Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33975958

ABSTRACT

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.


Subject(s)
Genetic Engineering , HIV Infections/therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Female , Gene Editing , Humans , Male , Mice
2.
AIDS ; 34(8): 1141-1149, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32287059

ABSTRACT

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.


Subject(s)
HIV Infections/therapy , HIV-1/genetics , Induced Pluripotent Stem Cells , Leukocytes, Mononuclear , Receptors, HIV/physiology , CRISPR-Associated Protein 9 , HIV-1/physiology , Humans , Receptors, CCR5/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/physiology
4.
Proc Natl Acad Sci U S A ; 115(9): 2180-2185, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29386396

ABSTRACT

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.


Subject(s)
Erythroid Cells , Hematopoietic Stem Cells/physiology , Induced Pluripotent Stem Cells/drug effects , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Cell Differentiation , Gene Expression Regulation/physiology , Humans , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Teratoma
5.
Proc Natl Acad Sci U S A ; 113(38): 10661-5, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601644

ABSTRACT

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.


Subject(s)
Anemia, Sickle Cell/genetics , Fetal Hemoglobin/genetics , Gene Editing/methods , Genetic Therapy , beta-Thalassemia/genetics , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/therapy , CRISPR-Cas Systems , DNA End-Joining Repair/genetics , Fetal Hemoglobin/therapeutic use , Genome, Human , Genotype , Hematopoietic Stem Cells/metabolism , Heterozygote , Humans , Sequence Deletion , beta-Thalassemia/pathology , beta-Thalassemia/therapy
6.
Stem Cells Dev ; 25(16): 1243-8, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27328768

ABSTRACT

Fibroblasts can be transdifferentiated directly into other somatic cells such as cardiomyocytes, hematopoietic cells, and neurons. An advantage of somatic cell differentiation without first generating induced pluripotent stem cells (iPSCs) is that it avoids contamination of the differentiated cells with residual iPSCs, which may cause teratoma. However, since primary fibroblasts from biopsy undergo senescence during repeated culture, it may be difficult to grow transdifferentiated cells in sufficient numbers for future therapeutic purposes. To circumvent this problem, we reversibly immortalized primary fibroblasts by using the piggyBac transposon to deliver the human telomerase reverse transcriptase (hTERT) gene hTERT plus SV40 Large T. Both approaches enabled fibroblasts to grow continuously without senescence, and neither caused teratoma formation in immunodeficient mice. However, fibroblasts immortalized with hTERT plus SV40 large T antigen accumulated chromosomal rearrangements, whereas fibroblasts immortalized with hTERT retained the normal karyotype. To transdifferentiate hTERT-immortalized fibroblasts into other somatic lineage cells, we transiently transfected them with episomal OCT4 and cultured them under neural cell growth condition with transposase to remove the transposon. Tripotent neural progenitor cells were seamlessly and efficiently generated. Thus, reversible immortalization of primary fibroblasts with hTERT will allow potential autologous cell-based therapeutics that bypass and simulate iPSC generation.


Subject(s)
Cell Lineage , Cell Transdifferentiation , Fibroblasts/cytology , Animals , Carcinogenesis/pathology , Cell Line, Transformed , Colony-Forming Units Assay , DNA Transposable Elements/genetics , Humans , Karyotyping , Mice, SCID , Neural Stem Cells/cytology , Transfection
7.
Clin Sci (Lond) ; 129(12): 989-99, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26386022

ABSTRACT

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.


Subject(s)
NF-E2-Related Factor 2/metabolism , Oxidative Stress , Signal Transduction , Animals , Antioxidants/pharmacology , Drug Discovery , Gene Expression Regulation , Humans , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Liver Diseases/drug therapy , Liver Diseases/metabolism , Lung Diseases/drug therapy , Lung Diseases/metabolism , Molecular Targeted Therapy , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects
8.
J Pathol ; 234(4): 538-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25111334

ABSTRACT

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.


