodos , Infecções por HIV/genética , Células-Tronco Pluripotentes Induzidas/imunologia , Receptores CCR5/genética , Deleção de Sequência/genética , Southern Blotting , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Primers do DNA/genética , Desoxirribonucleases/metabolismo , Resistência à Doença/imunologia , Imunofluorescência , Vetores Genéticos/genética , Infecções por HIV/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Macrófagos/citologia , Monócitos/citologia , Mutagênese/genética , Transposases/metabolismo
7.
Clin Sci (Lond) ; 129(12): 989-99, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26386022

RESUMO

The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) is a major regulator of oxidative stress defence in the human body. As Nrf2 regulates the expression of a large battery of cytoprotective genes, it plays a crucial role in the prevention of degenerative disease in multiple organs. Thus it has been the focus of research as a pharmacological target that could be used for prevention and treatment of chronic diseases such as multiple sclerosis, chronic kidney disease or cardiovascular diseases. The present review summarizes promising findings from basic research and shows which Nrf2-targeting therapies are currently being investigated in clinical trials and which agents have already entered clinical practice.


Assuntos
Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Transdução de Sinais , Animais , Antioxidantes/farmacologia , Descoberta de Drogas , Regulação da Expressão Gênica , Humanos , Nefropatias/tratamento farmacológico , Nefropatias/metabolismo , Hepatopatias/tratamento farmacológico , Hepatopatias/metabolismo , Pneumopatias/tratamento farmacológico , Pneumopatias/metabolismo , Terapia de Alvo Molecular , Fator 2 Relacionado a NF-E2/agonistas , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/genética , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
J Pathol ; 234(4): 538-47, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25111334

RESUMO

Skeletal muscles harbour a resident population of stem cells, termed satellite cells (SCs). After trauma, SCs leave their quiescent state to enter the cell cycle and undergo multiple rounds of proliferation, a process regulated by MyoD. To initiate differentiation, fusion and maturation to new skeletal muscle fibres, SCs up-regulate myogenin. However, the regulation of these myogenic factors is not fully understood. In this study we demonstrate that Nrf2, a major regulator of oxidative stress defence, plays a role in the expression of these myogenic factors. In both promoter studies with myoblasts and a mouse model of muscle injury in Nrf2-deficient mice, we show that Nrf2 prolongs SC proliferation by up-regulating MyoD and suppresses SC differentiation by down-regulating myogenin. Moreover, we show that IL-6 and HGF, both factors that facilitate SC activation, induce Nrf2 activity in myoblasts. Thus, Nrf2 activity promotes muscle regeneration by modulating SC proliferation and differentiation and thereby provides implications for tissue regeneration.


Assuntos
Músculo Esquelético/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo , Regeneração/fisiologia , Traumatismo por Reperfusão/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Animais , Western Blotting , Diferenciação Celular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/patologia , Proteína MyoD/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Transfecção
9.
Am J Respir Cell Mol Biol ; 51(5): 668-77, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24921206

RESUMO

Mechanical ventilation (MV) elicits complex and clinically relevant cellular responses in the lungs. The current study was designed to define the role of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), a major regulator of the cellular antioxidant defense system, in the pulmonary response to MV. Nrf2 activity was quantified in ventilated isolated perfused mouse lungs (IPL). Regulation of amphiregulin (AREG) was investigated in BEAS-2B cells with inactivated Nrf2 or Keap1, the inhibitor of Nrf2, using a luciferase vector with AREG promoter. AREG-dependent Nrf2 activity was examined in BEAS-2B cells, murine precision-cut lung slices (PCLS), and IPL. Finally, Nrf2 knockout and wild-type mice were ventilated to investigate the interplay between Nrf2 and AREG during MV in vivo. Lung functions and inflammatory parameters were measured. Nrf2 was activated in a ventilation-dependent manner. The knockdown of Nrf2 and Keap1 via short hairpin RNA in BEAS-2B cells and an EMSA with lung tissue revealed that AREG is regulated by Nrf2. Conversely, AREG application induced a significant Nrf2 activation in BEAS-2B cells, PCLS, and IPL. The signal transduction of ventilation-induced Nrf2 activation was shown to be p38 MAP kinase-dependent. In vivo ventilation experiments indicated that AREG is regulated by Nrf2 during MV. We conclude that Areg expression is regulated by Nrf2. During high-pressure ventilation, Nrf2 becomes activated and induces AREG, leading to a positive feedback loop between Nrf2 and AREG, which involves the p38 MAPK and results in the expression of cytoprotective genes.


