Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Acta Neuropathol ; 144(5): 1005-1025, 2022 11.
Article in English | MEDLINE | ID: mdl-36074148

ABSTRACT

Myasthenia gravis is an autoimmune disorder defined by muscle weakness and fatigability associated with antibodies against proteins of the neuromuscular junction (NMJ). The most common autoantibody target is the acetylcholine receptor (AChR). Three mechanisms have been postulated by which autoantibodies might interfere with neurotransmission: direct antagonism of the receptor, complement-mediated destruction of the postsynaptic membrane, and enhanced internalization of the receptor. It is very likely that more than one of these mechanisms act in parallel. Dissecting the mechanisms of autoantibody-mediated pathology requires patient-derived, monoclonal antibodies. Using membrane antigen capture activated cell sorting (MACACS), we isolated AChR-specific B cells from patients with myasthenia gravis, and produced six recombinant antibodies. All AChR-specific antibodies were hypermutated, including isotypes IgG1, IgG3, and IgG4, and recognized different subunits of the AChR. Despite clear binding, none of the individual antibodies showed significant antagonism of the AChR measured in an in vitro neuromuscular synapse model, or AChR-dependent complement activation, and they did not induce myasthenic signs in vivo. However, combinations of antibodies induced strong complement activation in vitro, and severe weakness in a passive transfer myasthenia gravis rat model, associated with NMJ destruction and complement activation in muscle. The strongest complement activation was mediated by combinations of antibodies targeting disparate subunits of the AChR, and such combinations also induced the formation of large clusters of AChR on the surface of live cells in vitro. We propose that synergy between antibodies of different epitope specificities is a fundamental feature of this disease, and possibly a general feature of complement-mediated autoimmune diseases. The importance of synergistic interaction between antibodies targeting different subunits of the receptor can explain the well-known discrepancy between serum anti-AChR titers and clinical severity, and has implications for therapeutic strategies currently under investigation.


Subject(s)
Myasthenia Gravis , Animals , Antibodies, Monoclonal , Autoantibodies , Cluster Analysis , Complement Activation , Complement System Proteins , Epitopes , Immunoglobulin G/metabolism , Myasthenia Gravis/pathology , Rats , Receptors, Cholinergic , Receptors, Complement
2.
Stem Cells ; 34(2): 299-310, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26577380

ABSTRACT

The multilineage differentiation capacity of mouse and human embryonic stem (ES) cells offers a testing platform for small molecules that mediate mammalian lineage determination and cellular specialization. Here we report the identification of two small molecules which drives mouse 129 ES cell differentiation to skeletal muscle with high efficiency without any genetic modification. Mouse embryoid bodies (EBs) were used to screen a library of 1,000 small molecules to identify compounds capable of inducing high levels of Pax3 mRNA. Stimulation of EBs with SMIs (skeletal muscle inducer, SMI1 and SMI2) from the screen resulted in a high percentage of intensively twitching skeletal muscle fibers 3 weeks after induction. Gene expression profiling studies that were carried out for mode of actions analysis showed that SMIs activated genes regulated by the Wnt pathway and inhibited expression of Smad2/3 and Sonic Hedgehog (Shh) target genes. A combination of three small molecules known to modulate these three pathways acted similarly to the SMIs found here, driving ES cells from 129 as well as Balb/c and C57Bl/6 to skeletal muscle. Taken together, these data demonstrate that the SMI drives ES cells to skeletal muscle via concerted activation of the Wnt pathway, and inhibition of Smad2/3 signaling and Shh pathways. This provides important developmental biological information about skeletal muscle differentiation from embryonic stem cells and may lead to the development of new therapeutics for muscle disease.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Wnt Signaling Pathway , Animals , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Muscle Fibers, Fast-Twitch/cytology
3.
Front Neurosci ; 17: 1103437, 2023.
Article in English | MEDLINE | ID: mdl-37250404