Subject(s)
Muscle, Skeletal/physiology , NF-E2-Related Factor 2/metabolism , Regeneration/physiology , Reperfusion Injury/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Animals , Blotting, Western , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , MyoD Protein/metabolism , Oligonucleotide Array Sequence Analysis , Transfection
9.
Genome Res ; 24(9): 1526-33, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25096406

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Induced Pluripotent Stem Cells/metabolism , Mutation , Targeted Gene Repair/methods , beta-Globins/genetics , Animals , DNA Transposable Elements/genetics , Erythroblasts/cytology , Erythroblasts/metabolism , Erythropoiesis , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Mice , beta-Thalassemia/genetics
10.
Calcif Tissue Int ; 95(4): 349-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25096517

ABSTRACT

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.


Subject(s)
Fracture Healing , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Animals , Biomechanical Phenomena , Bone Regeneration , Cell Differentiation , Chondrocytes/cytology , Femur/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteocalcin/metabolism , Osteoclasts/metabolism , Oxidative Stress , Vascular Endothelial Growth Factor A/metabolism , X-Ray Microtomography
11.
Am J Respir Cell Mol Biol ; 51(5): 668-77, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24921206

ABSTRACT

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.


Subject(s)
Bronchi/physiology , EGF Family of Proteins/metabolism , NF-E2-Related Factor 2/genetics , Respiration, Artificial , Amphiregulin , Animals , Antioxidant Response Elements/physiology , Bronchi/cytology , Cells, Cultured , Feedback, Physiological/physiology , Gene Expression Regulation/physiology , Humans , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Organ Culture Techniques , Promoter Regions, Genetic/physiology , Respiratory Mucosa/cytology , Respiratory Mucosa/physiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Proc Natl Acad Sci U S A ; 111(26): 9591-6, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24927590

ABSTRACT

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.


Subject(s)
Cell Differentiation/immunology , Disease Resistance/genetics , Genetic Engineering/methods , HIV Infections/genetics , Induced Pluripotent Stem Cells/immunology , Receptors, CCR5/genetics , Sequence Deletion/genetics , Blotting, Southern , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Primers/genetics , Deoxyribonucleases/metabolism , Disease Resistance/immunology , Fluorescent Antibody Technique , Genetic Vectors/genetics , HIV Infections/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Macrophages/cytology , Monocytes/cytology , Mutagenesis/genetics , Transposases/metabolism
13.
Sci Rep ; 4: 3625, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24406502

ABSTRACT

Skeletal muscle (SM) regeneration after injury is impaired by excessive inflammation. Particularly, the inflammatory cytokine tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a potent inducer of skeletal muscle wasting and fibrosis. In this study we investigated the role of Nrf2, a major regulator of oxidative stress defence, in SM ischemia/reperfusion (I/R) injury and TWEAK induced atrophy. We explored the time-dependent expression of TWEAK after I/R in SM of Nrf2-wildtype (WT) and knockout (KO) mice. Nrf2-KO mice expressed significant higher levels of TWEAK as compared to WT mice. Consequently, Nrf2-KO mice present an insufficient regeneration as compared to Nrf2-WT mice. Moreover, TWEAK stimulation activates Nrf2 in the mouse myoblast cell line C2C12. This Nrf2 activation inhibits TWEAK induced atrophy in C2C12 differentiated myotubes. In summary, we show that Nrf2 protects SM from TWEAK-induced cell death in vitro and that Nrf2-deficient mice therefore have poorer muscle regeneration.


Subject(s)
Muscle, Skeletal/pathology , Muscular Atrophy/prevention & control , NF-E2-Related Factor 2/physiology , Tumor Necrosis Factors/physiology , Animals , Apoptosis , Base Sequence , Cell Line , Cytokine TWEAK , DNA Primers , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction
14.
Stem Cells Transl Med ; 2(8): 558-66, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23847002

