Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Depress Anxiety ; 34(1): 68-78, 2017 01.
Article in English | MEDLINE | ID: mdl-27557481

ABSTRACT

BACKGROUND: Somatostatin (SST) is a neuropeptide expressed in a subtype of gamma-aminobutyric acid (GABA) interneurons that target the dendrites of pyramidal neurons. We previously reported reduced levels of SST gene and protein expression in the postmortem amygdala of subjects with major depressive disorder (MDD). This reduction was specific to female subjects with MDD. METHODS: Here, we used in situ hybridization to examine the regional and cellular patterns of reductions in SST expression in a cohort of female MDD subjects with known SST deficits in the amygdala (N = 10/group). RESULTS: We report a significant reduction in the density of SST-labeled neurons in the lateral, basolateral, and basomedial nuclei of the amygdala of MDD subjects compared to controls. SST mRNA levels per neuron did not differ between MDD and control subjects in the lateral or basolateral nuclei, but were lower in the basomedial nucleus. There was no difference in cross-sectional density of total cells. CONCLUSIONS: In summary, we report an MDD-related reduction in the density of detectable SST-positive neurons across several nuclei in the amygdala, with a reduction in SST mRNA per cell restricted to the basomedial nucleus. In the absence of changes in total cell density, these results suggest the possibility of a change in SST cell phenotype rather than cell death in the amygdala of female MDD subjects.


Subject(s)
Amygdala/metabolism , Amygdala/pathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Somatostatin/metabolism , Adult , Autopsy , Cell Count , Female , Humans , Middle Aged
2.
Nucleic Acids Res ; 43(18): 8638-50, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26338776

ABSTRACT

With many safety and technical limitations partly mitigated through chemical modifications, antisense oligonucleotides (ASOs) are gaining recognition as therapeutic entities. The increase in potency realized by 'third generation chemistries' may, however, simultaneously increase affinity to unintended targets with partial sequence complementarity. However, putative hybridization-dependent off-target effects (OTEs), a risk historically regarded as low, are not being adequately investigated. Here we show an unexpectedly high OTEs confirmation rate during screening of fully phosphorothioated (PS)-LNA gapmer ASOs designed against the BACH1 transcript. We demonstrate in vitro mRNA and protein knockdown of off-targets with a wide range of mismatch (MM) and gap patterns. Furthermore, with RNase H1 activity residing within the nucleus, hybridization predicted against intronic regions of pre-mRNAs was tested and confirmed. This dramatically increased ASO-binding landscape together with relatively high potency of such interactions translates into a considerable safety concern. We show here that with base pairing-driven target recognition it is possible to predict the putative off-targets and address the liability during lead design and optimization phases. Moreover, in silico analysis performed against both primary as well as spliced transcripts will be invaluable in elucidating the mechanism behind the hepatoxicity observed with some LNA-modified gapmers.


Subject(s)
Exons , Gene Knockdown Techniques , Introns , Oligonucleotides, Antisense , Base Pair Mismatch , Cells, Cultured , Computer Simulation , Gene Silencing , Humans , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/therapeutic use , Ribonuclease H/metabolism
3.
Am J Geriatr Psychiatry ; 21(5): 450-60, 2013 May.
Article in English | MEDLINE | ID: mdl-23570888

ABSTRACT

OBJECTIVES: Brain molecular aging, the pervasive and consistent transcriptome changes associated with normal brain aging, appears to overlap with disease pathways and may be anticipated in neurodegenerative and neuropsychiatric diseases, including major depressive disorder (MDD). Here, we characterize the global interaction of MDD-related gene changes with age, starting from our previous report of downregulated brain-derived neurotrophic factor (BDNF) and BDNF-dependent genes in the amygdala of women with MDD. METHODS: A large-scale gene expression data set in the amygdala from a postmortem cohort of 21 women with MDD and 21 age-matched controls (age range: 16-74 years) was analyzed for correlations of gene transcript changes with age, in the presence or absence of a diagnosis of MDD. RESULTS: 1) The age-related decrease in BDNF transcripts observed in control subjects corresponds with further age-related decreases in BDNF and BDNF-dependent gene expression in MDD subjects; 2) most MDD-related genes are frequently age-regulated in both MDD and control subjects; 3) the effects of MDD and age are positively correlated; 4) most genes that are age-dependent in control subjects display greater age effects in MDD subjects; and 5) the increased prevalence of age effects in MDD corresponds to similar trends in controls, rather than representing de novo age effects. CONCLUSIONS: MDD strongly associates with robust and anticipated gene expression changes that occur during normal aging of the brain, suggesting that an older molecular age of the brain represents an early biological event and/or a marker of risk for subsequent onset of MDD symptoms.