ABSTRACT

Novel in vitro platforms based on human neurons are needed to improve early drug testing and address the stalling drug discovery in neurological disorders. Topologically controlled circuits of human induced pluripotent stem cell (iPSC)-derived neurons have the potential to become such a testing system. In this work, we build in vitro co-cultured circuits of human iPSC-derived neurons and rat primary glial cells using microfabricated polydimethylsiloxane (PDMS) structures on microelectrode arrays (MEAs). Our PDMS microstructures are designed in the shape of a stomach, which guides axons in one direction and thereby facilitates the unidirectional flow of information. Such circuits are created by seeding either dissociated cells or pre-aggregated spheroids at different neuron-to-glia ratios. Furthermore, an antifouling coating is developed to prevent axonal overgrowth in undesired locations of the microstructure. We assess the electrophysiological properties of different types of circuits over more than 50 days, including their stimulation-induced neural activity. Finally, we demonstrate the inhibitory effect of magnesium chloride on the electrical activity of our iPSC circuits as a proof-of-concept for screening of neuroactive compounds.

4.
Front Mol Neurosci ; 16: 1287510, 2023.
Article in English | MEDLINE | ID: mdl-38235149

ABSTRACT

Neuron-to-neuron transmission of aggregation-prone, misfolded proteins may potentially explain the spatiotemporal accumulation of pathological lesions in the brains of patients with neurodegenerative protein-misfolding diseases (PMDs). However, little is known about protein transmission from the central nervous system to the periphery, or how this propagation contributes to PMD pathology. To deepen our understanding of these processes, we established two functional neuromuscular systems derived from human iPSCs. One was suitable for long-term high-throughput live-cell imaging and the other was adapted to a microfluidic system assuring that connectivity between motor neurons and muscle cells was restricted to the neuromuscular junction. We show that the Huntington's disease (HD)-associated mutant HTT exon 1 protein (mHTTEx1) is transmitted from neurons to muscle cells across the human neuromuscular junction. We found that transmission is an active and dynamic process that starts before aggregate formation and is regulated by synaptic activity. We further found that transmitted mHTTEx1 causes HD-relevant pathology at both molecular and functional levels in human muscle cells, even in the presence of the ubiquitous expression of mHTTEx1. In conclusion, we have uncovered a causal link between mHTTEx1 synaptic transmission and HD pathology, highlighting the therapeutic potential of blocking toxic protein transmission in PMDs.

5.
Lab Chip ; 22(7): 1386-1403, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35253810

ABSTRACT

Bottom-up neuroscience, which consists of building and studying controlled networks of neurons in vitro, is a promising method to investigate information processing at the neuronal level. However, in vitro studies tend to use cells of animal origin rather than human neurons, leading to conclusions that might not be generalizable to humans and limiting the possibilities for relevant studies on neurological disorders. Here we present a method to build arrays of topologically controlled circuits of human induced pluripotent stem cell (iPSC)-derived neurons. The circuits consist of 4 to 50 neurons with well-defined connections, confined by microfabricated polydimethylsiloxane (PDMS) membranes. Such circuits were characterized using optical imaging and microelectrode arrays (MEAs), suggesting the formation of functional connections between the neurons of a circuit. Electrophysiology recordings were performed on circuits of human iPSC-derived neurons for at least 4.5 months. We believe that the capacity to build small and controlled circuits of human iPSC-derived neurons holds great promise to better understand the fundamental principles of information processing and storing in the brain.