Assuntos
Brônquios/fisiologia , Família de Proteínas EGF/metabolismo , Fator 2 Relacionado a NF-E2/genética , Respiração Artificial , Anfirregulina , Animais , Elementos de Resposta Antioxidante/fisiologia , Brônquios/citologia , Células Cultivadas , Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/metabolismo , Técnicas de Cultura de Órgãos , Regiões Promotoras Genéticas/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Calcif Tissue Int ; 95(4): 349-61, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25096517

RESUMO

Oxidative stress plays an important role in wound healing but data relating oxidative stress to fracture healing are scarce. Nuclear factor erythroid 2-related factor 2 (Nrf2) is the major transcription factor that controls the cellular defence essential to combat oxidative stress by regulating the expression of antioxidative enzymes. This study examined the impact of Nrf2 on fracture healing using a standard closed femoral shaft fracture model in wild-type (WT) and Nrf2-knockout (Nrf2-KO)-mice. Healing was evaluated by histology, real-time RT-PCR, µCT and biomechanical measurements. We showed that Nrf2 expression is activated during fracture healing. Bone healing and remodelling were retarded in the Nrf2-KO compared to the WT-mice. Nrf2-KO-mice developed significantly less callus tissue compared to WT-mice. In addition, biomechanical testing demonstrated lower strength against shear stress in the Nrf2-KO-group compared to WT. The expression of vascular endothelial growth factor (VEGF) and osteocalcin is reduced during fracture healing in Nrf2-KO-mice. Taken together, our results demonstrate that Nrf2 deficiency in mice results in impaired fracture healing suggesting that Nrf2 plays an essential role in bone regeneration. Pharmacological activation of Nrf2 may have therapeutic potential for the enhancement of fracture healing.


Assuntos
Consolidação da Fratura , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Animais , Fenômenos Biomecânicos , Regeneração Óssea , Diferenciação Celular , Condrócitos/citologia , Fêmur/patologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteocalcina/metabolismo , Osteoclastos/metabolismo , Estresse Oxidativo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Microtomografia por Raio-X
11.
Proc Natl Acad Sci U S A ; 108(5): 2064-9, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21245320

RESUMO

VEGF and angiopoietin-1 (Ang1) are two major angiogenic factors being investigated for the treatment of myocardial infarction (MI). Targeting VEGF and Ang1 expression in the ischemic myocardium can increase their local therapeutic effects and reduce possible adverse effects. Adeno-associated viral vectors (AAVs) expressing cardiac-specific and hypoxia-inducible VEGF [AAV-myosin light chain-2v (MLC)VEGF] and Ang1 (AAV-MLCAng1) were coinjected (VEGF/Ang1 group) into six different sites of the porcine myocardium at the peri-infarct zone immediately after ligating the left descending coronary artery. An identical dose of AAV-Cytomegalovirus (CMV)LacZ or saline was injected into control animals. AAV genomes were detected in the liver in addition to the heart. RT-PCR, Western blotting, and ELISA analyses showed that VEGF and Ang1 were predominantly expressed in the myocardium in the infarct core and border of the infarct heart. Gated single-photon emission computed tomography analyses showed that the VEGF/Ang1 group had better cardiac function and myocardial perfusion at 8 wk than at 2 wk after vector injection. Compared with the saline and LacZ controls, the VEGF/Ang1 group expressed higher phosphorylated Akt and Bcl-xL, less Caspase-3 and Bad, and had higher vascular density, more proliferating cardiomyocytes, and less apoptotic cells in the infarct and peri-infarct zones. Thus, cardiac-specific and hypoxia-induced coexpression of VEGF and Ang1 improves the perfusion and function of porcine MI heart through the induction of angiogenesis and cardiomyocyte proliferation, activation of prosurvival pathways, and reduction of cell apoptosis.


