Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Huntingtons Dis ; 13(1): 41-53, 2024.
Article in English | MEDLINE | ID: mdl-38427495

ABSTRACT

Background: Mutations in the Huntingtin (HTT) gene cause Huntington's disease (HD), a neurodegenerative disorder. As a scaffold protein, HTT is involved in numerous cellular functions, but its normal and pathogenic functions during human forebrain development are poorly understood. Objective: To investigate the developmental component of HD, with a specific emphasis on understanding the functions of wild-type and mutant HTT alleles during forebrain neuron development in individuals carrying HD mutations. Methods: We used CRISPR/Cas9 gene-editing technology to disrupt the ATG region of the HTT gene via non-homologous end joining to produce mono- or biallelic HTT knock-out human induced pluripotent stem cell (iPSC) clones. Results: We showed that the loss of wild-type, mutant, or both HTT isoforms does not affect the pluripotency of iPSCs or their transition into neural cells. However, we observed that HTT loss causes division impairments in forebrain neuro-epithelial cells and alters maturation of striatal projection neurons (SPNs) particularly in the acquisition of DARPP32 expression, a key functional marker of SPNs. Finally, young post-mitotic neurons derived from HTT-/- human iPSCs display cellular dysfunctions observed in adult HD neurons. Conclusions: We described a novel collection of isogenic clones with mono- and biallelic HTT inactivation that complement existing HD-hiPSC isogenic series to explore HTT functions and test therapeutic strategies in particular HTT-lowering drugs. Characterizing neural and neuronal derivatives from human iPSCs of this collection, we show evidence that HTT loss or mutation has impacts on neuro-epithelial and striatal neurons maturation, and on basal DNA damage and BDNF axonal transport in post-mitotic neurons.


Subject(s)
Huntington Disease , Induced Pluripotent Stem Cells , Adult , Humans , Induced Pluripotent Stem Cells/metabolism , Huntington Disease/metabolism , Neurons/metabolism , Corpus Striatum/metabolism , Alleles , Huntingtin Protein/genetics , Huntingtin Protein/metabolism
2.
Stem Cell Res Ther ; 14(1): 110, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37106426

ABSTRACT

BACKGROUND: CRISPR/Cas9 editing systems are currently used to generate mutations in a particular gene to mimic a genetic disorder in vitro. Such "disease in a dish" models based on human pluripotent stem cells (hPSCs) offer the opportunity to have access to virtually all cell types of the human body. However, the generation of mutated hPSCs remains fastidious. Current CRISPR/Cas9 editing approaches lead to a mixed cell population containing simultaneously non-edited and a variety of edited cells. These edited hPSCs need therefore to be isolated through manual dilution cloning, which is time-consuming, labor intensive and tedious. METHODS: Following CRISPR/Cas9 edition, we obtained a mixed cell population with various edited cells. We then used a semi-automated robotic platform to isolate single cell-derived clones. RESULTS: We optimized CRISPR/Cas9 editing to knock out a representative gene and developed a semi-automated method for the clonal isolation of edited hPSCs. This method is faster and more reliable than current manual approaches. CONCLUSIONS: This novel method of hPSC clonal isolation will greatly improve and upscale the generation of edited hPSCs required for downstream applications including disease modeling and drug screening.


Subject(s)
CRISPR-Cas Systems , Pluripotent Stem Cells , Humans , CRISPR-Cas Systems/genetics , Gene Editing/methods , Pluripotent Stem Cells/metabolism , Mutation , Clone Cells
3.
Am J Hum Genet ; 108(11): 2171-2185, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34699745

ABSTRACT

Recent studies indicate that neurodegenerative processes that appear during childhood and adolescence in individuals with Wolfram syndrome (WS) occur in addition to early brain development alteration, which is clinically silent. Underlying pathological mechanisms are still unknown. We have used induced pluripotent stem cell-derived neural cells from individuals affected by WS in order to reveal their phenotypic and molecular correlates. We have observed that a subpopulation of Wolfram neurons displayed aberrant neurite outgrowth associated with altered expression of axon guidance genes. Selective inhibition of the ATF6α arm of the unfolded protein response prevented the altered phenotype, although acute endoplasmic reticulum stress response-which is activated in late Wolfram degenerative processes-was not detected. Among the drugs currently tried in individuals with WS, valproic acid was the one that prevented the pathological phenotypes. These results suggest that early defects in axon guidance may contribute to the loss of neurons in individuals with WS.