Subject(s)
Induced Pluripotent Stem Cells , Animals , Electrophysiological Phenomena , Electrophysiology , Humans , Induced Pluripotent Stem Cells/physiology , Microelectrodes , Neurons/physiology
6.
Eur Heart J ; 30(1): 116-24, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18784063

ABSTRACT

AIMS: Duchenne muscular dystrophy (DMD) is a severe and still incurable disease, with heart failure as a leading cause of death. The identification of a disease-modifying therapy may require early-initiated and long-term administration, but such type of therapeutic trial is not evident in humans. We have performed such a trial of SNT-MC17/idebenone in the mdx mouse model of DMD, based on the drug's potential to improve mitochondrial respiratory chain function and reduce oxidative stress. METHODS AND RESULTS: In this study, 200 mg/kg bodyweight of either SNT-MC17/idebenone or placebo was given from age 4 weeks until 10 months in mdx and wild-type mice. All evaluators were blinded to mouse type and treatment groups. Idebenone treatment significantly corrected cardiac diastolic dysfunction and prevented mortality from cardiac pump failure induced by dobutamine stress testing in vivo, significantly reduced cardiac inflammation and fibrosis, and significantly improved voluntary running performance in mdx mice. CONCLUSION: We have identified a novel potential therapeutic strategy for human DMD, as SNT-MC17/idebenone was cardioprotective and improved exercise performance in the dystrophin-deficient mdx mouse. Our data also illustrate that the mdx mouse provides unique opportunities for long-term controlled prehuman therapeutic studies.


Subject(s)
Antioxidants/therapeutic use , Muscular Dystrophy, Animal/drug therapy , Ubiquinone/analogs & derivatives , Animals , Biomarkers/blood , Cardiotonic Agents , Diastole , Dobutamine , Echocardiography , Fibrosis , Male , Mice , Mice, Inbred mdx , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/physiopathology , Myocardium/pathology , Oxidative Stress , Physical Conditioning, Animal , Placebos , Single-Blind Method , Time Factors , Troponin I/blood , Ubiquinone/therapeutic use
7.
J Pharmacol Exp Ther ; 331(3): 787-95, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19759319

ABSTRACT

Laminin alpha2-deficient congenital muscular dystrophy, called MDC1A, is a rare, devastating genetic disease characterized by severe neonatal hypotonia ("floppy infant syndrome"), peripheral neuropathy, inability to stand or walk, respiratory distress, and premature death in early life. Transgenic overexpression of the apoptosis inhibitor protein BCL-2, or deletion of the proapoptotic Bax gene in a mouse model for MDC1A prolongs survival and mitigates pathology, indicating that apoptotic events are involved in the pathology. Here we demonstrate that the proapoptotic glyceraldehyde-3-phosphate dehydrogenase (GAPDH)-Siah1-CBP/p300-p53 pathway is activated in a mouse model for MDC1A. Moreover, we show that omigapil, which inhibits GAPDH-Siah1-mediated apoptosis, ameliorates several pathological hallmarks in the MDC1A mouse model. Specifically, we demonstrate that treatment with omigapil inhibits apoptosis in muscle, reduces body weight loss and skeletal deformation, increases locomotive activity, and protects from early mortality. These data qualify omigapil, which is in late phase of clinical development for human use, as a drug candidate for the treatment of MDC1A.


Subject(s)
Apoptosis/drug effects , Laminin/deficiency , Muscle, Skeletal/drug effects , Muscular Dystrophy, Animal/drug therapy , Oxepins/therapeutic use , Animals , Body Weight/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Laminin/genetics , Mice , Mice, Knockout , Motor Activity/drug effects , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Nuclear Proteins/metabolism , Oxepins/administration & dosage , Oxepins/pharmacology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , p300-CBP Transcription Factors/metabolism
8.
FASEB J ; 22(12): 4190-200, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18728218

ABSTRACT

Dystrophin deficiency is the underlying molecular cause of progressive muscle weakness observed in Duchenne muscular dystrophy (DMD). Loss of functional dystrophin leads to elevated levels of intracellular Ca(2+), a key step in the cellular pathology of DMD. The cysteine protease calpain is activated in dystrophin-deficient muscle, and its inhibition is regarded as a potential therapeutic approach. In addition, previous work has shown that the ubiquitin-proteasome system also contributes to muscle protein breakdown in dystrophic muscle and, therefore, also qualifies as a potential target for therapeutic intervention in DMD. The relative contribution of calpain- and proteasome-mediated proteolysis induced by increased Ca(2+) levels was characterized in cultured muscle cells and revealed initial Ca(2+) influx-dependent calpain activity and subsequent Ca(2+)-independent activity of the ubiquitin-proteasome system. We then set out to optimize novel small-molecule inhibitors that inhibit both calpain as well as the 20S proteasome in a cellular system with impaired Ca(2+) homeostasis. On administration of such inhibitors to mdx mice, quantitative histological parameters improved significantly, in particular with compounds strongly inhibiting the 20S proteasome. To investigate the role of calpain inhibition without interfering with the ubiquitin-proteasome system, we crossed mdx mice with transgenic mice, overexpressing the endogenous calpain inhibitor calpastatin. Although our data show that proteolysis by calpain is strongly inhibited in the transgenic mdx mouse, this calpain inhibition did not ameliorate muscle histology. Our results indicate that inhibition of the proteasome rather than calpain is required for histological improvement of dystrophin-deficient muscle. In conclusion, we have identified novel proteasome inhibitors that qualify as potential candidates for pharmacological intervention in muscular dystrophy.


Subject(s)
Calcium/adverse effects , Calpain/antagonists & inhibitors , Muscles/pathology , Muscular Dystrophy, Duchenne/physiopathology , Protease Inhibitors/therapeutic use , Proteasome Inhibitors , Animals , Calcium-Binding Proteins/biosynthesis , Cells, Cultured , Humans , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscles/drug effects , Muscles/metabolism , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/pathology , Myoblasts/metabolism , Oligopeptides/pharmacology
9.
Cell Rep ; 29(6): 1539-1554.e7, 2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31693894

ABSTRACT

Age-related loss of skeletal muscle innervation by motor neurons leads to impaired neuromuscular function and is a well-established clinical phenomenon. However, the underlying pathogenesis remains unclear. Studying mice, we find that the number of motor units (MUs) can be maintained by counteracting neurotoxic microglia in the aged spinal cord. We observe that marked innervation changes, detected by motor unit number estimation (MUNE), occur prior to loss of muscle function in aged mice. This coincides with gene expression changes indicative of neuronal remodeling and microglial activation in aged spinal cord. Voluntary exercise prevents loss of MUs and reverses microglia activation. Depleting microglia by CSF1R inhibition also prevents the age-related decline in MUNE and neuromuscular junction disruption, implying a causal link. Our results suggest that age-related changes in spinal cord microglia contribute to neuromuscular decline in aged mice and demonstrate that removal of aged neurotoxic microglia can prevent or reverse MU loss.


Subject(s)
Aging/metabolism , Microglia/metabolism , Motor Neurons/metabolism , Physical Conditioning, Animal/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Aging/pathology , Animals , Cell Line , Databases, Genetic , Humans , Induced Pluripotent Stem Cells , Macrophages , Male , Mice , Mice, Inbred C57BL , Microglia/enzymology , Microglia/physiology , Motor Neurons/cytology , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Neuromuscular Junction/metabolism , Neuronal Plasticity/genetics , RNA-Seq , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Spinal Cord/enzymology , Spinal Cord/metabolism , Spinal Cord/physiopathology
10.
Sci Rep ; 8(1): 1799, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29379065

ABSTRACT

We generated induced pluripotent stem cells (iPSCs) from patient fibroblasts to yield cell lines containing varying degrees of heteroplasmy for a m.13514 A > G mtDNA point mutation (2 lines) and for a ~6 kb single, large scale mtDNA deletion (3 lines). Long term culture of the iPSCs containing a single, large-scale mtDNA deletion showed consistent increase in mtDNA deletion levels with time. Higher levels of mtDNA heteroplasmy correlated with increased respiratory deficiency. To determine what changes occurred in deletion level during differentiation, teratomas comprising all three embryonic germ layers were generated from low (20%) and intermediate heteroplasmy (55%) mtDNA deletion clones. Regardless of whether iPSCs harbouring low or intermediate mtDNA heteroplasmy were used, the final levels of heteroplasmy in all teratoma germ layers increased to a similar high level (>60%). Thus, during human stem cell division, cells not only tolerate high mtDNA deletion loads but seem to preferentially replicate deleted mtDNA genomes. This has implications for the involvement of mtDNA deletions in both disease and ageing.


Subject(s)
DNA, Mitochondrial/genetics , Sequence Deletion/genetics , Cell Differentiation/genetics , Cell Line , Clone Cells/metabolism , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mitochondria/genetics , Point Mutation/genetics
11.
Science ; 351(6278): 1199-203, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26847545

ABSTRACT

SH3 and multiple ankyrin repeat domains 3 (SHANK3) haploinsufficiency is causative for the neurological features of Phelan-McDermid syndrome (PMDS), including a high risk of autism spectrum disorder (ASD). We used unbiased, quantitative proteomics to identify changes in the phosphoproteome of Shank3-deficient neurons. Down-regulation of protein kinase B (PKB/Akt)-mammalian target of rapamycin complex 1 (mTORC1) signaling resulted from enhanced phosphorylation and activation of serine/threonine protein phosphatase 2A (PP2A) regulatory subunit, B56ß, due to increased steady-state levels of its kinase, Cdc2-like kinase 2 (CLK2). Pharmacological and genetic activation of Akt or inhibition of CLK2 relieved synaptic deficits in Shank3-deficient and PMDS patient-derived neurons. CLK2 inhibition also restored normal sociability in a Shank3-deficient mouse model. Our study thereby provides a novel mechanistic and potentially therapeutic understanding of deregulated signaling downstream of Shank3 deficiency.


Subject(s)
Autism Spectrum Disorder/drug therapy , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Amino Acid Sequence , Animals , Autism Spectrum Disorder/enzymology , Autism Spectrum Disorder/genetics , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 22/genetics , Disease Models, Animal , Down-Regulation , Gene Knockdown Techniques , Humans , Insulin-Like Growth Factor I/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Microfilament Proteins , Molecular Sequence Data , Multiprotein Complexes/metabolism , Neurons/enzymology , Phosphorylation , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteomics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
12.
Stem Cell Reports ; 7(6): 1059-1071, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27840045

ABSTRACT

In fragile X syndrome (FXS), CGG repeat expansion greater than 200 triplets is believed to trigger FMR1 gene silencing and disease etiology. However, FXS siblings have been identified with more than 200 CGGs, termed unmethylated full mutation (UFM) carriers, without gene silencing and disease symptoms. Here, we show that hypomethylation of the FMR1 promoter is maintained in induced pluripotent stem cells (iPSCs) derived from two UFM individuals. However, a subset of iPSC clones with large CGG expansions carries silenced FMR1. Furthermore, we demonstrate de novo silencing upon expansion of the CGG repeat size. FMR1 does not undergo silencing during neuronal differentiation of UFM iPSCs, and expression of large unmethylated CGG repeats has phenotypic consequences resulting in neurodegenerative features. Our data suggest that UFM individuals do not lack the cell-intrinsic ability to silence FMR1 and that inter-individual variability in the CGG repeat size required for silencing exists in the FXS population.


Subject(s)
DNA Methylation/genetics , Fragile X Mental Retardation Protein/genetics , Gene Silencing , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Neurons/metabolism , Trinucleotide Repeat Expansion/genetics , Cell Differentiation/genetics , Clone Cells , Epigenesis, Genetic , Female , Fragile X Syndrome/genetics , Genetic Loci , Humans , Induced Pluripotent Stem Cells/cytology , Male , Pedigree
13.
J Biomol Screen ; 20(9): 1101-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26024946

ABSTRACT

Fragile X syndrome (FXS) is the most common form of inherited mental retardation, and it is caused in most of cases by epigenetic silencing of the Fmr1 gene. Today, no specific therapy exists for FXS, and current treatments are only directed to improve behavioral symptoms. Neuronal progenitors derived from FXS patient induced pluripotent stem cells (iPSCs) represent a unique model to study the disease and develop assays for large-scale drug discovery screens since they conserve the Fmr1 gene silenced within the disease context. We have established a high-content imaging assay to run a large-scale phenotypic screen aimed to identify compounds that reactivate the silenced Fmr1 gene. A set of 50,000 compounds was tested, including modulators of several epigenetic targets. We describe an integrated drug discovery model comprising iPSC generation, culture scale-up, and quality control and screening with a very sensitive high-content imaging assay assisted by single-cell image analysis and multiparametric data analysis based on machine learning algorithms. The screening identified several compounds that induced a weak expression of fragile X mental retardation protein (FMRP) and thus sets the basis for further large-scale screens to find candidate drugs or targets tackling the underlying mechanism of FXS with potential for therapeutic intervention.


Subject(s)
Fragile X Syndrome/drug therapy , Gene Silencing/drug effects , Induced Pluripotent Stem Cells/drug effects , Neural Stem Cells/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Trinucleotide Repeats
14.
Neuromuscul Disord ; 13(9): 699-704, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14561491

ABSTRACT

Anecdotal reports of positive influence of certain traditional Chinese medicines on the progression of neuromuscular diseases in general and Duchenne muscular dystrophy (DMD) in particular has raised interest in patient support groups and clinical experts alike. However, clinical signs of steroid-specific side effects in patients treated with a particular form of Chinese medicine raised the concern that they may contain glucocorticoids, which in turn could also explain the mild beneficial effects seen in some of the patients. We have extracted and fractionated capsules containing pulverized Chinese medicine that had been used for the treatment of DMD patients and analyzed their content for glucocorticoid-like activity using promoter-reporter assays. We demonstrate that extracts from this Chinese medicine activate a prototype glucocorticoid-response element, increase the level of utrophin protein in human muscle cells and activate the utrophin promoter A. Based on our bioassays we conclude that this particular Chinese medicine used for the treatment of muscular dystrophy patients contains glucocorticoids as one of its active ingredients.


Subject(s)
Glucocorticoids/analysis , Medicine, Chinese Traditional/statistics & numerical data , Methylprednisolone/pharmacology , Animals , COS Cells/drug effects , Cells, Cultured , Chemical Fractionation , Chlorocebus aethiops , Chromatography, High Pressure Liquid , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/biosynthesis , Drug Interactions , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Genes, Reporter , Glucocorticoids/pharmacology , Hormone Antagonists/pharmacology , Humans , Membrane Proteins/analysis , Membrane Proteins/biosynthesis , Mifepristone/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Transfection , Utrophin
15.
Neuromuscul Disord ; 12 Suppl 1: S95-104, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12206803

ABSTRACT

Previous studies on transgenic mice indicate that upregulation of utrophin protein may offer a potential treatment strategy for Duchenne muscular dystrophy. We have analyzed the effect of the glucocorticoid 6alpha-methylprednisolone-21 sodium succinate on utrophin protein levels, using a cell-based assay with differentiated human myotubes, derived from biopsies of healthy individuals or Duchenne muscular dystrophy patients. We found that within 5-7 days 6alpha-methylprednisolone-21 sodium succinate increases utrophin protein up to approximately 40% in both normal and dystrophin-deficient myotubes compared to untreated control cultures. When analyzed in promoter-reporter assays 6alpha-methylprednisolone-21 sodium succinate activated a utrophin promoter A-fragment but did not activate a utrophin promoter B-fragment. Surprisingly, endogenous levels of utrophin mRNA in 6alpha-methylprednisolone-21 sodium succinate-treated muscle cells were unaltered indicating that the utrophin-inducing effect of glucocorticoids may be a result of post-transcriptional mechanisms. We have also analyzed 66 glucocorticoids for their effect on utrophin protein levels and found that glucocorticoids in general are able to induce utrophin protein in human myotubes.