Assuntos
Angiopoietina-1/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Angiopoietina-1/fisiologia , Animais , Proliferação de Células , Vasos Coronários/crescimento & desenvolvimento , Suínos , Fator A de Crescimento do Endotélio Vascular/fisiologia
12.
Proc Natl Acad Sci U S A ; 107(45): 19467-72, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974949

RESUMO

To date, a large number of reports have described reprogramming many somatic cell types into induced pluripotent stem (iPS) cells, using different numbers of transcription factors and devising alternate methods of introducing the transcription factor genes or proteins into the somatic cells. Here, we describe a method using bacteriophage ΦC31 integrase to reprogram mouse embryonic fibroblasts and human amniotic fluid cells into iPS cells. These iPS cells showed morphology, surface antigens, gene expression, and epigenetic states similar to ES cells and formed teratomas with three germ layers in nonobese diabetic/severely compromised immunodeficient mice. Importantly, these iPS cells have only a single integration site in each cell line. The locations of integration favor the intergenic regions, and their distances from the adjacent genes extended from several hundred to >1 million bp. The effect of the insertion on the expression of these genes can be studied by RT-PCR. No insertion into microRNA gene loci was detected. Hence, it is possible to select cells in which adjacent gene functions are not affected, or the inserts can be removed if necessary. We conclude that phage integrase-mediated site-specific recombination can produce iPS cells that have undisturbed endogenous gene function and could be safe for future human therapeutic application.


Assuntos
Bacteriófagos/enzimologia , Células-Tronco Pluripotentes Induzidas/citologia , Integrases/genética , Líquido Amniótico/citologia , Animais , Técnicas de Cultura de Células , Reprogramação Celular , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Humanos , Camundongos
13.
J Biol Chem ; 286(6): 4493-9, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21127061

RESUMO

IL-6 gene expression is controlled by a promoter region containing multiple regulatory elements such as NF-κB, NF-IL6, CRE, GRE, and TRE. In this study, we demonstrated that TRE, found within the IL-6 promoter, is embedded in a functional antioxidant response element (ARE) matching an entire ARE consensus sequence. Further, point mutations of the ARE consensus sequence in the IL-6 promoter construct selectively eliminate ARE but not TRE activity. Nrf2 is a redox-sensitive transcription factor which provides cytoprotection against electrophilic and oxidative stress and is the most potent activator of ARE-dependent transcription. Using Nrf2 knock-out mice we demonstrate that Nrf2 is a potent activator of IL-6 gene transcription in vivo. Moreover, we show evidence that Nrf2 is the transcription factor that activates IL6 expression in a cholestatic hepatitis mouse model. Our findings suggest a possible role of IL-6 in oxidative stress defense and also give indication about an important function for Nrf2 in the regulation of hematopoietic and inflammatory processes.


Assuntos
Antioxidantes , Regulação da Expressão Gênica , Interleucina-6/biossíntese , Fator 2 Relacionado a NF-E2/metabolismo , Elementos de Resposta , Animais , Modelos Animais de Doenças , Células Hep G2 , Hepatite/genética , Hepatite/metabolismo , Humanos , Interleucina-6/genética , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/genética , Mutação Puntual , Transcrição Gênica
14.
EMBO J ; 27(1): 212-23, 2008 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-18059474

RESUMO

The liver is frequently challenged by surgery-induced metabolic overload, viruses or toxins, which induce the formation of reactive oxygen species. To determine the effect of oxidative stress on liver regeneration and to identify the underlying signaling pathways, we studied liver repair in mice lacking the Nrf2 transcription factor. In these animals, expression of several cytoprotective enzymes was reduced in hepatocytes, resulting in oxidative stress. After partial hepatectomy, liver regeneration was significantly delayed. Using in vitro and in vivo studies, we identified oxidative stress-mediated insulin/insulin-like growth factor resistance as an underlying mechanism. This deficiency impaired the activation of p38 mitogen-activated kinase, Akt kinase and downstream targets after hepatectomy, resulting in enhanced death and delayed proliferation of hepatocytes. Our results reveal novel roles of Nrf2 in the regulation of growth factor signaling and in tissue repair. In addition, they provide new insight into the mechanisms underlying oxidative stress-induced defects in liver regeneration. These findings may provide the basis for the development of new strategies to improve regeneration in patients with acute or chronic liver damage.