ABSTRACT

The discovery of induced pluripotent stem cells (iPSCs) holds great promise for regenerative medicine since it is possible to produce patient-specific pluripotent stem cells from affected individuals for potential autologous treatment. Using nonintegrating cytoplasmic Sendai viral vectors, we generated iPSCs efficiently from adult mobilized CD34⁺ and peripheral blood mononuclear cells. After 5-8 passages, the Sendai viral genome could not be detected by real-time quantitative reverse transcription-polymerase chain reaction. Using the spin embryoid body method, we showed that these blood cell-derived iPSCs could efficiently be differentiated into hematopoietic stem and progenitor cells without the need of coculture with either mouse or human stromal cells. We obtained up to 40% CD34⁺ of which ~25% were CD34⁺/CD43⁺ hematopoietic precursors that could readily be differentiated into mature blood cells. Our study demonstrated a reproducible protocol for reprogramming blood cells into transgene-free iPSCs by the Sendai viral vector method. Maintenance of the genomic integrity of iPSCs without integration of exogenous DNA should allow the development of therapeutic-grade stem cells for regenerative medicine.


Subject(s)
Blood Cells/cytology , Cellular Reprogramming , Genetic Vectors/metabolism , Induced Pluripotent Stem Cells/cytology , Sendai virus/genetics , Transcription Factors/metabolism , Adult , Animals , Antigens, CD34/metabolism , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Female , Gene Expression Profiling , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred NOD , Transgenes/genetics
15.
Cold Spring Harb Perspect Med ; 3(2): a011775, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23378598

ABSTRACT

The thalassemias are among the most common inherited diseases worldwide, affecting individuals originating from the Mediterranean area, Middle East, Transcaucasia, Central Asia, Indian subcontinent, and Southeast Asia. As the diseases require long-term care, prevention of the homozygous state constitutes a major armament in the management. This article discusses the major prevention programs that are set up in many countries in Europe, Asia, and Australia, often drawing from the experience in Sardinia. These comprehensive programs involve carrier detections, molecular diagnostics, genetic counseling, and prenatal diagnosis. Variability of clinical severity can be attributable to interactions with α-thalassemia and mutations that increase fetal productions. Special methods that are currently quite expensive and not widely applicable are preimplantation and preconception diagnosis. The recent successful studies of fetal DNA in maternal plasma may allow future prenatal diagnosis that is noninvasive for the fetus.


Subject(s)
Thalassemia/prevention & control , Cost of Illness , Female , Forecasting , Genetic Counseling/methods , Genetic Testing/methods , Global Health , Health Education/methods , Health Promotion/methods , Humans , Molecular Diagnostic Techniques/methods , Phenotype , Preconception Care/methods , Pregnancy , Prenatal Diagnosis/methods , Prognosis , Thalassemia/epidemiology
16.
Proc Natl Acad Sci U S A ; 108(5): 2064-9, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21245320

ABSTRACT

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.


Subject(s)
Angiopoietin-1/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-1/physiology , Animals , Cell Proliferation , Coronary Vessels/growth & development , Swine , Vascular Endothelial Growth Factor A/physiology
17.
Ann Rheum Dis ; 70(5): 844-50, 2011 May.
Article in English | MEDLINE | ID: mdl-21173018

ABSTRACT

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.


Subject(s)
Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , NF-E2-Related Factor 2/physiology , Oxidative Stress/physiology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/metabolism , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/physiopathology , Cartilage, Articular/pathology , Cartilage, Articular/physiopathology , Fractures, Spontaneous/etiology , Fractures, Spontaneous/pathology , Gene Expression , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/deficiency , RNA, Messenger/genetics , Thioredoxins/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
18.
J Biol Chem ; 286(6): 4493-9, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21127061

ABSTRACT

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.


Subject(s)
Antioxidants , Gene Expression Regulation , Interleukin-6/biosynthesis , NF-E2-Related Factor 2/metabolism , Response Elements , Animals , Disease Models, Animal , Hep G2 Cells , Hepatitis/genetics , Hepatitis/metabolism , Humans , Interleukin-6/genetics , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Point Mutation , Transcription, Genetic
19.
Proc Natl Acad Sci U S A ; 107(45): 19467-72, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974949

ABSTRACT

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.


Subject(s)
Bacteriophages/enzymology , Induced Pluripotent Stem Cells/cytology , Integrases/genetics , Amniotic Fluid/cytology , Animals , Cell Culture Techniques , Cellular Reprogramming , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...