Subject(s)
Aging/genetics , Aging/psychology , Amygdala/metabolism , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Genetic Predisposition to Disease/genetics , Adolescent , Adult , Aged , Biomarkers/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Case-Control Studies , Depressive Disorder, Major/psychology , Female , Humans , Middle Aged
4.
IUBMB Life ; 63(11): 979-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22002928

ABSTRACT

Glycogen storage disease type II or Pompe disease (GSD II, MIM 232300) is a rare inherited metabolic myopathy caused by a deficiency of lysosomal acid α-glucosidase or acid maltase (GAA; EC 3.2.1.20), resulting in a massive lysosomal glycogen accumulation in cardiac and skeletal muscles. Affected individuals exhibit either severe hypotonia associated with hypertrophic cardiomyopathy (infantile forms) or progressive muscle weakness (late-onset forms). Even if enzyme replacement therapy has recently become a standard treatment, it suffers from several limitations. This review will present the main results of enzyme replacement therapy and the recent findings concerning alternative treatments for Pompe disease, such as gene therapy, enzyme enhancement therapy, and substrate reduction therapy.


Subject(s)
Glycogen Storage Disease Type II/therapy , Animals , Drug Carriers , Enzyme Activators/therapeutic use , Enzyme Replacement Therapy/adverse effects , Enzyme Replacement Therapy/methods , Genetic Therapy , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/genetics , Humans , Immune Tolerance , Liver/drug effects , Liver/physiopathology , Muscles/drug effects , Muscles/physiopathology , alpha-Glucosidases/genetics , alpha-Glucosidases/immunology , alpha-Glucosidases/therapeutic use
5.
Methods Mol Biol ; 709: 223-35, 2011.
Article in English | MEDLINE | ID: mdl-21194031

ABSTRACT

RNA interference has emerged as a powerful technique to down-regulate gene expression. The lentiviral vector-mediated expression of small hairpin RNAs (shRNAs) from polymerase III promoters allows permanent down-regulation of a specific gene in a wide range of cell types both in vitro and in vivo. In this chapter, we describe a method permitting the expression of shRNA from lentiviral vectors in primary murine myogenic cells. We designed shRNAs targeted to the muscular glycogen synthase isoform (shGYS1), a highly regulated enzyme responsible for glycogen synthesis, in order to modulate the muscle glycogen biosynthetic pathway and to improve the phenotype in primary myogenic cells from a murine model of glycogen storage disease type II (GSDII). This method based on shRNA-mediated down-regulation could be applied to other muscular disorders to evaluate new therapeutic options.


Subject(s)
Genetic Vectors , Glycogen Synthase/genetics , Lentivirus/genetics , Myoblasts , RNA Interference , RNA, Small Interfering/genetics , Animals , Cells, Cultured , Gene Expression , Glycogen/biosynthesis , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Membrane Glycoproteins/genetics , Mice , Muscular Diseases/therapy , RNA, Small Interfering/therapeutic use , Viral Envelope Proteins/genetics
6.
Hum Mol Genet ; 19(4): 684-96, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19959526

ABSTRACT

Glycogen storage disease type II (GSDII) or Pompe disease is an autosomal recessive disorder caused by acid alpha-glucosidase (GAA) deficiency, leading to lysosomal glycogen accumulation. Affected individuals store glycogen mainly in cardiac and skeletal muscle tissues resulting in fatal hypertrophic cardiomyopathy and respiratory failure in the most severe infantile form. Enzyme replacement therapy has already proved some efficacy, but results remain variable especially in skeletal muscle. Substrate reduction therapy was successfully used to improve the phenotype in several lysosomal storage disorders. We have recently demonstrated that shRNA-mediated reduction of glycogen synthesis led to a significant reduction of glycogen accumulation in skeletal muscle of GSDII mice. In this paper, we analyzed the effect of a complete genetic elimination of glycogen synthesis in the same GSDII model. GAA and glycogen synthase 1 (GYS1) KO mice were inter-crossed to generate a new double-KO model. GAA/GYS1-KO mice exhibited a profound reduction of the amount of glycogen in the heart and skeletal muscles, a significant decrease in lysosomal swelling and autophagic build-up as well as a complete correction of cardiomegaly. In addition, the abnormalities in glucose metabolism and insulin tolerance observed in the GSDII model were corrected in double-KO mice. Muscle atrophy observed in 11-month-old GSDII mice was less pronounced in GAA/GYS1-KO mice, resulting in improved exercise capacity. These data demonstrate that long-term elimination of muscle glycogen synthesis leads to a significant improvement of structural, metabolic and functional defects in GSDII mice and offers a new perspective for the treatment of Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/physiopathology , Glycogen/biosynthesis , Muscle, Skeletal/physiopathology , Animals , Disease Models, Animal , Female , Glucose/metabolism , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/therapy , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Humans , Lysosomes/genetics , Lysosomes/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
7.
Neurobiol Dis ; 37(3): 656-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005954

ABSTRACT

Sandhoff disease is an autosomal recessive lysosomal disorder due to mutations in the beta-hexosaminidase beta-chain gene, resulting in beta-hexosaminidases A (alphabeta) and B (betabeta) deficiency and GM2 ganglioside accumulation in the brain. In this study, our aim was to demonstrate that transduction of cerebral endothelial cells cultured in two-chamber culture inserts with a lentiviral vector encoding the hexosaminidases alpha and beta chains could induce a vectorial secretion of hexosaminidases. Therefore, the human cerebral endothelial cell line hCMEC/D3 was infected with the bicistronic vector from the apical compartment, and beta-hexosaminidase activity was measured in transduced cells and in deficient fibroblasts co-cultured in the basal (i.e. brain) compartment. Induced beta-hexosaminidase secretion by transduced hCMEC/D3 cells was sufficient to allow for a 70-90% restoration of beta-hexosaminidase activity in deficient fibroblasts. On the basis of these in vitro data, we propose that brain endothelium be considered as a novel therapeutic target in Sandhoff disease.


Subject(s)
Endothelial Cells/enzymology , Genetic Therapy/methods , Sandhoff Disease/enzymology , Sandhoff Disease/therapy , Transduction, Genetic/methods , beta-N-Acetylhexosaminidases/metabolism , Cell Line, Transformed , Cerebral Arteries/cytology , Cerebral Arteries/enzymology , Cerebrum/blood supply , Cerebrum/enzymology , Cerebrum/physiopathology , Coculture Techniques , Diffusion Chambers, Culture , Endothelial Cells/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , G(M2) Ganglioside/metabolism , Genetic Vectors/pharmacology , Genetic Vectors/therapeutic use , Humans , Lentivirus/genetics , Sandhoff Disease/genetics , beta-N-Acetylhexosaminidases/genetics
8.
Biochem Biophys Res Commun ; 388(2): 333-8, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19665008

ABSTRACT

Glycogen storage disease type II (GSDII) is an autosomal recessive disorder caused by defects in the acid alpha-glucosidase (GAA) gene leading to lysosomal glycogen accumulation, mainly in cardiac and muscle tissues. In order to facilitate biological investigation on this disease and to avoid time-consuming direct cell isolation and culture, we have established murine myogenic GSDII cell lines. Lentiviral/retroviral expression of SV40 T antigen, Bmi-1 or cyclin-dependent kinase 4 (CDK4) genes was used to induce the immortalization of primary satellite cells from GSDII mice. The resulting immortalized myoblasts exhibit phenotypic characteristics of their parental cells, including profound GAA deficiency, glycogen accumulation and the ability to fully differentiate into myotubes when placed in proper culture conditions. These cell lines will constitute a powerful tool for both basic and applied studies focused on a better understanding of the pathophysiological mechanisms involved in GSDII and for assessing putative therapeutic strategies.


Subject(s)
Cell Line , Glycogen Storage Disease Type II/enzymology , Myoblasts/cytology , alpha-Glucosidases/genetics , Animals , Disease Models, Animal , Glycogen Storage Disease Type II/drug therapy , Lysosomes/enzymology , Mice , Mice, Knockout , Muscle Development , Myoblasts/enzymology , Myoblasts/physiology
9.
J Gene Med ; 11(4): 279-87, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19263466

ABSTRACT

BACKGROUND: Glycogen storage disease type II (GSDII) or Pompe disease is an inherited disease of glycogen metabolism caused by a lack of functional lysosomal acid alpha-glucosidase (GAA). Affected individuals store glycogen in lysosomes resulting in fatal hypertrophic cardiomyopathy and respiratory failure in the most severe form. Even if enzyme replacement therapy (ERT) has already proven some efficacy, its results remain heterogeneous in skeletal muscle, especially in cross reactive immunological material (CRIM)-negative patients. We investigated for the first time the use of hematopoietic stem cell (HSC) gene therapy in a murine model of GSDII. METHODS: Deficient HSC were transduced with a lentiviral vector expressing human GAA or enhanced green fluorescent protein (GFP) under the control of the retroviral MND promoter and transplanted into lethally irradiated GSDII mice. Animals were then subjected to an ERT protocol for 5 weeks and monitored for metabolic correction and GAA-induced immune reaction. RESULTS: GAA was expressed as a correctly processed protein, allowing a complete enzymatic correction in transduced deficient cells without toxicity. Seventeen weeks after transplantation, a partial restoration of the GAA enzymatic activity was observed in bone marrow and peripheral blood cells of GSDII mice, allowing a significant glycogen clearance in skeletal muscle. ERT induced a robust antibody response in GFP-transplanted mice, whereas no immune reaction could be detected in GAA-transplanted mice. CONCLUSIONS: Lentiviral vector-mediated HSC gene therapy leads to a partial metabolic correction and induces a tolerance to ERT in GSDII mice. This strategy could enhance the efficacy of ERT in CRIM-negative Pompe patients.


Subject(s)
Genetic Therapy , Glycogen Storage Disease Type II/therapy , Hematopoietic Stem Cells/metabolism , Immune Tolerance , alpha-Glucosidases/administration & dosage , Animals , Disease Models, Animal , Enzyme Therapy , Gene Transfer Techniques , Humans , Mice , Mice, Transgenic , Phenotype , Treatment Outcome , alpha-Glucosidases/genetics
10.
Hum Mol Genet ; 17(24): 3876-86, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18782850

ABSTRACT

Glycogen storage disease type II (GSDII) or Pompe disease is an autosomal recessive disorder caused by defects in the acid alpha-glucosidase gene, which leads to lysosomal glycogen accumulation and enlargement of the lysosomes mainly in cardiac and muscle tissues, resulting in fatal hypertrophic cardiomyopathy and respiratory failure in the most severely affected patients. Enzyme replacement therapy has already proven to be beneficial in this disease, but correction of pathology in skeletal muscle still remains a challenge. As substrate deprivation was successfully used to improve the phenotype in other lysosomal storage disorders, we explore here a novel therapeutic approach for GSDII based on a modulation of muscle glycogen synthesis. Short hairpin ribonucleic acids (shRNAs) targeted to the two major enzymes involved in glycogen synthesis, i.e. glycogenin (shGYG) and glycogen synthase (shGYS), were selected. C2C12 cells and primary myoblasts from GSDII mice were stably transduced with lentiviral vectors expressing both the shRNAs and the enhanced green fluorescent protein (EGFP) reporter gene. Efficient and specific inhibition of GYG and GYS was associated not only with a decrease in cytoplasmic and lysosomal glycogen accumulation in transduced cells, but also with a strong reduction in the lysosomal size, as demonstrated by confocal microscopy analysis. A single intramuscular injection of recombinant AAV-1 (adeno-associated virus-1) vectors expressing shGYS into newborn GSDII mice led to a significant reduction in glycogen accumulation, demonstrating the in vivo therapeutic efficiency. These data offer new perspectives for the treatment of GSDII and could be relevant to other muscle glycogenoses.


Subject(s)
Genetic Therapy , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Glycogen/biosynthesis , Glycogen/genetics , RNA Interference/physiology , Animals , Animals, Newborn , Cell Line , Dependovirus/genetics , Genetic Vectors/administration & dosage , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , Glycogen Storage Disease Type II/enzymology , Glycogen Synthase/antagonists & inhibitors , Glycogen Synthase/genetics , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , Humans , Mice , Mice, Knockout
11.
In Vitro Cell Dev Biol Anim ; 44(10): 397-406, 2008.
Article in English | MEDLINE | ID: mdl-18810562

ABSTRACT

Glycogen storage disease type II (GSDII) or Pompe disease is an inherited disease of glycogen metabolism caused by a lack of functional lysosomal acid alpha-glucosidase (GAA). Affected individuals store glycogen in lysosomes resulting in fatal hypertrophic cardiomyopathy and respiratory failure in the most severe form. We investigated for the first time the use of lentiviral vectors to correct the GSDII phenotype in human and murine GAA-deficient cells. Fibroblasts from infantile and adult GSDII patients were efficiently transduced by a GAA-expressing lentiviral vector placed under the control of the strong MND promoter, leading to a complete restoration of enzymatic activity. We also developed a muscle-specific lentiviral vector based on the synthetic C5-12 promoter and tested it on deficient myogenic satellite cells derived from a GSDII mouse model. GAA was expressed as a correctly processed protein allowing a complete enzymatic and metabolic correction in myoblasts and differentiated myotubes, as well as a significant mannose-6-phosphate (M6P)-dependent secretion reuptake by naive cells. Transduced cells showed lysosomal glycogen clearance, as demonstrated by electron microscopy. These results form the basis for a therapeutic approach of GSDII using lentiviral vector-mediated gene transfer into muscle stem cells.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Lentivirus/genetics , Muscles/metabolism , Animals , Cell Line , Fibroblasts/enzymology , Fibroblasts/pathology , Glycogen/metabolism , Humans , Lysosomes/metabolism , Mice , Myoblasts/enzymology , Myoblasts/pathology , Myoblasts/ultrastructure , Organ Specificity , Receptor, IGF Type 2/metabolism , Transduction, Genetic , Virus Inactivation , alpha-Glucosidases/genetics , alpha-Glucosidases/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...