Assuntos
Resistência à Insulina , Fator de Crescimento Insulin-Like I/fisiologia , Insulina/fisiologia , Regeneração Hepática/genética , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Hepatócitos/metabolismo , Hepatócitos/patologia , Resistência à Insulina/genética , Masculino , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/fisiologia , Tamanho do Órgão/genética , Oxirredução , Estresse Oxidativo/genética
15.
Proc Natl Acad Sci U S A ; 106(24): 9826-30, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19482945

RESUMO

The innovation of reprogramming somatic cells to induced pluripotent stem cells provides a possible new approach to treat beta-thalassemia and other genetic diseases such as sickle cell anemia. Induced pluripotent stem (iPS) cells can be made from these patients' somatic cells and the mutation in the beta-globin gene corrected by gene targeting, and the cells differentiated into hematopoietic cells to be returned to the patient. In this study, we reprogrammed the skin fibroblasts of a patient with homozygous beta(0) thalassemia into iPS cells, and showed that the iPS cells could be differentiated into hematopoietic cells that synthesized hemoglobin. Prenatal diagnosis and selective abortion have been effective in decreasing the number of beta-thalassemia births in some countries that have instituted carrier screening and genetic counseling. To make use of the cells from the amniotic fluid or chorionic villus sampling that are used for prenatal diagnosis, we also showed that these cells could be reprogrammed into iPS cells. This raises the possibility of providing a new option following prenatal diagnosis of a fetus affected by a severe illness. Currently, the parents would choose either to terminate the pregnancy or continue it and take care of the sick child after birth. The cells for prenatal diagnosis can be converted into iPS cells for treatment in the perinatal periods. Early treatment has the advantage of requiring much fewer cells than adult treatment, and can also prevent organ damage in those diseases in which damage can begin in utero or at an early age.


Assuntos
Anemia Falciforme/terapia , Células-Tronco Pluripotentes/citologia , Diagnóstico Pré-Natal , Talassemia beta/terapia , Anemia Falciforme/diagnóstico , Células Cultivadas , Humanos , Talassemia beta/diagnóstico
16.
Proc Natl Acad Sci U S A ; 106(8): 2933-8, 2009 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19196989

RESUMO

Oxidative stress has been implicated in the etiology of Parkinson's disease (PD) and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) animal model of PD. It is known that under conditions of oxidative stress, the transcription factor NF-E2-related factor (Nrf2) binds to antioxidant response element (ARE) to induce antioxidant and phase II detoxification enzymes. To investigate the role of Nrf2 in the process of MPTP-induced toxicity, mice expressing the human placental alkaline phosphatase (hPAP) gene driven by a promoter containing a core ARE sequence (ARE-hPAP) were used. ARE-hPAP mice were injected (30 mg/kg) once per day for 5 days and killed 7 days after the last MPTP injection. In response to this design, ARE-dependent gene expression was decreased in striatum whereas it was increased in substantia nigra. The same MPTP protocol was applied in Nrf2(+/+) and Nrf2(-/-) mice; Nrf2 deficiency increases MPTP sensitivity. Furthermore, we evaluated the potential for astrocytic Nrf2 overexpression to protect from MPTP toxicity. Transgenic mice with Nrf2 under control of the astrocyte-specific promoter for the glial fribillary acidic protein (GFAP-Nrf2) on both a Nrf2(+/+) and Nrf2(-/-) background were administered MPTP. In the latter case, only the astrocytes expressed Nrf2. Independent of background, MPTP-mediated toxicity was abolished in GFAP-Nrf2 mice. These striking results indicate that Nrf2 expression restricted to astrocytes is sufficient to protect against MPTP and astrocytic modulation of the Nrf2-ARE pathway is a promising target for therapeutics aimed at reducing or preventing neuronal death in PD.


Assuntos
Astrócitos/patologia , Modelos Animais de Doenças , Intoxicação por MPTP/fisiopatologia , Fator 2 Relacionado a NF-E2/fisiologia , Doença de Parkinson/fisiopatologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Fosfatase Alcalina/genética , Animais , Sequência de Bases , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Primers do DNA , Proteína Glial Fibrilar Ácida/genética , Imuno-Histoquímica , Intoxicação por MPTP/patologia , Camundongos , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/genética , Doença de Parkinson/patologia , Substância Negra/efeitos dos fármacos , Substância Negra/patologia
17.
Ann Rheum Dis ; 70(5): 844-50, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21173018