Subject(s)
Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Methylprednisolone/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Cytoskeletal Proteins/genetics , Glucocorticoids/antagonists & inhibitors , Hormone Antagonists/pharmacology , Humans , Membrane Proteins/genetics , Mifepristone/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Messenger/metabolism , Up-Regulation , Utrophin
16.
Neuromuscul Disord ; 14(10): 675-82, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15351425

ABSTRACT

Duchenne muscular dystrophy is a severe X-linked hereditary disease caused by the absence of functional dystrophin. The dystrophin-deficient mdx-mouse strain is a widely used animal model for dystrophin-deficiency. Several therapeutic approaches for muscular dystrophy have been proposed by different laboratories. In order to compare the efficacy of these therapies in the mdx-mouse, it is essential to implement standardized protocols for the assessment of functional and histological parameters in this mouse model. Here, we determine that the minimal 'Feret's diameter' is a geometrical parameter that allows for reliable measure of muscle fiber cross-sectional size. Using this geometrical parameter we calculate variance coefficients of the muscle fiber size and provide reference values for the quantitative assessment of dystrophic symptoms in frequently investigated muscles of wild-type and mdx-mouse. In addition, we compare the variance coefficients of the muscle fiber size with the percentage of muscle fibers with centralized nuclei; another histological hallmark of muscular dystrophy.


Subject(s)
Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Age Factors , Animals , Disease Models, Animal , Histological Techniques/methods , Indoles , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Reference Values
17.
PLoS One ; 6(3): e17963, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21483849

ABSTRACT

Short-chain quinones are described as potent antioxidants and in the case of idebenone have already been under clinical investigation for the treatment of neuromuscular disorders. Due to their analogy to coenzyme Q10 (CoQ10), a long-chain quinone, they are widely regarded as a substitute for CoQ10. However, apart from their antioxidant function, this provides no clear rationale for their use in disorders with normal CoQ10 levels. Using recombinant NAD(P)H:quinone oxidoreductase (NQO) enzymes, we observed that contrary to CoQ10 short-chain quinones such as idebenone are good substrates for both NQO1 and NQO2. Furthermore, the reduction of short-chain quinones by NQOs enabled an antimycin A-sensitive transfer of electrons from cytosolic NAD(P)H to the mitochondrial respiratory chain in both human hepatoma cells (HepG2) and freshly isolated mouse hepatocytes. Consistent with the substrate selectivity of NQOs, both idebenone and CoQ1, but not CoQ10, partially restored cellular ATP levels under conditions of impaired complex I function. The observed cytosolic-mitochondrial shuttling of idebenone and CoQ1 was also associated with reduced lactate production by cybrid cells from mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS) patients. Thus, the observed activities separate the effectiveness of short-chain quinones from the related long-chain CoQ10 and provide the rationale for the use of short-chain quinones such as idebenone for the treatment of mitochondrial disorders.


Subject(s)
NAD(P)H Dehydrogenase (Quinone)/metabolism , Ubiquinone/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HEK293 Cells , Hep G2 Cells , Humans , Lactic Acid/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , NAD/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxidation-Reduction/drug effects , Quinones/metabolism , Rats , Rotenone/pharmacology , Ubiquinone/metabolism , Ubiquinone/pharmacology
18.
PLoS One ; 4(3): e4774, 2009.
Article in English | MEDLINE | ID: mdl-19295909

ABSTRACT

BACKGROUND: Cachexia is among the most debilitating and life-threatening aspects of cancer. It represents a metabolic syndrome affecting essential functional circuits involved in the regulation of homeostasis, and includes anorexia, fat and muscle tissue wasting. The anorexigenic peptide alpha-MSH is believed to be crucially involved in the normal and pathologic regulation of food intake. It was speculated that blockade of its central physiological target, the melanocortin (MC)-4 receptor, might provide a promising anti-cachexia treatment strategy. This idea is supported by the fact that in animal studies, agouti-related protein (AgRP), the endogenous inverse agonist at the MC-4 receptor, was found to affect two hallmark features of cachexia, i.e. to increase food intake and to reduce energy expenditure. METHODOLOGY/PRINCIPAL FINDINGS: SNT207707 and SNT209858 are two recently discovered, non peptidic, chemically unrelated, orally active MC-4 receptor antagonists penetrating the blood brain barrier. Both compounds were found to distinctly increase food intake in healthy mice. Moreover, in mice subcutaneously implanted with C26 adenocarcinoma cells, repeated oral administration (starting the day after tumor implantation) of each of the two compounds almost completely prevented tumor induced weight loss, and diminished loss of lean body mass and fat mass. CONCLUSIONS/SIGNIFICANCE: In contrast to the previously reported peptidic and small molecule MC-4 antagonists, the compounds described here work by the oral administration route. Orally active compounds might offer a considerable advantage for the treatment of cachexia patients.


Subject(s)
Cachexia/drug therapy , Eating/drug effects , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Animals , Body Weight , Brain Chemistry , Cell Line, Tumor , Female , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Random Allocation
19.
Muscle Nerve ; 33(6): 753-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16598790

ABSTRACT

Calpains are Ca2+ -dependent cytosolic cysteine proteases that participate in the pathology of Duchenne muscular dystrophy (DMD). Utrophin is a functional homolog of dystrophin that partially compensates for dystrophin deficiency in myofibers of mdx mice. In this study, we investigated the susceptibility of utrophin to cleavage by calpain in vitro and in muscle cells. We found that utrophin is a direct in vitro substrate of purified calpain I and II. Cleavage of utrophin by calpain I or II generates specific degradation products that are also found in cultured control and DMD myotubes under conditions with elevated intracellular Ca2+ levels. In addition, we showed that activation of cellular calpains by Ca2+ ionophore treatment reduces utrophin protein levels in muscle cells and that calpain inhibition prevents this Ca2+ -induced reduction in utrophin levels. These observations suggest that, beside its known effect on general muscle protein degradation, calpain contributes to DMD pathology by specifically degrading the compensatory protein utrophin.


Subject(s)
Calpain/metabolism , Muscle Cells/enzymology , Muscle, Skeletal/cytology , Muscular Dystrophy, Duchenne/metabolism , Utrophin/metabolism , Animals , Calpain/antagonists & inhibitors , Calpain/isolation & purification , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , In Vitro Techniques , Ionophores/pharmacology , Kidney/cytology , Mice , Muscle Fibers, Skeletal/enzymology , Muscular Dystrophy, Duchenne/pathology , Substrate Specificity , Utrophin/genetics
20.
Bioorg Med Chem Lett ; 15(23): 5176-81, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16185867

ABSTRACT

Dipeptide-derived alpha-keto-amide compounds with potent calpain inhibitory activity have been identified. These reversible covalent inhibitors have IC(50) values down to 25nM and exhibit greatly improved activity in muscle cells compared to the reference compound MDL28170. Several novel calpain inhibitors have shown positive effects on histological parameters in an animal model of Duchenne muscular dystrophy demonstrating their potential as a treatment option for this fatal disease.


Subject(s)
Amides/therapeutic use , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/therapeutic use , Dipeptides/chemistry , Muscular Dystrophy, Animal/drug therapy , Amides/chemistry , Animals , Diaphragm/drug effects , Diaphragm/pathology , Mice , Mice, Inbred mdx , Molecular Structure , Muscular Dystrophy, Animal/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL