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










Publication year range
1.
Am J Pathol ; 189(2): 354-369, 2019 02.
Article in English | MEDLINE | ID: mdl-30448410

ABSTRACT

In muscular dystrophies, muscle membrane fragility results in a tissue-specific increase of danger-associated molecular pattern molecules (DAMPs) and infiltration of inflammatory cells. The DAMP extracellular ATP (eATP) released by dying myofibers steadily activates muscle and immune purinergic receptors exerting dual negative effects: a direct damage linked to altered intracellular calcium homeostasis in muscle cells and an indirect toxicity through the triggering of the immune response and inhibition of regulatory T cells. Accordingly, pharmacologic and genetic inhibition of eATP signaling improves the phenotype in models of chronic inflammatory diseases. In α-sarcoglycanopathy, eATP effects may be further amplified because α-sarcoglycan extracellular domain binds eATP and displays an ecto-ATPase activity, thus controlling eATP concentration at the cell surface and attenuating the magnitude and/or the duration of eATP-induced signals. Herein, we show that in vivo blockade of the eATP/P2X purinergic pathway by a broad-spectrum P2X receptor-antagonist delayed the progression of the dystrophic phenotype in α-sarcoglycan-null mice. eATP blockade dampened the muscular inflammatory response and enhanced the recruitment of forkhead box protein P3-positive immunosuppressive regulatory CD4+ T cells. The improvement of the inflammatory features was associated with increased strength, reduced necrosis, and limited expression of profibrotic factors, suggesting that pharmacologic purinergic antagonism, altering the innate and adaptive immune component in muscle infiltrates, might provide a therapeutic approach to slow disease progression in α-sarcoglycanopathy.


Subject(s)
Adenosine Triphosphate/immunology , Muscular Dystrophy, Animal , Myofibrils , Sarcoglycans/deficiency , T-Lymphocytes, Regulatory , Adenosine Triphosphate/genetics , Animals , Calcium/immunology , Chronic Disease , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/pathology , Myofibrils/immunology , Myofibrils/pathology , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/immunology , Sarcoglycans/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
2.
DNA Cell Biol ; 2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30388038

ABSTRACT

Mandibular hypoplasia, deafness, and progeroid features, with concomitant lipodystrophy, define a multisystem disorder named MDPL syndrome. MDPL has been associated with heterozygous mutations in POLD1 gene resulting in loss of DNA polymerase δ activity. In this study, we report clinical, genetic, and cellular studies of a 13-year-old Pakistani girl, presenting growth retardation, sensorineural deafness, altered distribution of subcutaneous adipose tissue, and insulin resistance. We performed Sanger sequencing of POLD1 gene in the proband and the healthy parents. Fibroblasts obtained from dermal biopsy were evaluated for the specific hallmarks of cellular senescence and for their response to the DNA-induced damage. Patient carried the recurrent heterozygous de novo in frame deletion (c.1812_1814delCTC, p.Ser605del ) within POLD1 gene, previously detected in 16 MDPL patients. In patient's fibroblasts we observed severe nuclear envelope anomalies, presence of micronuclei, accumulation of prelamin A, altered cell growth, and cellular senescence. In addition, we observed a persistence of DNA damage after cisplatin exposure, compared to control cells. In conclusion, the MDPL nuclear and cellular findings resemble features observed in other progeroid syndromes and familial lipodystrophies. Although further investigations will be necessary, these information could be used to establish targeted therapeutic approaches.

3.
J Hum Genet ; 63(6): 761-764, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29556034

ABSTRACT

We present a 13-year-old patient with persistent increase of serum Creatine Kinase (CK) and myalgia after exertion. Skeletal muscle biopsy showed marked reduction of dystrophin expression leading to genetic analysis of DMD gene by MLPA, which detected a single deletion of exon 78. To the best of our knowledge, DMD exon 78 deletion has never been described in literature and, according to prediction, it should lead to loss of reading frame in the dystrophin gene. To further assess the actual effect of exon 78 deletion, we analysed cDNA from muscle mRNA. This analysis confirmed the absence of 32 bp of exon 78. Exclusion of exon 78 changes the open reading frame of exon 79 and generate a downstream stop codon, producing a dystrophin protein of 3703 amino acids instead of 3685 amino acids. Albeit loss of reading frame usually leads to protein degradation and severe phenotype, in this case, we demonstrated that deletion of DMD exon 78 can be associated with a functional protein able to bind DGC complex and a very mild phenotype. This study adds a novel deletion in DMD gene in human and helps to define the compliance between maintaining/disrupting the reading frame and clinical form of the disease.


Subject(s)
Creatine Kinase/blood , Dystrophin/genetics , Exons , Gene Deletion , Muscular Dystrophy, Duchenne/diagnosis , Adolescent , Biopsy , Codon, Terminator , DNA, Complementary/genetics , Humans , Male , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Myalgia/physiopathology , Open Reading Frames , Phenotype , RNA, Messenger/genetics
4.
Lab Invest ; 96(8): 862-71, 2016 08.
Article in English | MEDLINE | ID: mdl-27295345

ABSTRACT

Activation of the proteasome pathway is one of the secondary processes of cell damage, which ultimately lead to muscle degeneration and necrosis in Duchenne muscular dystrophy (DMD). In mdx mice, the proteasome inhibitor bortezomib up-regulates the membrane expression of members of the dystrophin complex and reduces the inflammatory reaction. However, chronic inhibition of the 26S proteasome may be toxic, as indicated by the systemic side-effects caused by this drug. Therefore, we sought to determine the components of the ubiquitin-proteasome pathway that are specifically activated in human dystrophin-deficient muscles. The analysis of a cohort of patients with genetically determined DMD or Becker muscular dystrophy (BMD) unveiled a selective up-regulation of the ubiquitin ligase tripartite motif-containing protein 32 (TRIM32). The induction of TRIM32 was due to a transcriptional effect and it correlated with disease severity in BMD patients. In contrast, atrogin1 and muscle RING-finger protein-1 (MuRF-1), which are strongly increased in distinct types of muscular atrophy, were not affected by the DMD dystrophic process. Knock-out models showed that TRIM32 is involved in ubiquitination of muscle cytoskeletal proteins as well as of protein inhibitor of activated STAT protein gamma (Piasγ) and N-myc downstream-regulated gene, two inhibitors of satellite cell proliferation and differentiation. Accordingly, we showed that in DMD/BMD muscle tissue, TRIM32 induction was more pronounced in regenerating myofibers rather than in necrotic muscle cells, thus pointing out a role of this protein in the regulation of human myoblast cell fate. This finding highlights TRIM32 as a possible therapeutic target to favor skeletal muscle regeneration in DMD patients.


Subject(s)
Muscular Dystrophy, Duchenne/metabolism , Transcription Factors/biosynthesis , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Animals , Case-Control Studies , Humans , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regeneration , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Up-Regulation
5.
Nat Cell Biol ; 18(1): 132-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26571211

ABSTRACT

Genetic defects in myelin formation and maintenance cause leukodystrophies, a group of white matter diseases whose mechanistic underpinnings are poorly understood. Hypomyelination and congenital cataract (HCC), one of these disorders, is caused by mutations in FAM126A, a gene of unknown function. We show that FAM126A, also known as hyccin, regulates the synthesis of phosphatidylinositol 4-phosphate (PtdIns(4)P), a determinant of plasma membrane identity. HCC patient fibroblasts exhibit reduced PtdIns(4)P levels. FAM126A is an intrinsic component of the plasma membrane phosphatidylinositol 4-kinase complex that comprises PI4KIIIα and its adaptors TTC7 and EFR3 (refs 5,7). A FAM126A-TTC7 co-crystal structure reveals an all-α-helical heterodimer with a large protein-protein interface and a conserved surface that may mediate binding to PI4KIIIα. Absence of FAM126A, the predominant FAM126 isoform in oligodendrocytes, destabilizes the PI4KIIIα complex in mouse brain and patient fibroblasts. We propose that HCC pathogenesis involves defects in PtdIns(4)P production in oligodendrocytes, whose specialized function requires massive plasma membrane expansion and thus generation of PtdIns(4)P and downstream phosphoinositides. Our results point to a role for FAM126A in supporting myelination, an important process in development and also following acute exacerbations in multiple sclerosis.


Subject(s)
Cell Membrane/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Animals , Humans , Mice , Mutation/genetics , Phosphatidylinositol Phosphates/genetics , Protein Structure, Tertiary , Protein Transport/genetics , Protein Transport/physiology
6.
Am J Pathol ; 185(12): 3349-60, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26465071

ABSTRACT

Infiltration of immune cells and chronic inflammation substantially affect skeletal and cardiac muscle degeneration in Duchenne muscular dystrophy. In the immune system, extracellular adenosine triphosphate (ATP) released by dying cells is sensed as a danger associated molecular pattern through P2 purinergic receptors. Specifically, the P2X7 subtype has a prominent role in regulating immune system physiology and contributes to inflammasome activation also in muscle cells. Here, we show that in vivo blockade of the extracellular ATP/P2X purinergic signaling pathway by periodate-oxidized ATP delayed the progression of the dystrophic phenotype and dampened the local inflammatory response in mdx mice, a spontaneous mouse model of dystrophin deficiency. Reduced infiltration of leukocytes and macrophages and decreased expression of IL-6 were revealed in the muscles of periodate-oxidized ATP-treated mdx mice. Concomitantly, an increase in Foxp3(+) immunosuppressive regulatory T cells was observed and correlated with enhanced myofiber regeneration. Moreover, we detected reduced concentrations of profibrotic cytokines, including transforming growth factor-ß and connective tissue growth factor, in muscles of periodate-oxidized ATP-treated mdx mice. The improvement of inflammatory features was associated with increased strength and reduced necrosis, thus suggesting that pharmacologic purinergic antagonism altering the adaptive immune component in the muscle infiltrates might represent a promising therapeutic approach in Duchenne muscular dystrophy.


Subject(s)
Muscle, Skeletal/immunology , Muscular Dystrophy, Duchenne/immunology , Receptors, Purinergic P2X/physiology , T-Lymphocytes, Regulatory/immunology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/immunology , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/therapeutic use , Animals , Disease Progression , Drug Evaluation, Preclinical/methods , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/prevention & control , Physical Conditioning, Animal , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2X Receptor Antagonists/therapeutic use , Receptors, Purinergic P2X/metabolism , Regeneration/drug effects , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects
7.
Biochem Biophys Res Commun ; 458(3): 601-604, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25680467

ABSTRACT

Mitochondrial disorders are a heterogeneous group of diseases sharing a defect of the oxidative phosphorylation system. Point mutations in the mitochondrial DNA are a common cause of mitochondrial disorders and frequently affect the sequences encoding mitochondrial transfer RNAs. The m.3271T>C mutation in the mitochondrial tRNA(Leu(UUR)) is traditionally reported in patients with clinical features of the mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) syndrome and in mitochondrial diabetes. Here we describe the clinical, pathological, and molecular features of an Italian child and his asymptomatic mother, carrying the m.3271T>C mutation in the mitochondrial tRNA(Leu(UUR)) gene, in association with an unusual clinical phenotype dominated by hypertrophic cardiomyopathy and provide review literature of cases with this mutation. To the best of our knowledge, there are no reports describing the association of this mutation with cardiomyopathy, and our cases suggest that the m.3271T>C mutation has to be taken into account in the diagnostic approach of maternally inherited cardiomyopathies.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Point Mutation , RNA, Transfer, Leu/genetics , Adolescent , Cardiomyopathy, Hypertrophic/pathology , Child, Preschool , Female , Humans , MELAS Syndrome/genetics , Male , Middle Aged
8.
Hum Mutat ; 34(6): 869-72, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23526554

ABSTRACT

Early-onset epileptic encephalopathies (EOEEs) are a group of rare devastating epileptic syndromes of infancy characterized by severe drug-resistant seizures and electroencephalographic abnormalities. The current study aims to determine the genetic etiology of a familial form of EOEE fulfilling the diagnosis criteria for malignant migrating partial seizures of infancy (MMPSI). We identified two inherited novel mutations in TBC1D24 in two affected siblings. Mutations severely impaired TBC1D24 expression and function, which is critical for maturation of neuronal circuits. The screening of TBC1D24 in an additional set of eight MMPSI patients was negative. TBC1D24 loss of function has been associated to idiopathic infantile myoclonic epilepsy, as well as to drug-resistant early-onset epilepsy with intellectual disability. Here, we describe a familial form of MMPSI due to mutation in TBC1D24, revealing a devastating epileptic phenotype associated with TBC1D24 dysfunction.


Subject(s)
Carrier Proteins/genetics , Heterozygote , Mutation , Spasms, Infantile/genetics , Brain/metabolism , Carrier Proteins/metabolism , Exome , Female , GTPase-Activating Proteins , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Membrane Proteins , Nerve Tissue Proteins , Phenotype , Spasms, Infantile/diagnosis
9.
Epilepsia ; 53(12): 2120-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23205931

ABSTRACT

PURPOSE: Unverricht-Lundborg disease (EPM1A) is frequently due to an unstable expansion of a dodecamer repeat in the CSTB gene, whereas other types of mutations are rare. EPM1A due to homozygous expansion has a rather stereotyped presentation with prominent action myoclonus. We describe eight patients with five different compound heterozygous CSTB point or indel mutations in order to highlight their particular phenotypical presentations and evaluate their genotype-phenotype relationships. METHODS: We screened CSTB mutations by means of Southern blotting and the sequencing of the genomic DNA of each proband. CSTB messenger RNA (mRNA) aberrations were characterized by sequencing the complementary DNA (cDNA) of lymphoblastoid cells, and assessing the protein concentrations in the lymphoblasts. The patient evaluations included the use of a simplified myoclonus severity rating scale, multiple neurophysiologic tests, and electroencephalography (EEG)-polygraphic recordings. To highlight the particular clinical features and disease time-course in compound heterozygous patients, we compared some of their characteristics with those observed in a series of 40 patients carrying the common homozygous expansion mutation observed at the C. Besta Foundation, Milan, Italy. KEY FINDINGS: The eight compound heterozygous patients belong to six EPM1A families (out of 52; 11.5%) diagnosed at the Laboratory of Genetics of the Galliera Hospitals in Genoa, Italy. They segregated five different heterozygous point or indel mutations in association with the common dodecamer expansion. Four patients from three families had previously reported CSTB mutations (c.67-1G>C and c.168+1_18del); one had a novel nonsense mutation at the first exon (c.133C>T) leading to a premature stop codon predicting a short peptide; the other three patients from two families had a complex novel indel mutation involving the donor splice site of intron 2 (c.168+2_169+21delinsAA) and leading to an aberrant transcript with a partially retained intron. The protein dose (cystatin B/ß-actin) in our heterozygous patients was 0.24 ± 0.02, which is not different from that assessed in patients bearing the homozygous dodecamer expansion. The compound heterozygous patients had a significantly earlier disease onset (7.4 ± 1.7 years) than the homozygous patients, and their disease presentations included frequent myoclonic seizures and absences, often occurring in clusters throughout the course of the disease. The seizures were resistant to the pharmacologic treatments that usually lead to complete seizure control in homozygous patients. EEG-polygraphy allowed repeated seizures to be recorded. Action myoclonus progressively worsened and all of the heterozygous patients older than 30 years were in wheelchairs. Most of the patients showed moderate to severe cognitive impairment, and six had psychiatric symptoms. SIGNIFICANCE: EPM1A due to compound heterozygous CSTB mutations presents with variable but often markedly severe and particular phenotypes. Most of our patients presented with the electroclinical features of severe epilepsy, which is unexpected in homozygous patients, and showed frequent seizures resistant to pharmacologic treatment. The presence of variable phenotypes (even in siblings) suggests interactions with other genetic factors influencing the final disease presentation.


Subject(s)
Cystatin B/genetics , INDEL Mutation/genetics , Phenotype , Point Mutation/genetics , Unverricht-Lundborg Syndrome/genetics , Acoustic Stimulation , Adolescent , Adult , Cystatin B/metabolism , DNA Mutational Analysis , Electrodiagnosis , Electroencephalography , Evoked Potentials, Auditory, Brain Stem , Heterozygote , Humans , Immunoprecipitation , Magnetic Resonance Imaging , Male , Neurologic Examination , RNA, Messenger/metabolism , Retrospective Studies , Young Adult
10.
Am J Pathol ; 176(4): 1863-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20304949

ABSTRACT

Duchenne muscular dystrophy (DMD) and its milder allelic variant, Becker muscular dystrophy (BMD), result from mutations of the dystrophin gene and lead to progressive muscle deterioration. Enhanced activation of proteasomal degradation underlies critical steps in the pathogenesis of the DMD/BMD dystrophic process. Previously, we demonstrated that treatment with the proteasome inhibitor MG-132 rescues the cell membrane localization of dystrophin and the dystrophin glycoprotein complex in mdx mice, a natural genetic mouse model of DMD. The current work aims to thoroughly define the therapeutic potential in dystrophinopathies of Velcade, a drug that selectively blocks the ubiquitin-proteasome pathway. Velcade is particularly intriguing since it has been approved for the treatment of multiple myeloma. Therefore, its side effects in humans have been explored. Velcade effects were analyzed through two independent methodological approaches. First, we administered the drug systemically in mdx mice over a 2-week period. In this system, Velcade restores the membrane expression of dystrophin and dystrophin glycoprotein complex members and improves the dystrophic phenotype. In a second approach, we treated with the compound explants from muscle biopsies of DMD or BMD patients. We show that the inhibition of the proteasome pathway up-regulates dystrophin, alpha-sarcoglycan, and beta-dystroglycan protein levels in explants from BMD patients, whereas it increases the proteins of the dystrophin glycoprotein complex in DMD cases.


Subject(s)
Gene Expression Regulation , Muscular Dystrophy, Duchenne/metabolism , Proteasome Inhibitors , Animals , Boronic Acids/pharmacology , Bortezomib , Disease Models, Animal , Dystroglycans/metabolism , Evans Blue/pharmacology , Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Muscular Dystrophy, Duchenne/therapy , Mutation , Pyrazines/pharmacology , Sarcoglycans/metabolism , Ubiquitin/chemistry
11.
Cell Cycle ; 7(14): 2199-207, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18641458

ABSTRACT

Aquaporin-4 (AQP4) is the major water channel expressed in fast-twitch skeletal muscle fibers. AQP4 is reduced in Duchenne and Becker Muscular Dystrophies, but not in caveolinopathies, thus suggesting an interaction with dystrophin or with members of the dystrophin-glycoprotein complex (DGC) rather than a nonspecific effect due to muscle membrane damage. To establish the role of sarcoglycans in AQP4 decrease occurring in muscular dystrophy, AQP4 expression was analyzed in muscle biopsies from patients affected by Limb Girdle Muscular Dystrophies (LGMDs) 2C-F genetically confirmed. In all the LGMD 2C-F (2alpha-, 1beta-, 2gamma-, 1delta-deficiency), AQP4 was severely decreased. This effect was associated to a marked reduction in alpha1-syntrophin levels. In control muscle AQP4 did not show a direct interaction with any of the four sarcoglycans but, it co-immunoprecipitated with alpha1-syntrophin, indicating that this modular protein may link AQP4 levels with the DGC complex. To determine whether AQP4 expression could be affected in other LGMDs due to the defect of a membrane protein not associated to the dystrophin complex, we examined AQP4 expression in 6 patients affected by dysferlin deficiency genetically confirmed. All the patients displayed a reduction of the water channel, and AQP4 expression appeared to correlate with the severity of the muscle histopathological lesions. However, differently from what observed in the sarcoglycans, alpha1-syntrophin expression was normal or just slightly reduced. These results seem to indicate an additional mechanism of regulation of AQP4 levels in muscle cells. In accordance with a specific effect of membrane muscle disorders, AQP4 protein levels were not changed in 3 mitochondrial and 3 metabolic myopathies. In conclusion, AQP4 expression and membrane localization are markedly reduced in LGMD 2B-2F. The role of AQP4 in the degenerative mechanism occurring in these diseases will be the object of our future research.


Subject(s)
Aquaporin 4/metabolism , Membrane Proteins/deficiency , Muscle Proteins/deficiency , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Sarcoglycans/deficiency , Adolescent , Adult , Aged , Caveolin 3/metabolism , Child , Child, Preschool , Dysferlin , Dystrophin/metabolism , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology
12.
Lab Invest ; 88(3): 275-83, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18253147

ABSTRACT

Caveolins are the principal protein components of caveolae, invaginations of the plasma membrane involved in cell signaling and trafficking. Caveolin-3 (Cav-3) is the muscle-specific isoform of the caveolin family and mutations in the CAV3 gene lead to a large group of neuromuscular disorders. In unrelated patients, we identified two distinct CAV3 mutations involving the same codon 78. Patient 1, affected by dilated cardiomyopathy and limb girdle muscular dystrophy (LGMD)-1C, shows an autosomal recessive mutation converting threonine to methionine (T78M). Patient 2, affected by isolated familiar hyperCKemia, shows an autosomal dominant mutation converting threonine to lysine (T78K). Cav-3 wild type (WT) and Cav-3 mutations were transiently transfected into Cos-7 cells. Cav-3 WT and Cav-3 T78M mutant localized at the plasma membrane, whereas Cav-3 T78K was retained in a perinuclear compartment. Cav-3 T78K expression was decreased by 87% when compared with Cav-3 WT, whereas Cav-3 T78M protein levels were unchanged. To evaluate whether Cav-3 T78K and Cav-3 T78M mutants behaved with a dominant negative pattern, Cos-7 cells were cotransfected with green fluorescent protein (GFP)-Cav-3 WT in combination with either mutant or WT Cav-3. When cotransfected with Cav-3 WT or Cav-3 T78M, GFP-Cav-3 WT was localized at the plasma membrane, as expected. However, when cotransfected with Cav-3 T78K, GFP-Cav-3 WT was retained in a perinuclear compartment, and its protein levels were reduced by 60%, suggesting a dominant negative action. Accordingly, Cav-3 protein levels in muscles from a biopsy of patient 2 (T78K mutation) were reduced by 80%. In conclusion, CAV3 T78M and T78K mutations lead to distinct disorders showing different clinical features and inheritance, and displaying distinct phenotypes in vitro.


Subject(s)
Caveolin 3/genetics , Muscular Dystrophies/genetics , Mutation, Missense , Phenotype , Adult , Alleles , Amino Acid Substitution , Animals , COS Cells , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Caveolin 3/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Chlorocebus aethiops , Codon , DNA/analysis , DNA/genetics , Electromyography , Female , Fluorescent Antibody Technique, Indirect , Genes, Dominant , Genes, Recessive , Green Fluorescent Proteins/metabolism , Histocytochemistry , Homozygote , Humans , Immunohistochemistry , In Vitro Techniques , Lysine/metabolism , Male , Methionine/metabolism , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/surgery , Muscle, Smooth/metabolism , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Myocardium/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transfection
13.
Biochem Biophys Res Commun ; 361(2): 445-50, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17662246

ABSTRACT

Glycogen branching enzyme deficiency (glycogen storage disease type IV, GSD-IV) is a rare autosomal recessive disorder of the glycogen synthesis with high mortality. Two female newborns showed severe hypotonia at birth and both died of cardiorespiratory failure, at 4 and 12 weeks, respectively. In both patients, muscle biopsies showed deposits of PAS-positive diastase-resistant material and biochemical analysis in cultured fibroblasts showed markedly reduced glycogen branching enzyme activity. Direct sequencing of GBE1 gene revealed that patient 1 was homozygous for a novel c.691+5 g>c in intron 5 (IVS5+5 g>c). RT-PCR analysis of GBE1 transcripts from fibroblasts cDNA showed that this mutation produce aberrant splicing. Patient 2 was homozygous for a novel c.1643G>A mutation leading to a stop at codon 548 in exon 13 (p.W548X). These data underscore that in GSD-IV a severe phenotype correlates with null mutations, and indicate that RNA analysis is necessary to characterize functional consequences of intronic mutations.


Subject(s)
Glycogen Storage Disease Type IV/genetics , Mutation/genetics , 1,4-alpha-Glucan Branching Enzyme/chemistry , 1,4-alpha-Glucan Branching Enzyme/genetics , Amino Acid Sequence , Base Sequence , Biopsy , Blotting, Western , DNA Mutational Analysis , DNA, Complementary , Exons/genetics , Fatal Outcome , Female , Fibroblasts/enzymology , Humans , Infant, Newborn , Introns/genetics , Molecular Sequence Data , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology
14.
Hum Mutat ; 28(5): 524, 2007 May.
Article in English | MEDLINE | ID: mdl-17397052

ABSTRACT

Molecular characterization of twelve unrelated patients affected by the autosomal recessive osteosclerotic skeletal dysplasia, Pycnodysostosis (cathepsin k deficiency), revealed 11 different genotypes. The mutational profile consisted of 12 different mutations, including nine previously unreported ones, spread throughout the whole gene. One mutation occurred in regions coding predomain, two affected the prodomain and nine others occurred in the mature domain. The novel lesions consisted in six missense mutations c.20T>C (p.L7P), c.494A>G (p.Q165R), c.580G>A (p.G194S), c.746T>C (p.I249T), c.749A>G (p.D250G), c.955G>T (p.G319C), two frameshifts c.60_61dupGA (p.I21RfsX29), c.282dupA (p.S95VfsX9) and a splicing mutation c.890G>A (r.785_890del). The six new missense mutations were examined by western blots of COS-7 cells transfected with mutant CTSK genes. The L7P, occurring within the predicted hydrophobic domain of signal peptide, showed a significantly reduced expression level compared to the wild type control. These findings suggested that the mutation affected targeting and translocation of the nascent lysosomal protein across the endoplasmatic reticulum membrane. The novel amino acid changes were also modeled into the three-dimensional structure that predicted incorrect protein folding for all of them. Molecular characterization of the patients is of particular value for genetic counseling of patients and their families as diagnosis of Pycnodysostosis based on enzyme assay is unpractical and thus not offered routinely.


Subject(s)
Cathepsins/genetics , Dysostoses/genetics , Mutation , Cathepsin K , Exons , Humans , Polymerase Chain Reaction , Polymorphism, Genetic
15.
J Child Neurol ; 21(11): 983-5, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17092469

ABSTRACT

We report an 11-year-old boy with short stature, bilateral ptosis, sensorineural hearing loss, muscle weakness, and endocrine abnormalities. Brain magnetic resonance imaging (MRI) showed a bilateral abnormal signal in the globus pallidus and in the midbrain tegment. Muscle biopsy specimens showed ragged red and cytochrome c oxidase negative fibers, and biochemical analysis of muscle homogenate showed a partial defect of complex I and IV activities of the respiratory chain enzymes. Analysis of mitochondrial DNA by a polymerase chain reaction screening procedure and Southern blot revealed a novel heteroplasmic single mitochondrial DNA deletion of 7.8 kb in different tissues. This deletion was absent in the blood DNA of his mother and brother. This case further expands and confirms the wide clinical spectrum of mitochondrial disorders associated with single large-scale mitochondrial DNA deletions.


Subject(s)
DNA, Mitochondrial/genetics , Gene Deletion , Growth Disorders/genetics , Human Growth Hormone/deficiency , Hypoparathyroidism/genetics , Mitochondrial Encephalomyopathies/genetics , Child , Globus Pallidus/pathology , Growth Disorders/complications , Growth Disorders/pathology , Humans , Hypoparathyroidism/complications , Hypoparathyroidism/pathology , Magnetic Resonance Imaging , Male , Mitochondrial Encephalomyopathies/complications , Mitochondrial Encephalomyopathies/pathology
16.
Nat Genet ; 38(10): 1111-3, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16951682

ABSTRACT

We describe a new autosomal recessive white matter disorder ('hypomyelination and congenital cataract') characterized by hypomyelination of the central and peripheral nervous system, progressive neurological impairment and congenital cataract. We identified mutations in five affected families, resulting in a deficiency of hyccin, a newly identified 521-amino acid membrane protein. Our study highlights the essential role of hyccin in central and peripheral myelination.


Subject(s)
Cataract/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Animals , COS Cells , Cataract/etiology , Child , Chlorocebus aethiops , Genes, Recessive , Hereditary Central Nervous System Demyelinating Diseases/etiology , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Oncogene Proteins/metabolism , Pedigree
17.
Am J Physiol Cell Physiol ; 290(2): C577-82, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16192300

ABSTRACT

In this report, we have developed a novel method to identify compounds that rescue the dystrophin-glycoprotein complex (DGC) in patients with Duchenne or Becker muscular dystrophy. Briefly, freshly isolated skeletal muscle biopsies (termed skeletal muscle explants) from patients with Duchenne or Becker muscular dystrophy were maintained under defined cell culture conditions for a 24-h period in the absence or presence of a specific candidate compound. Using this approach, we have demonstrated that treatment with a well-characterized proteasome inhibitor, MG-132, is sufficient to rescue the expression of dystrophin, beta-dystroglycan, and alpha-sarcoglycan in skeletal muscle explants from patients with Duchenne or Becker muscular dystrophy. These data are consistent with our previous findings regarding systemic treatment with MG-132 in a dystrophin-deficient mdx mouse model (Bonuccelli G, Sotgia F, Schubert W, Park D, Frank PG, Woodman SE, Insabato L, Cammer M, Minetti C, and Lisanti MP. Am J Pathol 163: 1663-1675, 2003). Our present results may have important new implications for the possible pharmacological treatment of Duchenne or Becker muscular dystrophy in humans.


Subject(s)
Cysteine Proteinase Inhibitors/metabolism , Dystrophin/metabolism , Glycoproteins/metabolism , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/metabolism , Proteasome Inhibitors , Animals , Biopsy , Cysteine Proteinase Inhibitors/therapeutic use , Dystroglycans/genetics , Dystroglycans/metabolism , Dystrophin/genetics , Glycoproteins/genetics , Humans , Leupeptins/metabolism , Mice , Multiprotein Complexes , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Proteasome Endopeptidase Complex/metabolism , Sarcoglycans/genetics , Sarcoglycans/metabolism , Tissue Culture Techniques
18.
Biochem Biophys Res Commun ; 333(2): 540-3, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15950936

ABSTRACT

To better elucidate the pathogenesis of lymphocyte recruitment of memory CD4(+) T cells in inflammatory myopathies, we studied the expression of CCR5 and CCR7 on CD4 memory T cells in muscle tissue from 11 patients with juvenile dermatomyositis, six adult patients with polymyositis, two patients with Duchenne muscular dystrophy, and two patients with spinal muscular atrophy. A prevalent infiltration of CCR5(+) effector CD4 T memory cells is observed in inflammatory myopathies. Moreover, we found a strong expression of CCR7 in perifascicular atrophic and in degenerating/regenerating muscle fibers in juvenile dermatomyositis (JDM) but not in fibers from adult polymyositis and Duchenne muscular dystrophy. The selective expression of CCR7 in JDM may open new perspectives in the understanding of the pathogenesis of inflammatory myopathies, offering a new tool for the differential diagnosis of these disorders.


Subject(s)
Dermatomyositis/immunology , Muscle Fibers, Skeletal/immunology , Muscle, Skeletal/immunology , Receptors, CCR5/immunology , Receptors, Chemokine/immunology , T-Lymphocytes/immunology , Cells, Cultured , Child , Child, Preschool , Dermatomyositis/metabolism , Female , Humans , Male , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Receptors, CCR5/metabolism , Receptors, CCR7 , Receptors, Chemokine/metabolism , T-Lymphocytes/metabolism , Tissue Distribution
19.
Muscle Nerve ; 28(4): 508-11, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14506725

ABSTRACT

We report a novel nonsense mitochondrial cytochrome b mutation (G15170A) in a 40-year-old woman with progressive exercise intolerance and lactic acidosis. Muscle biopsy showed several cytochrome c oxidase-positive ragged-red fibers, and reduced activities of respiratory chain complexes I and III. This mutation, resulting in the loss of 228 amino acids of the protein, was very abundant in the patient's muscle, but undetectable in lymphocytes and fibroblasts. Clinical and laboratory data indicate that this defect is the primary cause of the disease, thus adding a new mutation in the cytochrome b gene among the growing number of patients with exercise intolerance and lactic acidosis.


Subject(s)
Codon, Nonsense , Cytochrome b Group/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Exercise/physiology , Muscle, Skeletal/metabolism , Acidosis, Lactic/genetics , Adult , Base Sequence/genetics , Electron Transport Complex I , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Female , Humans , Muscle Cramp/genetics , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , NADH, NADPH Oxidoreductases/metabolism
20.
J Child Neurol ; 18(4): 300-3, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12760436

ABSTRACT

We report a novel T14687C mutation in the mitochondrial transfer ribonucleic acid glutamic acid gene in a 16-year-old boy with myopathy and lactic acidosis, retinopathy, and progressive respiratory failure leading to death. A muscle biopsy showed cytochrome c oxidase-negative ragged-red fibers, and biochemical analysis of the respiratory chain enzymes in muscle homogenate revealed complex I and complex IV deficiencies. The mutation, which affects the trinucleotide (TpsiC) loop, was nearly homoplasmic in the muscle DNA of the proband, but it was absent in his blood and in the blood from the asymptomatic mother, suggesting that it may have been a spontaneous somatic mutation in muscle.


Subject(s)
Mitochondria/genetics , Mitochondrial Myopathies/genetics , Mutation/genetics , RNA, Transfer, Amino Acyl/genetics , Respiratory Insufficiency/genetics , Adolescent , Humans , Male , Mitochondria/pathology , Mitochondrial Myopathies/pathology , Respiratory Insufficiency/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...