RESUMO

OBJECTIVES: Increasing evidence suggests that oxidative stress may play a key role in joint destruction due to rheumatoid arthritis (RA). The aim of this study was to elucidate the role of nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that maintains the cellular defence against oxidative stress, in RA. METHODS: The activation status of Nrf2 was assessed in synovial tissue from patients with RA using immunohistochemistry. Antibody-induced arthritis (AIA) was induced in Nrf2-knockout and Nrf2-wild-type control mice. The severity of cartilage destruction was evaluated using a damage score. The extent of oxidative stress, the activation state of Nrf2 and the expression level of Nrf2 target genes were analysed by immunhistological staining. The expression of vascular endothelial growth factor (VEGF)-A was examined on mRNA and protein using the Luminex technique. A Xenogen imaging system was used to measure Nrf2 activity in an antioxidant response element-luciferase transgenic mouse during AIA. RESULTS: Nrf2 was activated in the joints of arthritic mice and of patients with RA. Nrf2-knockout mice had more severe cartilage injuries and more oxidative damage, and the expression of Nrf2 target genes was enhanced in Nrf2-wild-type but not in knockout mice during AIA. Both VEGF-A mRNA and protein expression was upregulated in Nrf2-knockout mice during AIA. An unexpected finding was the number of spontaneously fractured bones in Nrf2-knockout mice with AIA. CONCLUSION: These results provide strong evidence that oxidative stress is significantly involved in cartilage degradation in experimental arthritis, and indicate that the presence of a functional Nrf2 gene is a major requirement for limiting cartilage destruction.


Assuntos
Artrite Experimental/patologia , Artrite Reumatoide/patologia , Fator 2 Relacionado a NF-E2/fisiologia , Estresse Oxidativo/fisiologia , Animais , Artrite Experimental/complicações , Artrite Experimental/metabolismo , Artrite Experimental/fisiopatologia , Artrite Reumatoide/complicações , Artrite Reumatoide/metabolismo , Artrite Reumatoide/fisiopatologia , Cartilagem Articular/patologia , Cartilagem Articular/fisiopatologia , Fraturas Espontâneas/etiologia , Fraturas Espontâneas/patologia , Expressão Gênica , Glutamato-Cisteína Ligase/metabolismo , Heme Oxigenase-1/metabolismo , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/deficiência , RNA Mensageiro/genética , Tiorredoxinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
18.
AIDS ; 34(8): 1141-1149, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32287059

RESUMO

OBJECTIVE: To assess the in-vitro CCR5---tropic and CXCR4---tropic HIV---1 infectivity of immune cells, particularly macrophages, derived from CCR5 gene---edited induced pluripotent stem cells (iPSCs) obtained from the peripheral blood mononuclear cells (PBMC) of HIV---infected patients on antiretroviral therapy (ART). DESIGN: PBMC were obtained from six patients who had been HIV---infected for over 20 years and were on ART for 1---12 years prior to this study. METHODS: The PBMC were derived into iPSCs and genetically edited with TALENs or CRISPR---cas9 endonucleases combined with PiggyBac technology to introduce the naturally occurring 32---bp deletion to the CCR5 gene. These iPSCs were differentiated into macrophages, and subsequently challenged with CCR5---tropic or CCR5/CXCR4 dual--- tropic HIV---1 strains. iPSC derivation, gene editing and immune cell differentiation were done in feeder---free, xeno---free in-vitro conditions. RESULTS: Multiple unedited (wild---type) and CCR5 gene---edited (mutant) iPSCs were derived from patients' PBMC. When differentiated into immune cells and HIV---1 challenged, mutant iPSC lines were resistant to CCR5---tropic and to some extent to CCR5/CXCR4 dual---tropic HIV---1 infection when compared to wild---type iPSC lines. CONCLUSION: Our study demonstrates that iPSC---derived, gene---edited immune cells are resistant to distinct HIV---1 strains. These findings have important implications for both in-vitro stem cell development and therapeutic approaches to cure HIV infection.