Subject(s)
Age of Onset , Induced Pluripotent Stem Cells/cytology , Neurites , Neurons/cytology , Wolfram Syndrome/pathology , CRISPR-Cas Systems , Case-Control Studies , Endoplasmic Reticulum Stress , Gene Expression Regulation , Humans , Neurites/drug effects , Valproic Acid/pharmacology , Wolfram Syndrome/genetics
4.
Hum Mol Genet ; 31(1): 41-56, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34312665

ABSTRACT

Alternative splicing has emerged as a fundamental mechanism for the spatiotemporal control of development. A better understanding of how this mechanism is regulated has the potential not only to elucidate fundamental biological principles, but also to decipher pathological mechanisms implicated in diseases where normal splicing networks are misregulated. Here, we took advantage of human pluripotent stem cells to decipher during human myogenesis the role of muscleblind-like (MBNL) proteins, a family of tissue-specific splicing regulators whose loss of function is associated with myotonic dystrophy type 1 (DM1), an inherited neuromuscular disease. Thanks to the CRISPR/Cas9 technology, we generated human-induced pluripotent stem cells (hiPSCs) depleted in MBNL proteins and evaluated the consequences of their losses on the generation of skeletal muscle cells. Our results suggested that MBNL proteins are required for the late myogenic maturation. In addition, loss of MBNL1 and MBNL2 recapitulated the main features of DM1 observed in hiPSC-derived skeletal muscle cells. Comparative transcriptomic analyses also revealed the muscle-related processes regulated by these proteins that are commonly misregulated in DM1. Together, our study reveals the temporal requirement of MBNL proteins in human myogenesis and should facilitate the identification of new therapeutic strategies capable to cope with the loss of function of these MBNL proteins.


Subject(s)
Induced Pluripotent Stem Cells , Myotonic Dystrophy , Alternative Splicing , Gene Editing , Humans , Induced Pluripotent Stem Cells/metabolism , Muscle Development/genetics , Myotonic Dystrophy/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-31990683

ABSTRACT

Lesch-Nyhan disease (LND) is a rare monogenic disease caused by deficiency of the salvage pathway enzyme hypoxanthine-guanine phosphoribosyltransferase (HGPRT). LND is characterized by severe neuropsychiatric symptoms that currently cannot be treated. Predictive in vivo models are lacking for screening and evaluating candidate drugs because LND-associated neurological symptoms are not recapitulated in HGPRT-deficient animals. Here, we used human neural stem cells and neurons derived from induced pluripotent stem cells (iPSCs) of children affected with LND to identify neural phenotypes of interest associated with HGPRT deficiency to develop a target-agnostic-based drug screening system. We screened more than 3000 molecules and identified 6 pharmacological compounds, all possessing an adenosine moiety, that corrected HGPRT deficiency-associated neuronal phenotypes by promoting metabolism compensations in an HGPRT-independent manner. This included S-adenosylmethionine, a compound that had already been used as a compassionate approach to ease the neuropsychiatric symptoms in LND. Interestingly, these compounds compensate abnormal metabolism in a manner complementary to the gold standard allopurinol and can be provided to patients with LND via simple food supplementation. This experimental paradigm can be easily adapted to other metabolic disorders affecting normal brain development and functioning in the absence of a relevant animal model.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Lesch-Nyhan Syndrome/drug therapy , Lesch-Nyhan Syndrome/therapy , Neural Stem Cells/cytology , Allopurinol/therapeutic use , Animals , Case-Control Studies , Cell Differentiation , Disease Models, Animal , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Neural Stem Cells/enzymology , Phenotype
6.
Cells ; 8(12)2019 11 26.
Article in English | MEDLINE | ID: mdl-31779280

ABSTRACT

Substantial variations in differentiation properties have been reported among human pluripotent cell lines (hPSC), which could affect their utility and clinical safety. We characterized the variable osteogenic capacity observed between different human pluripotent stem cell lines. By focusing on the miRNA expression profile, we demonstrated that the osteogenic differentiation propensity of human pluripotent stem cell lines could be associated with the methylation status and the expression of miRNAs from the imprinted DLK1/DIO3 locus. More specifically, quantitative analysis of the expression of six different miRNAs of that locus prospectively identified human embryonic stem cells and human-induced pluripotent stem cells with differential osteogenic differentiation capacities. At the molecular and functional levels, we showed that these miRNAs modulated the expression of the activin receptor type 2B and the downstream signal transduction, which impacted osteogenesis. In conclusion, miRNAs of the imprinted DLK1/DIO3 locus appear to have both a predictive value and a functional impact in determining the osteogenic fate of human pluripotent stem cells.


Subject(s)
Calcium-Binding Proteins/genetics , Cell Differentiation/genetics , Iodide Peroxidase/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Osteogenesis/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Biomarkers , Calcium-Binding Proteins/metabolism , Cell Line , Gene Expression Regulation, Developmental , Genomic Imprinting , Humans , Immunophenotyping , Iodide Peroxidase/metabolism , Membrane Proteins/metabolism , Quantitative Trait Loci , RNA Interference
7.
Biochem Pharmacol ; 102: 45-63, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26707799

ABSTRACT

Cucurbitacins are cytotoxic triterpenoid sterols isolated from plants. One of their earliest cellular effect is the aggregation of actin associated with blockage of cell migration and division that eventually lead to apoptosis. We unravel here that cucurbitacin I actually induces the co-aggregation of actin with phospho-myosin II. This co-aggregation most probably results from the stimulation of the Rho/ROCK pathway and the direct inhibition of the LIMKinase. We further provide data that suggest that the formation of these co-aggregates is independent of a putative pro-oxidant status of cucurbitacin I. The results help to understand the impact of cucurbitacins on signal transduction and actin dynamics and open novel perspectives to use it as drug candidates for cancer research.


Subject(s)
Actins/metabolism , Lim Kinases/antagonists & inhibitors , Lim Kinases/metabolism , Myosin Type II/metabolism , Triterpenes/pharmacology , rho-Associated Kinases/metabolism , Actins/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Fosfomycin/chemistry , Fosfomycin/metabolism , HeLa Cells , Humans , Myosin Type II/chemistry , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Seeds , Signal Transduction/drug effects , Signal Transduction/physiology , Triterpenes/chemistry , Triterpenes/isolation & purification , rho-Associated Kinases/chemistry
8.
Hum Mol Genet ; 22(25): 5188-98, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-23922231

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an RNA-mediated disorder caused by a non-coding CTG repeat expansion that, in particular, provokes functional alteration of CUG-binding proteins. As a consequence, several genes with misregulated alternative splicing have been linked to clinical symptoms. In our search for additional molecular mechanisms that would trigger functional defects in DM1, we took advantage of mutant gene-carrying human embryonic stem cell lines to identify differentially expressed genes. Among the different genes found to be misregulated by DM1 mutation, one strongly downregulated gene encodes a transcription factor, ZNF37A. In this paper, we show that this defect in expression, which derives from a loss of RNA stability, is controlled by the RNA-binding protein, CUGBP1, and is associated with impaired myogenesis-a functional defect reminiscent of that observed in DM1. Loss of the ZNF37A protein results in changes in the expression of the subunit α1 of the receptor for the interleukin 13. This suggests that the pathological molecular mechanisms linking ZNF37A and myogenesis may involve the signaling pathway that is known to promote myoblast recruitment during development and regeneration.


Subject(s)
Alternative Splicing/genetics , Kruppel-Like Transcription Factors/genetics , Muscle Development/genetics , Myotonic Dystrophy/genetics , Trinucleotide Repeat Expansion/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Embryonic Stem Cells , Humans , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/metabolism , Mutation , Myotonic Dystrophy/physiopathology , Signal Transduction/genetics
9.
Stem Cells ; 31(9): 1816-28, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712629

ABSTRACT

Decreased expression of neuronal genes such as brain-derived neurotrophic factor (BDNF) is associated with several neurological disorders. One molecular mechanism associated with Huntington disease (HD) is a discrete increase in the nuclear activity of the transcriptional repressor REST/NRSF binding to repressor element-1 (RE1) sequences. High-throughput screening of a library of 6,984 compounds with luciferase-assay measuring REST activity in neural derivatives of human embryonic stem cells led to identify two benzoimidazole-5-carboxamide derivatives that inhibited REST silencing in a RE1-dependent manner. The most potent compound, X5050, targeted REST degradation, but neither REST expression, RNA splicing nor binding to RE1 sequence. Differential transcriptomic analysis revealed the upregulation of neuronal genes targeted by REST in wild-type neural cells treated with X5050. This activity was confirmed in neural cells produced from human induced pluripotent stem cells derived from a HD patient. Acute intraventricular delivery of X5050 increased the expressions of BDNF and several other REST-regulated genes in the prefrontal cortex of mice with quinolinate-induced striatal lesions. This study demonstrates that the use of pluripotent stem cell derivatives can represent a crucial step toward the identification of pharmacological compounds with therapeutic potential in neurological affections involving decreased expression of neuronal genes associated to increased REST activity, such as Huntington disease.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , High-Throughput Screening Assays/methods , Neural Stem Cells/metabolism , Neurons/metabolism , Repressor Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Cell Line , Disease Models, Animal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Genes, Reporter , Humans , Huntington Disease/pathology , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurons/drug effects , Repressor Proteins/metabolism , Transcriptome/drug effects , Transcriptome/genetics
10.
Cell Transplant ; 21(12): 2587-602, 2012.
Article in English | MEDLINE | ID: mdl-22889472

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are a most appealing source for cell replacement therapy in acute brain lesions. We evaluated the potential of hiPSC therapy in stroke by transplanting hiPSC-derived neural progenitor cells (NPCs) into the postischemic striatum. Grafts received host tyrosine hydroxylase-positive afferents and contained developing interneurons and homotopic GABAergic medium spiny neurons that, with time, sent axons to the host substantia nigra. Grafting reversed stroke-induced somatosensory and motor deficits. Grafting also protected the host substantia nigra from the atrophy that follows disruption of reciprocal striatonigral connections. Graft innervation by tyrosine hydoxylase fibers, substantia nigra protection, and somatosensory functional recovery were early events, temporally dissociated from the slow maturation of GABAergic neurons in the grafts and innervation of substantia nigra. This suggests that grafted hiPSC-NPCs initially exert trophic effects on host brain structures, which precede integration and potential pathway reconstruction. We believe that transplantation of NPCs derived from hiPSCs can provide useful interventions to limit the functional consequences of stroke through both neuroprotective effects and reconstruction of impaired pathways.


Subject(s)
Brain/pathology , Induced Pluripotent Stem Cells/cytology , Stroke/therapy , Animals , Brain/metabolism , Cell Differentiation , Cell Line , Embryoid Bodies/pathology , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Humans , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Teratoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism
11.
In Vitro Cell Dev Biol Anim ; 46(3-4): 376-85, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20217271

ABSTRACT

Pre-implantation genetic diagnosis allows the characterisation of embryos that carry a gene responsible for a severe monogenic disease and to transfer to the mother's uterus only the unaffected one(s). The genetically affected embryos can be used to establish human embryonic stem cell (hESC) lines. We are currently establishing a cell bank of ESC lines carrying specific disease-causing mutant genes. These cell lines are available to the scientific community. For this purpose, we have designed a technique that requires only minimal manipulation of the embryos. At the blastocyst stage, we just removed the zona pellucida before seeding the embryo as a whole on a layer of feeder cells. This approach gave a good success rate (>20%), whatever the quality of the embryos, and allowed us to derive 11 new hESC lines, representing seven different pathologies. Full phenotypic validation of the cell lines according to ISCI guidelines confirmed their pluripotent nature, as they were positive for hESC markers and able to differentiate in vitro in all three germ layers derivatives. Nine out of 11 stem cell lines had normal karyotypes. Our results indicate that inner cell mass isolation is not mandatory for hESC derivation and that minimal manipulation of embryos can lead to high success rate.


Subject(s)
Blastocyst/cytology , Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Preimplantation Diagnosis/methods , Animals , Antigens, Surface/metabolism , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Line , Female , Gene Expression Regulation, Developmental , Humans , Karyotyping , Male , Mice , Pedigree , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Tissue Eng Part C Methods ; 14(4): 289-98, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18710335

ABSTRACT

In parallel to the active search for therapeutic and industrial applications of human embryonic stem cells (hESCs), designing automated means of producing those cells is a timely goal. Slow-turning lateral vessels (STLVs) with low shear stress have shown promise for expanding the cells at the embryoid body stage. We have improved this technology by developing two complementary systems, allowing continuous optimization of the culture conditions. First, perfused STLV bioreactors were set up, to provide continuous delivery of culture medium to the cells growing in the rotating chamber. This allowed the external control of the culture medium, and consequently optimized oxygenation, pH, nutrient supply, and waste elimination. Second, a dialysis chamber was adapted. This led to a further enhanced controlled environment and a decrease in the quantity of adjunct products (e.g., growth factors) necessary to the cells inside the bioreactor chamber. hESC aggregation and initial differentiation-taking neural induction as an example-were compared between the perfused and dialyzed STLV system and static cultures. Perfused and dialyzed STLV bioreactors promoted formation of embryoid bodies that were differentiated more rapidly and were homogeneously synchronized in a statistically significant manner.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Stem Cells/cytology , Stem Cells/physiology , Tissue Engineering/instrumentation , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Cell Survival/physiology , Embryonic Induction/physiology , Equipment Design , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Neurons/cytology , Tissue Engineering/methods
13.
J Biol Chem ; 280(19): 18881-90, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15728587

ABSTRACT

We have recently demonstrated that in human heart, beta2-adrenergic receptors (beta2-ARs) are biochemically coupled not only to the classical adenylyl cyclase (AC) pathway but also to the cytosolic phospholipase A2 (cPLA2) pathway (Pavoine, C., Behforouz, N., Gauthier, C., Le Gouvello, S., Roudot-Thoraval, F., Martin, C. R., Pawlak, A., Feral, C., Defer, N., Houel, R., Magne, S., Amadou, A., Loisance, D., Duvaldestin, P., and Pecker, F. (2003) Mol. Pharmacol. 64, 1117-1125). In this study, using Fura-2-loaded cardiomyocytes isolated from adult rats, we showed that stimulation of beta2-ARs triggered an increase in the amplitude of electrically stimulated [Ca2+]i transients and contractions. This effect was abolished with the PKA inhibitor, H89, but greatly enhanced upon addition of the selective cPLA2 inhibitor, AACOCF3. The beta2-AR/cPLA2 inhibitory pathway involved G(i) and MSK1. Potentiation of beta2-AR/AC/PKA-induced Ca2+ responses by AACOCF3 did not rely on the enhancement of AC activity but was associated with eNOS phosphorylation (Ser1177) and L-NAME-sensitive NO production. This was correlated with PKA-dependent phosphorylation of PLB (Ser16). The constraint exerted by the beta2-AR/cPLA2 pathway on the beta2-AR/AC/PKA-induced Ca2+ responses required integrity of caveolar structures and was impaired by Filipin III treatment. Immunoblot analyses demonstrated zinterol-induced translocation of cPLA and its cosedimentation with MSK1, eNOS, PLB, and sarcoplasmic reticulum Ca2+ pump (SERCA) 2a in a low density caveolin-3-enriched membrane fraction. This inferred the gathering of beta2-AR signaling effectors around caveolae/sarcoplasmic reticulum (SR) functional platforms. Taken together, these data highlight cPLA as a cardiac beta2-AR signaling pathway that limits beta2-AR/AC/PKA-induced Ca2+ responses in adult rat cardiomyocytes through the impairment of eNOS activation and PLB phosphorylation.


Subject(s)
Cytosol/enzymology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Phospholipases A/physiology , Receptors, Adrenergic, beta-2/metabolism , Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Caveolin 1 , Caveolin 3 , Caveolins/metabolism , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Ethanolamines/pharmacology , Immunoblotting , Immunohistochemistry , Indoles/pharmacology , Isoquinolines/pharmacology , Lysophospholipase/metabolism , Microscopy, Confocal , NG-Nitroarginine Methyl Ester/pharmacology , Pertussis Toxin/pharmacology , Phospholipases A/metabolism , Phospholipases A2 , Phosphorylation , Rats , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Serine/chemistry , Sulfonamides/pharmacology , Time Factors
14.
Circulation ; 110(14): 2003-9, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15451797

ABSTRACT

BACKGROUND: Studies in isolated cardiomyocytes showed that replenishment in cellular glutathione, achieved with the glutathione precursor N-acetylcysteine (NAC), abrogated deleterious effects of tumor necrosis factor-alpha (TNF-alpha). METHODS AND RESULTS: We examined the ability of NAC to limit the progression of cardiac injury in the rat model of hypertension, induced by the nitric oxide synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) (50 mg/kg per day SC) and high-salt diet (HS) (8% NaCl). Four-week HS/L-NAME administration induced hypertension (193+/-8 versus 122+/-4 mm Hg for low-salt diet [LS] group) and left ventricular (LV) dysfunction, revealed by echocardiography and characterized by decreased LV shortening fraction (38+/-2% versus 49+/-4% for LS group; P<0.05) and decreased LV posterior wall thickening (49+/-3% versus 70+/-4% for LS group; P<0.05). LV dysfunction worsened further after 6-week HS/L-NAME administration. Importantly, increase in serum TNF-alpha level was strongly correlated with shortening fraction decrease and cardiac glutathione depletion. NAC (75 mg/d) was given as a therapeutic treatment in a subgroup of HS/L-NAME animals during weeks 5 and 6 of HS/L-NAME administration. NAC treatment, which replenished cardiac glutathione, had no effect on hypertension but reduced LV remodeling and dysfunction, normalized serum TNF-alpha level, and limited activation of matrix metalloproteinases -2 and -9 and collagen deposition in LV tissues. CONCLUSIONS: These findings suggest that glutathione status determines the adverse effects of TNF-alpha in cardiac failure and that TNF-alpha antagonism may be achieved by glutathione supplementation.


Subject(s)
Acetylcysteine/therapeutic use , Antioxidants/therapeutic use , Glutathione/physiology , Hypertension/drug therapy , Tumor Necrosis Factor-alpha/analysis , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling/drug effects , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Collagen/analysis , Disease Progression , Drug Evaluation, Preclinical , Glutathione/deficiency , Heart Ventricles/chemistry , Hypertension/blood , Hypertension/chemically induced , Hypertension/complications , Male , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Myocardial Contraction , Myocardium/metabolism , NG-Nitroarginine Methyl Ester/toxicity , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sodium Chloride, Dietary/toxicity , Sphingomyelin Phosphodiesterase/metabolism , Ultrasonography , Ventricular Dysfunction, Left/blood , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology
15.
Circulation ; 109(3): 406-11, 2004 Jan 27.
Article in English | MEDLINE | ID: mdl-14732751

ABSTRACT

BACKGROUND: The negative effect of tumor necrosis factor-alpha (TNF-alpha) on heart contraction, which is mediated by sphingosine, is a major component in heart failure. Because the cellular level of glutathione may limit sphingosine production via the inhibition of the Mg-dependent neutral sphingomyelinase (N-SMase), we hypothesized that cardiac glutathione status might determine the negative contractile response to TNF-alpha. METHODS AND RESULTS: We examined the effects of TNF-alpha in isolated cardiomyocytes obtained from control rats or rats that were given the glutathione precursor N-acetylcysteine (NAC, 100 mg IP per animal). In cardiomyocytes obtained from control rats, 25 ng/mL TNF-alpha increased reactive oxygen species generation and N-SMase activity (500% and 34% over basal, respectively) and decreased the amplitude of [Ca(2+)](i) in response to electrical stimulation (22% below basal). NAC treatment increased cardiac glutathione content by 42%. In cardiomyocytes obtained from NAC-treated rats, 25 ng/mL TNF-alpha had no effect on reactive oxygen species production or N-SMase activity but increased the amplitude of [Ca(2+)](i) transients and contraction in response to electrical stimulation by 40% to 50% over basal after 20 minutes. This was associated with a hastened relaxation (20% reduction in t(1/2) compared with basal) and an increased phosphorylation of both Ser(16)- and Thr(17)-phospholamban residues (260% and 115% of maximal isoproterenol effect, respectively). CONCLUSIONS: It is concluded that cardiac glutathione status, by controlling N-SMase activation, determines the severity of the adverse effects of TNF-alpha on heart contraction. Glutathione supplementation may therefore provide therapeutic benefits for vulnerable hearts.


Subject(s)
Acetylcysteine/pharmacology , Calcium/metabolism , Glutathione/analogs & derivatives , Myocytes, Cardiac/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Glutathione/metabolism , Glutathione/pharmacology , Male , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor-alpha/toxicity
16.
Development ; 129(8): 2003-13, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11934866

ABSTRACT

In this study, we have characterized the early steps of hematopoiesis during embryonic stem cell differentiation. The immunophenotype of hematopoietic progenitor cells derived from murine embryonic stem cells was determined using a panel of monoclonal antibodies specific for hematopoietic differentiation antigens. Surprisingly, the CD41 antigen (alphaIIb integrin, platelet GPIIb), essentially considered to be restricted to megakaryocytes, was found on a large proportion of cells within embryoid bodies although very few megakaryocytes were detected. In clonogenic assays, more than 80% of all progenitors (megakaryocytic, granulo-macrophagic, erythroid and pluripotent) derived from embryoid bodies expressed the CD41 antigen. CD41 was the most reliable marker of early steps of hematopoiesis. However, CD41 remained a differentiation marker because some CD41(-) cells from embryoid bodies converted to CD41(+) hematopoietic progenitors, whereas the inverse switch was not observed. Immunoprecipitation and western blot analysis confirmed that CD41 was present in cells from embryoid bodies associated with CD61 (beta3 integrin, platelet GPIIIa) in a complex. Analysis of CD41 expression during ontogeny revealed that most yolk sac and aorta-gonad-mesonephros hematopoietic progenitor cells were also CD41(+), whereas only a minority of bone marrow and fetal liver hematopoietic progenitors expressed this antigen. Differences in CD34 expression were also observed: hematopoietic progenitor cells from embryoid bodies, yolk sac and aorta-gonad-mesonephros displayed variable levels of CD34, whereas more than 90% of fetal liver and bone marrow progenitor cells were CD34(+). Thus, these results demonstrate that expression of CD41 is associated with early stages of hematopoiesis and is highly regulated during hematopoietic development. Further studies concerning the adhesive properties of hematopoietic cells are required to assess the biological significance of these developmental changes.


Subject(s)
Blood Platelets/immunology , Hematopoietic Stem Cells/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/biosynthesis , Animals , Antigens, CD/analysis , Biomarkers , Blood Platelets/classification , Cell Line , Hematopoietic Stem Cells/classification , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Glycoprotein GPIIb-IIIa Complex/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...