Assuntos
Infecções por HIV/terapia , HIV-1/genética , Células-Tronco Pluripotentes Induzidas , Leucócitos Mononucleares , Receptores de HIV/fisiologia , Proteína 9 Associada à CRISPR , HIV-1/fisiologia , Humanos , Receptores CCR5/genética , Receptores CXCR4/genética , Receptores CXCR4/fisiologia
19.
J Gene Med ; 11(9): 743-53, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19554624

RESUMO

BACKGROUND: Transplantation of stem cells from various sources into infarcted hearts has the potential to promote myocardial regeneration. However, the regenerative capacity is limited partly as a result of the low survival rate of the transplanted cells in the ischemic myocardium. In the present study, we tested the hypothesis that combining cell and angiogenic gene therapies would provide additive therapeutic effects via co-injection of bone marrow-derived mesenchymal stem cells (MSCs) with an adeno-associated viral vector (AAV), MLCVEGF, which expresses vascular endothelial growth factor (VEGF) in a cardiac-specific and hypoxia-inducible manner. METHODS: MSCs isolated from transgenic mice expressing green fluorescent protein and MLCVEGF packaged in AAV serotype 1 capsid were injected into mouse hearts at the border of ischemic area, immediately after occlusion of the left anterior descending coronary, individually or together. Engrafted cells were detected and quantified by real-time polymerase chain reaction and immunostaining. Angiogenesis and infarct size were analyzed on histological and immunohistochemical stained sections. Cardiac function was analyzed by echocardiography. RESULTS: We found that co-injection of AAV1-MLCVEGF with MSCs reduced cell loss. Although injection of MSCs and AAV1-MLCVEGF individually improved cardiac function and reduced infarct size, co-injection of MSC and AAV1-MLCVEGF resulted in the best improvement in cardiac function as well as the smallest infarct among all groups. Moreover, injection of AAV1-MLCVEGF induced neovasculatures. Nonetheless, injection of MSCs attracted endogenous stem cell homing and increased scar thickness. CONCLUSIONS: Co-injection of MLCVEGF and MSCs in ischemic hearts can result in better cardiac function and MSC survival, compared to their individual injections, as a result of the additive effects of each therapy.


Assuntos
Terapia Genética , Infarto do Miocárdio/terapia , Neovascularização Fisiológica/genética , Transplante de Células-Tronco , Fator A de Crescimento do Endotélio Vascular/genética , Adenoviridae/genética , Animais , Sequência de Bases , Western Blotting , Diferenciação Celular , Separação Celular , Primers do DNA , Feminino , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/cirurgia
20.
Mol Cell Biol ; 26(3): 940-54, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428448

RESUMO

Genetic and biochemical analyses have uncovered an essential role for nuclear factor erythroid 2-related factor 2 (Nrf2) in regulating phase II xenobiotic metabolism and antioxidant response. Here we show that Nrf2 protects against the ovarian toxicity of 4-vinylcyclohexene diepoxide (VCD) in mice. Nrf2-/- female mice exposed to VCD exhibit an age-dependent decline in reproduction leading to secondary infertility accompanied by hypergonadotropic hypogonadism after 30 weeks of age. VCD is shown to selectively destroy small ovarian follicles, resulting in early depletion of functional follicles. Treatment with VCD induces apoptotic death in cultured cells and in ovarian follicles, suggesting apoptosis as a mechanism of follicle loss. Loss of Nrf2 function blocks the basal and inducible expression of microsomal epoxide hydrolase, a key enzyme in the detoxification of VCD, and increases the oxidative stress in cells that is further exacerbated by VCD. Foxo3a, a repressor in the early stages of follicle activation, displays reduced expression in Nrf2-/- ovaries, causing accelerated growth of follicles in the absence of exposure to exogenous chemicals. Furthermore, Foxo3a is degraded through the 26S proteasome pathway in untreated cells and is induced by VCD via both Nrf2-dependent transcription and protein stabilization. This study demonstrates that Nrf2 serves as an essential sensor and regulator of chemical homeostasis in ovarian cells, protecting the cells from toxic chemicals by controlling metabolic detoxification, reactive oxygen species defense, and Foxo3a expression. In addition, these findings raise the possibility that exposure to environmental or occupational ovotoxicants plays a role in the premature ovarian failure commonly associated with infertility and premature aging in women.


Assuntos
Cicloexanos/toxicidade , Fatores de Transcrição Forkhead/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Folículo Ovariano/efeitos dos fármacos , Insuficiência Ovariana Primária/induzido quimicamente , Compostos de Vinila/toxicidade , Animais , Apoptose , Cicloexenos , Epóxido Hidrolases/metabolismo , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Inativação Metabólica/genética , Camundongos , Camundongos Mutantes , Mutação , Fator 2 Relacionado a NF-E2/genética , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Estresse Oxidativo/genética , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA