Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Pediatr Cardiol ; 43(8): 1870-1878, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35538321

ABSTRACT

Congenital heart disease (CHD) is one of the most specific and yet challenging fields of heart surgery. Apart from the known clinical approaches, including surgery, a significant scale of regenerative therapeutic options is available, which increase the number of cardiomyocytes and restore cardiac function. Although it has been revealed in recent years that mitochondrial transplantation can be used as a promising treatment option in this disease group, there is no clinical evidence for the significance of mitochondrial function in myocardial tissue of patients with CHD regarding cardiac surgery. In this study, mitochondrial morphology and function, myocardial fibrosis, and myocyte atypia were evaluated in myocardial biopsy tissue of pediatric patients with cyanotic and acyanotic CHD, five from each group. After histopathological evaluation of myocardial tissue specimens, mitochondrial morphology and network were analyzed by immunofluorescence staining using an anti-Tom20 antibody, electron transport chain complexes of myocardium were examined by cytochrome c oxidase/succinate dehydrogenase staining, and the amount of ATP was measured by bioluminescence assay. In addition, cardiac markers have been tested to be reviewed as a potential indicator for postoperative follow-up. Myocyte atypia and fibrosis were classified on a scale of 1 to 4. In this study, unlike patients with acyanotic CHD, alterations in mitochondrial network and reduction in ATP production were detected in all pediatric patients with cyanotic CHD. A statistically significant correlation was also determined between mitochondrial dysfunction and cardiac markers. These findings may be assumed as a promising pathway for evaluating the relationship between mitochondrial dysfunction and cyanotic CHD.


Subject(s)
Heart Defects, Congenital , Child , Humans , Adenosine Triphosphate , Cyanosis/etiology , Electron Transport Complex IV/metabolism , Heart Defects, Congenital/complications , Heart Defects, Congenital/surgery , Heart Defects, Congenital/metabolism , Mitochondria/metabolism , Succinate Dehydrogenase/metabolism
2.
Acta Neurol Belg ; 122(6): 1499-1509, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34241798

ABSTRACT

Skeletal muscle pathology is thought to have an important role in the onset and/or progression of amyotrophic lateral sclerosis (ALS), which is a neurodegenerative disorder characterized by progressive muscle weakness. Since miRNAs are recognized as important regulatory factors of essential biological processes, we aimed to identify differentially expressed miRNAs in the skeletal muscle of sporadic ALS patients through the combination of molecular-omic technologies and bioinformatic tools. We analyzed the miRnome profiles of skeletal muscle biopsies acquired from ten sALS patients and five controls with Affymetrix GeneChip miRNA 4.0 Array. To find out differentially expressed miRNAs in patients, data were analyzed by The Institute for Genomic Research-Multi Experiment Viewer (MeV) and miRNAs whose expression difference were statistically significant were identified as candidates. The potential target genes of these miRNAs were predicted by miRWalk 2.0 and were functionally enriched by gene ontology (GO) analysis. The expression level of priority candidates was validated by quantitative real-time PCR (qRT-PCR) analysis. We identified ten differentially expressed miRNAs in patients with a fold change threshold ≥ 2.0, FDR = 0. We identified ten differentially expressed miRNAs in patients with a fold change threshold ≥ 2.0, FDR = 0. Nine out of the ten miRNAs were found to be related to top three enriched ALS-related terms. Based on the qRT-PCR validation of candidate miRNAs, patients were separated into two groups: those with upregulated miR-4429 and miR-1825 expression and those with downregulated miR-638 expression. The different muscle-specific miRNA profiles in sALS patients may indicate the involvement of etiologic heterogeneity, which may allow the development of novel therapeutic strategies.


Subject(s)
Amyotrophic Lateral Sclerosis , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Amyotrophic Lateral Sclerosis/genetics , Gene Ontology , Muscle, Skeletal , Gene Expression Profiling
3.
Biol Futur ; 72(3): 281-290, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34554549

ABSTRACT

3D cell cultures and organoid approach are increasingly being used for basic research and drug discovery of several diseases. Recent advances in these technologies, enabling research on tissue-like structures created in vitro is very important for the value of the data produced. Application of 3D cultures will not only contribute to advancing basic research, but also help to reduce animal usage in biomedical science. The 3D organoid approach is important for research on diseases where patient tissue is difficult to obtain. Therefore, this review aims to show recent advances in the 3D organoid technology in disease modeling and potential usage in translational and personalized medicine of diseases with limited patient material such as neurological diseases and rare diseases.


Subject(s)
Forecasting/methods , Nervous System Diseases/drug therapy , Organoids/metabolism , Rare Diseases/drug therapy , Cell Culture Techniques/methods , Humans
4.
Sci Rep ; 11(1): 18161, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34518586

ABSTRACT

Megaconial Congenital Muscular Dystrophy (CMD) is a rare autosomal recessive disorder characterized by enlarged mitochondria located mainly at the periphery of muscle fibers and caused by mutations in the Choline Kinase Beta (CHKB) gene. Although the pathogenesis of this disease is not well understood, there is accumulating evidence for the presence of mitochondrial dysfunction. In this study, we aimed to investigate whether imbalanced mitochondrial dynamics affects mitochondrial function and bioenergetic efficiency in skeletal muscle cells of Megaconial CMD. Immunofluorescence, confocal and transmission electron microscopy studies revealed impaired mitochondrial network, morphology, and localization in primary skeletal muscle cells of Megaconial CMD. The organelle disruption was specific only to skeletal muscle cells grown in culture. The expression levels of mitochondrial fission proteins (DRP1, MFF, FIS1) were found to be decreased significantly in both primary skeletal muscle cells and tissue sections of Megaconial CMD by Western blotting and/or immunofluorescence analysis. The metabolomic and fluxomic analysis, which were performed in Megaconial CMD for the first time, revealed decreased levels of phosphonucleotides, Krebs cycle intermediates, ATP, and altered energy metabolism pathways. Our results indicate that reduced mitochondrial fission and altered mitochondrial energy metabolism contribute to mitochondrial dysmorphology and dysfunction in the pathogenesis of Megaconial CMD.


Subject(s)
Energy Metabolism , Mitochondrial Dynamics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Cells, Cultured , Fluorescence , Humans , Metabolic Flux Analysis , Metabolomics , Mitochondrial Proteins/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/ultrastructure
5.
J Allergy Clin Immunol Pract ; 9(10): 3752-3766.e4, 2021 10.
Article in English | MEDLINE | ID: mdl-34146746

ABSTRACT

BACKGROUND: Serine/threonine kinase-4 (STK4) deficiency is an autosomal recessive combined immunodeficiency. OBJECTIVE: We aimed to define characteristic clinical and laboratory features to aid the differential diagnosis and determine the most suitable therapy. METHODS: In addition to nine STK4 deficiency patients, we reviewed 15 patients from the medical literature. We compared B lymphocyte subgroups of the cohort with age-matched healthy controls. RESULTS: In the cohort, median age at symptom onset and age at diagnosis were 6 years 8 months (range, 6-248 months) and 7 years 5 months (range, 6-260 months), respectively. The main clinical findings were infections (in all nine patients [9 of 9]), autoimmune or inflammatory diseases (7 of 9), and atopy (4 of 9). CD4 lymphopenia (9 out 9), lymphopenia (7 out 9), intermittent eosinophilia (4 out 9), transient neutropenia (3 out 9), low IgM (4 out 9), and high IgE (4 out 9) were common. Decreased recent thymic emigrants, naive and central memory T cells, but increased effector memory T cells were present. The increase in plasmablasts (P = .003) and decrease in switched memory B cells (P = .022) were significant. When 24 patients are analyzed, cutaneous viral infections (n = 20), recurrent pneumonia (n = 18), Epstein Barr virus-associated lymphoproliferation (n = 11), atopic dermatitis (n = 10), autoimmune cytopenia (n = 7), and lymphoma (n = 6) were frequent. Lymphopenia, CD4 lymphopenia, high IgG, IgA, and IgE were common laboratory characteristics. CONCLUSIONS: The differential diagnosis with autosomal recessive hyper-IgE syndrome is crucial. Because, atopy and CD4 lymphopenia are common in both diseases. Immunoglobulins, antibacterial, and antiviral prophylaxis are the mainstays of treatment. Clinicians may use immunomodulatory therapies during inflammatory or autoimmune complications. However, more data are needed to recommend hematopoietic stem cell transplantation as a safe therapy.


Subject(s)
Epstein-Barr Virus Infections , Lymphopenia , Herpesvirus 4, Human , Humans , Intracellular Signaling Peptides and Proteins , Lymphopenia/diagnosis , Protein Serine-Threonine Kinases , Serine , Threonine
6.
Hum Mol Genet ; 29(24): 3935-3944, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33410474

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating childhood disease primarily affecting lower motoneurons in the spinal cord. SMA is caused by the loss of functional survival of motoneuron (SMN) protein, leading to structural and functional alterations of the cytoskeleton in motoneurons and other cells. Loss of SMN results in impairments of microtubule architecture, but the underlying mechanisms are not completely understood. In this study, we mechanistically analyzed the effects of SMN deficiency on microtubules, demonstrating a reduced stability together with a reduction in alpha tubulin detyrosination. This was caused by increased levels of microtubule-associated protein 1B and tubulin tyrosine ligase, resulting in mitochondrial mislocalization in SMA. Our findings suggest that altered tubulin post-translational modifications and microtubule-associated proteins are involved in the pathomechanisms of SMA, such as an impaired axonal transport of mitochondria.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/pathology , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology , Mutation , Peptide Synthases/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Axonal Transport , Biological Transport , Cells, Cultured , Humans , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Mitochondria , Motor Neurons/metabolism , Muscular Atrophy, Spinal/etiology , Muscular Atrophy, Spinal/metabolism , Peptide Synthases/genetics , Survival of Motor Neuron 1 Protein/genetics , Tubulin/metabolism , Tyrosine/metabolism
7.
Am J Pathol ; 190(10): 2136-2145, 2020 10.
Article in English | MEDLINE | ID: mdl-32650001

ABSTRACT

Secondary mitochondrial damage in skeletal muscles is a common feature of different neuromuscular disorders, which fall outside the mitochondrial cytopathies. The common cause of mitochondrial dysfunction and structural changes in skeletal muscle tissue remains to be discovered. Although they are associated with different clinical, genetic, and pathologic backgrounds, the pathomechanisms underlying neuromuscular disorders might be attributed to the complex interaction and cross talk between mitochondria and the associated miRNAs. This study aimed to identify the common miRNA signatures that are associated with mitochondrial damage in different muscular dystrophies (MDs; Duchenne muscular dystrophy, megaconial congenital muscular dystrophy, Ullrich congenital muscular dystrophy, and α-dystroglycanopathy). The miRNome profiles of skeletal muscle biopsies acquired from four different MD groups and control individuals were analyzed by miRNA microarray. We identified 17 common up-regulated miRNAs in all of the tested MD groups. A specific bioinformatics approach identified 10 of these miRNAs to be specifically related to the mitochondrial pathways. Six miRNAs, miR-134-5p, miR-199a-5p, miR-382-5p, miR-409-3p, miR-497-5p, and miR-708-5p, were associated with the top four mitochondrial pathways and were thus selected as priority candidates for further validation by quantitative real-time PCR analysis. We demonstrate, for the first time, common up-regulated miRNAs that are associated with mitochondrial damage in different MD groups, therefore contributing to the pathophysiology. Our findings may open a new gate toward therapeutics.


Subject(s)
Mitochondria/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophy, Duchenne/genetics , Sclerosis/genetics , Adolescent , Child , Child, Preschool , Computational Biology/methods , Female , Gene Expression Profiling/methods , Humans , Infant , Male , MicroRNAs/genetics
8.
Mol Immunol ; 121: 28-37, 2020 05.
Article in English | MEDLINE | ID: mdl-32151906

ABSTRACT

INTRODUCTION: H Syndrome is an autosomal recessive (AR) disease caused by defects in SLCA29A3 gene. This gene encodes the equilibrative nucleoside transporter, the protein which is highly expressed in spleen, lymph node and bone marrow. Autoinflammation and autoimmunity accompanies H Syndrome (HS). AIM: The aim was to further elucidate the mechanisms of disease by molecular studies in a patient with SLC29A3 gene defect. PATIENT AND METHODS: Mitochondrial dysfunction, lysosomal integrity, cytokine response in response to stimulation with different pattern recognition receptor ligands, and circulating cell-free mitochondrial-DNA(ccf-mtDNA) level in plasma were analyzed compared to controls to understand the cellular triggers of autoinflammation. RNA sequencing (RS) analyses were also performed in monocytes before/after culture with lipopolysaccharide. RESULTS: Patient had progressive destructive arthropathy in addition to clinical findings due to combined immunodeficiency. Pure red cell aplasia (PRCA), vitiligo, diabetes, multiple autoantibody positivity, lymphopenia, increased acute phase reactants were present. Recent thymic emigrants (RTE), naïve T cells were decreased, effector memory CD4 + T cells, nonclassical inflammatory monocytes were increased. Patient's peripheral blood mononuclear cells secreted more IL-1ß and IL-6, showed lysosomal disruption and significant mitochondrial dysfunction compared to healthy controls. Plasma ccf-mtDNA level was significantly elevated compared to age-matched controls (p < 0.05). RNA sequencing studies revealed decreased expression of NLR Family Caspase Recrument-Domain Containing 4(NLRC4), 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4(PFKFB4), serine dehydratase(SDS), heparan sulfate(Glucosamine) 3-O-sulfotransferase 1(HS3ST1), neutral cholesterol ester hydrolase 1 (NCEH1), and interleukin-8 (IL-8) in patient's monocytes compared to controls. Longstanding PRCA, which is possibly autoimmune, resolved after initiating monthly intravenous immunoglobulins (IVIG) and low dose steroids to the patient. CONCLUSION: Although autoinflammation and autoimmunity are reported in HS, by functional analyses we here show in the present patient that over-active inflammasome pathway in HS might be related with mitochondrial and lysosomal dysfunction. Increased plasma ccf-mtDNA may be used as a biomarker of inflammasomopathy in HS. HS should be included in the classification of primary immunodeficiency diseases.


Subject(s)
Autoimmunity/genetics , Contracture/genetics , Hearing Loss, Sensorineural/genetics , Histiocytosis/genetics , Immunologic Deficiency Syndromes/genetics , Nucleoside Transport Proteins/genetics , Adolescent , Contracture/drug therapy , Contracture/immunology , Contracture/pathology , Glucocorticoids/therapeutic use , Hearing Loss, Sensorineural/drug therapy , Hearing Loss, Sensorineural/immunology , Hearing Loss, Sensorineural/pathology , Histiocytosis/drug therapy , Histiocytosis/immunology , Histiocytosis/pathology , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Inflammasomes/immunology , Lysosomes/immunology , Lysosomes/pathology , Male , Mitochondria/immunology , Mitochondria/pathology , Treatment Outcome
9.
J Clin Immunol ; 39(7): 726-738, 2019 10.
Article in English | MEDLINE | ID: mdl-31432443

ABSTRACT

INTRODUCTION: Autosomal recessively inherited lipopolysaccharide-responsive beige-like anchor (LRBA) protein deficiency was shown to be responsible for different types of inborn errors of immunity, such as common variable immunodeficiency (CVID) and autoimmune lymphoproliferative syndrome (ALPS). The aim of this study was to compare patients with LRBA-related ALPS and LRBA-related CVID, to describe their clinical and laboratory phenotypes, and to prepare an algorithm for their diagnosis and management. METHODS: Fifteen LRBA-deficient patients were identified among 31 CVID and 14 possible ALPS patients with Western blotting (WB), primary immunodeficiency disease (PIDD) gene, next-generation panel screening (NGS), and whole exome sequencing (WES). RESULTS: The median age on admission and age of diagnosis were 7 years (0.3-16.5) and 11 years (5-44), respectively. Splenomegaly was seen in 93.3% (14/15) of the patients on admission. Splenectomy was performed to 1/5. Recurrent upper respiratory tract infections (93.3% (14/15)), autoimmune cytopenia (80% (12/15)), chronic diarrhea (53.3% (8/15)), lower respiratory tract infections (53.3% (8/15)), lymphoma (26.6% (4/15)), Evans syndrome (26.6% (4/15)), and autoimmune thyroiditis (20% (3/15)) were common clinical findings and diseases. Lymphopenia (5/15), intermittant neutropenia (4/15), eosinophilia (4/15), and progressive hypogammaglobulinemia are recorded in given number of patients. Double negative T cells (TCRαß+CD4-CD8-) were increased in 80% (8/10) of the patients. B cell percentage/numbers were low in 60% (9/15) of the patients on admission. Decreased switched memory B cells, decreased naive and recent thymic emigrant (RTE) Thelper (Th) cells, markedly increased effector memory/effector memory RA+ (TEMRA) Th were documented. Large PD1+ population, increased memory, and enlarged follicular helper T cell population in the CD4+ T cell compartment was seen in one of the patients. Most of the deleterious missense mutations were located in the DUF1088 and BEACH domains. Interestingly, one of the two siblings with the same homozygous LRBA defect did not have any clinical symptom. Hematopoietic stem cell transplantation (HSCT) was performed to 7/15 (46.6%) of the patients. Transplanted patients are alive and well after a median of 2 years (1-3). In total, one patient died from sepsis during adulthood before HSCT. CONCLUSION: Patients with LRBA deficiency may initially be diagnosed as CVID or ALPS in the clinical practice. Progressive decrease in B cells as well as IgG in ALPS-like patients and addition of IBD symptoms in the follow-up should raise the suspicion for LRBA deficiency. Decreased switched memory B cells, decreased naive and recent thymic emigrant (RTE) Th cells, and markedly increased effector memory/effector memory RA+ Th cells (TEMRA Th) cells are important for the diagnosis of the patients in addition to clinical features. Analysis of protein by either WB or flow cytometry is required when the clinicians come across especially with missense LRBA variants of uncertain significance. High rate of malignancy shows the regulatory T cell's important role of immune surveillance. HSCT is curative and succesful in patients with HLA-matched family donor.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Autoimmune Lymphoproliferative Syndrome/diagnosis , Autoimmune Lymphoproliferative Syndrome/etiology , Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/etiology , Genetic Association Studies , Genetic Predisposition to Disease , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Autoimmune Lymphoproliferative Syndrome/complications , Autoimmune Lymphoproliferative Syndrome/therapy , Biomarkers , Child , Child, Preschool , Combined Modality Therapy , Common Variable Immunodeficiency/complications , Common Variable Immunodeficiency/therapy , Communicable Diseases/etiology , Female , Genetic Association Studies/methods , Genetic Loci , Hematopoietic Stem Cell Transplantation , Humans , Immunophenotyping , Male , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Exome Sequencing , Young Adult
10.
Orphanet J Rare Dis ; 14(1): 179, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31311558

ABSTRACT

BACKGROUND: The protein O-mannosyltransferase 1, encoded by the POMT1 gene, is a key enzyme in the glycosylation of α-dystroglycan. POMT1-related disorders belong to the group of dystroglycanopathies characterized by a proximally pronounced muscular dystrophy with structural or functional involvement of the brain and/or the eyes. The phenotypic spectrum ranges from the severe Walker-Warburg syndrome (WWS) to milder forms of limb girdle muscular dystrophy (LGMD). The phenotypic severity of POMT1-related dystroglycanopathies depends on the residual enzyme activity. A genotype-phenotype correlation can be assumed. RESULTS: The clinical, neuroradiological, and genetic findings of 35 patients with biallelic POMT1 mutations (15 WWS, 1 MEB (muscle-eye-brain disease), 19 LGMD) from 27 independent families are reported. The representative clinical course of an infant with WWS and the long-term course of a 32 years old patient with LGMD are described in more detail. Specific features of 15 patients with the homozygous founder mutation p.Ala200Pro are defined as a distinct and mildly affected LGMD subgroup. Ten previously reported and 8 novel POMT1 mutations were identified. Type and location of each of the POMT1 mutations are evaluated in detail and a list of all POMT1 mutations reported by now is provided. Patients with two mutations leading to premature protein termination had a WWS phenotype, while the presence of at least one missense mutation was associated with milder phenotypes. In the patient with MEB-like phenotype two missense mutations were observed within the catalytic active domain of the enzyme. CONCLUSIONS: Our large cohort confirms the importance of type and location of each POMT1 mutation for the individual clinical manifestation and thereby expands the knowledge on the genotype-phenotype correlation in POMT1-related dystroglycanopathies. This genotype-phenotype correlation is further supported by the observation of an intrafamiliar analogous clinical manifestation observed in all affected 13 siblings from 5 independent families. Our data confirm the progressive nature of the disease also in milder LGMD phenotypes, ultimately resulting in loss of ambulation at a variable age. Our data define two major clinical POMT1 phenotypes, which should prompt genetic testing including the POMT1 gene: patients with a severe WWS manifestation predominantly present with profound neonatal muscular hypotonia and a severe and progressive hydrocephalus with involvement of brainstem and/or cerebellum. The presence of an occipital encephalocele in a WWS patient might point to POMT1 as causative gene within the different genes associated with WWS. The milder LGMD phenotypes constantly show markedly elevated creatine kinase values in combination with microcephaly and cognitive impairment.


Subject(s)
Mannosyltransferases/genetics , Mutation/genetics , Female , Genetic Association Studies , Humans , Infant , Infant, Newborn , Male , Muscular Dystrophies, Limb-Girdle/genetics , Walker-Warburg Syndrome/genetics
11.
BMC Neurol ; 18(1): 207, 2018 Dec 15.
Article in English | MEDLINE | ID: mdl-30553274

ABSTRACT

BACKGROUND: Alpha-dystroglycan (αDG) is an extracellular peripheral glycoprotein that acts as a receptor for both extracellular matrix proteins containing laminin globular domains and certain arenaviruses. An important enzyme, known as Like-acetylglucosaminyltransferase (LARGE), has been shown to transfer repeating units of -glucuronic acid-ß1,3-xylose-α1,3- (matriglycan) to αDG that is required for functional receptor as an extracellular matrix protein scaffold. The reduction in the amount of LARGE-dependent matriglycan result in heterogeneous forms of dystroglycanopathy that is associated with hypoglycosylation of αDG and a consequent lack of ligand-binding activity. Our aim was to investigate whether LARGE expression showed correlation with glycosylation of αDG and histopathological parameters in different types of muscular dystrophies, except for dystroglycanopathies. METHODS: The expression level of LARGE and glycosylation status of αDG were examined in skeletal muscle biopsies from 26 patients with various forms of muscular dystrophy [Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), sarcoglycanopathy, dysferlinopathy, calpainopathy, and merosin and collagen VI deficient congenital muscular dystrophies (CMDs)] and correlation of results with different histopathological features was investigated. RESULTS: Despite the fact that these diseases are not caused by defects of glycosyltransferases, decreased expression of LARGE was detected in many patient samples, partly correlating with the type of muscular dystrophy. Although immunolabelling of fully glycosylated αDG with VIA4-1 was reduced in dystrophinopathy patients, no significant relationship between reduction of LARGE expression and αDG hypoglycosylation was detected. Also, Merosin deficient CMD patients showed normal immunostaining with αDG despite severe reduction of LARGE expression. CONCLUSIONS: Our data shows that it is not always possible to correlate LARGE expression and αDG glycosylation in different types of muscular dystrophies and suggests that there might be differences in αDG processing by LARGE which could be regulated under different pathological conditions.


Subject(s)
Muscular Dystrophies/metabolism , N-Acetylglucosaminyltransferases/biosynthesis , Dystroglycans/metabolism , Female , Glycosylation , Humans , Male , Muscle, Skeletal/metabolism , N-Acetylglucosaminyltransferases/analysis
12.
J Neurol Sci ; 393: 100-104, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30153568

ABSTRACT

BACKGROUND: Primary myoblast cell cultures display the phenotypic characteristics and genetic defects of the donor tissue and represent an in vitro model system reflecting the disease pathology. They have been generated only from freshly harvested tissue biopsies. Here, we describe a novel technique to establish myoblast cell cultures from cryopreserved skeletal muscle biopsy tissues that are useful for diagnostic and research purposes. METHODS AND RESULTS: This protocol was performed on seven gradually frozen muscle biopsy specimens from various neuromuscular disorders that were stored in dimethylsulfoxide (DMSO)-supplemented freezing media at -80 °C for up to one year. After storage for varying periods of time, primary myoblast cultures were successfully established from all cryopreserved biopsy tissues without any chromosomal abnormality. Desmin immunoreactivity confirmed that the cell cultures contained >90% pure myoblasts. The myoblasts differentiated into multinucleated myotubes successfully. Furthermore, there were no statistically significant differences in cell viability, metabolic activity, population doubling time, and myocyte enhancer factor 2 (MEF2C) expression between cell cultures established from freshly harvested and one year-stored frozen tissue specimens. CONCLUSIONS: This protocol opens up new horizons for basic research and the pre-clinical studies of novel therapies by using cryopreserved skeletal muscle biopsies stored under suitable conditions in tissue banks.


Subject(s)
Cryopreservation , Muscle, Skeletal , Myoblasts , Primary Cell Culture/methods , Adult , Biopsy , Cell Proliferation , Cell Survival , Female , Humans , Karyotyping , MEF2 Transcription Factors/metabolism , Male , Middle Aged , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , Myoblasts/pathology , Myoblasts/physiology , Time Factors , Young Adult
13.
J Pregnancy ; 2018: 9718316, 2018.
Article in English | MEDLINE | ID: mdl-30151283

ABSTRACT

AIM: To show the importance of prenatal diagnosis of Duchenne Muscular Dystrophy (DMD) and to demonstrate the effect of DMD gene mutations on gestational outcomes. MATERIALS AND METHODS: We retrospectively evaluated 89 pregnancies in 81 individuals who were referred to Hacettepe University for prenatal diagnosis of DMD between January 2000 and December 2015. Prenatal diagnostic methods (chorionic villus sampling (CVS): 66, amniocentesis (AC): 23) were compared for test results, demographic features, and obstetric outcomes of pregnancies. The female fetuses were divided into two groups according to the DMD status (healthy or carrier) to understand the effect of DMD gene mutations on obstetric outcomes. RESULTS: Eight prenatally diagnosed disease-positive fetuses were terminated. There was no statistically significant difference between the CVS and AC groups in terms of study variables. There were 46 male fetuses (51.6%) and 43 female fetuses (48.4%). Fifteen of the female fetuses were carriers (34.8%). Median birthweight values were statistically insignificantly lower in the carrier group. CONCLUSION: Pregnancies at risk for DMD should be prenatally tested to prevent the effect of disease on families and DMD carrier fetuses had obstetric outcomes similar to DMD negative female fetuses.


Subject(s)
Amniocentesis/statistics & numerical data , Chorionic Villi Sampling/statistics & numerical data , Heterozygote , Muscular Dystrophy, Duchenne/diagnosis , Adult , Female , Genetic Carrier Screening/methods , Humans , Male , Muscular Dystrophy, Duchenne/genetics , Mutation/genetics , Pregnancy , Pregnancy Outcome/genetics , Retrospective Studies
14.
Neuromuscul Disord ; 24(7): 624-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24856141

ABSTRACT

We performed genome-wide homozygosity mapping and mapped a novel myopathic phenotype to chromosomal region 1q25 in a consanguineous family with three affected individuals manifesting proximal and distal weakness and atrophy, rigid spine and contractures of the proximal and distal interphalangeal hand joints. Additionally, cardiomyopathy and respiratory involvement were noted. DNA sequencing of torsinA-interacting protein 1 (TOR1AIP1) gene encoding lamina-associated polypeptide 1B (LAP1B), showed a homozygous c.186delG mutation that causes a frameshift resulting in a premature stop codon (p.E62fsTer25). We observed that expression of LAP1B was absent in the patient skeletal muscle fibres. Ultrastructural examination showed intact sarcomeric organization but alterations of the nuclear envelope including nuclear fragmentation, chromatin bleb formation and naked chromatin. LAP1B is a type-2 integral membrane protein localized in the inner nuclear membrane that binds to both A- and B-type lamins, and is involved in the regulation of torsinA ATPase. Interestingly, luminal domain-like LAP1 (LULL1)-an endoplasmic reticulum-localized partner of torsinA-was overexpressed in the patient's muscle in the absence of LAP1B. Therefore, the findings suggest that LAP1 and LULL1 might have a compensatory effect on each other. This study expands the spectrum of genes associated with nuclear envelopathies and highlights the critical function for LAP1B in striated muscle.


Subject(s)
Membrane Proteins/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Nuclear Envelope/ultrastructure , Nuclear Proteins/genetics , Adolescent , Adult , Amino Acid Sequence , Carrier Proteins/metabolism , Cytoskeletal Proteins , DNA Mutational Analysis , Family , Female , Fluorescent Antibody Technique , Frameshift Mutation , Humans , Male , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Molecular Sequence Data , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Pedigree , RNA, Messenger , Sarcomeres/metabolism , Sarcomeres/ultrastructure
15.
J Med Genet ; 50(7): 437-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23687351

ABSTRACT

BACKGROUND: Autosomal recessive limb girdle muscular dystrophy (LGMD2) is a heterogeneous group of myopathies characterised by progressive muscle weakness involving proximal muscles of the shoulder and pelvic girdles including at least 17 different genetic entities. Additional loci have yet to be identified as there are families which are unlinked to any of the known loci. Here we have investigated a consanguineous family with LGMD2 with two affected individuals in order to identify the causative gene defect. METHODS AND RESULTS: We performed genome wide homozygosity mapping and mapped the LGMD2 phenotype to chromosome 2q35-q36.3. DNA sequence analysis of the highly relevant candidate gene DES revealed a homozygous splice site mutation c.1289-2A>G in the two affected family members. Immunofluorescent staining and western blot analysis showed that the expression and the cytoskeletal network formation of mutant desmin were well preserved in skeletal muscle fibres. Unlike autosomal dominant desminopathies, ultrastructural alterations such as disruption of myofibrillar organisation, formation of myofibrillar degradation products and dislocation/aggregation of membranous organelles were not present. This novel splice site mutation results in addition of 16 amino acids within the tail domain of desmin, which has been suggested to interact with lamin B protein. We also detected a specific disruption of desmin-lamin B interaction in the skeletal muscle of the patient by confocal laser scanning microscopy. CONCLUSIONS: Our study reveals that autosomal recessive mutations in DES cause LGMD2 phenotype without features of myofibrillar myopathy.


Subject(s)
Desmin/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Mutation , Adult , Chromosome Mapping , Consanguinity , Genes, Recessive , Genotype , Homozygote , Humans , Male , Microscopy, Electron, Transmission , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/pathology , Pedigree , Phenotype , RNA Splice Sites
16.
Neuromuscul Disord ; 23(1): 15-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22985905

ABSTRACT

The identification of two independent mutations is rarely described between affected members of the same family with Duchenne Muscular Dystrophy. This study reports the presence of two distinct intragenic dystrophin deletions in a Turkish family. Exon 54 deletion was identified originally in the proband, whereas his maternal cousin had deletions of exons 43-50 in the dystrophin gene. As indicated, only the mother of the proband was identified as exon 54 deletion carrier however, the proband's cousin was detected as a sporadic case. These molecular genetic data reveal an interesting and novel mixture, in the same family, of both mutations of the same gene.


Subject(s)
Dystrophin/genetics , Family , Gene Deletion , Muscular Dystrophy, Duchenne/genetics , Child , Child, Preschool , Exons/genetics , Heterozygote , Humans , Male , Muscular Dystrophy, Duchenne/ethnology , Pedigree , Turkey
17.
N Engl J Med ; 364(10): 939-46, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21388311

ABSTRACT

Dystroglycan, which serves as a major extracellular matrix receptor in muscle and the central nervous system, requires extensive O-glycosylation to function. We identified a dystroglycan missense mutation (Thr192→Met) in a woman with limb-girdle muscular dystrophy and cognitive impairment. A mouse model harboring this mutation recapitulates the immunohistochemical and neuromuscular abnormalities observed in the patient. In vitro and in vivo studies showed that the mutation impairs the receptor function of dystroglycan in skeletal muscle and brain by inhibiting the post-translational modification, mediated by the glycosyltransferase LARGE, of the phosphorylated O-mannosyl glycans on α-dystroglycan that is required for high-affinity binding to laminin.


Subject(s)
Dystroglycans/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Mutation, Missense , Animals , Disease Models, Animal , Female , Humans , Mice , Pedigree , Phenotype , Sequence Analysis, DNA
18.
Cell Biol Int ; 35(1): 9-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20795944

ABSTRACT

The Abeta (amyloid-beta) peptide is derived from the sequential cleavage of AbetaPP (amyloid-beta precursor protein) by two enzymes, the ß- and γ-secretases. The major ß-secretase, identified as the novel transmembrane aspartic protease BACE1 (beta site APP-cleaving enzyme 1), mediates the primary amyloidogenic cleavage of AbetaPP and initiates the production of Abeta. It has been implicated in the proteolytic processing of another substrate, namely ST6Gal1 (ß galactoside α2,6-sialyltransferase 1), which is the major α2,6-sialyltransferase responsible for the broad synthesis of glycoproteins and glycolipids. The present study investigated the effect of overexpression of AbetaPP on expression and secretion of ST6Gal1 in skeletal muscle cells by inducing overexpression of wild-type full-length 751-AbetaPP in the mouse myogenic cell line C2C12. Expression and secretion of the ST6Gal1 enzyme were analysed by Western blot and/or immunofluorescence staining. The results of our study demonstrated that AbetaPP overexpression in C2C12 cells increased the expression and the secretion of ST6Gal1 enzyme in vitro.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Myoblasts, Skeletal/enzymology , Sialyltransferases/biosynthesis , Animals , Cells, Cultured , Gene Expression Regulation, Enzymologic , Humans , Mice , Myoblasts, Skeletal/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , beta-D-Galactoside alpha 2-6-Sialyltransferase
19.
Am J Hum Genet ; 87(6): 834-41, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21109228

ABSTRACT

Limb-girdle muscular dystrophy (LGMD) is a genetically heterogeneous group of inherited muscular disorders manifesting symmetric, proximal, and slowly progressive muscle weakness. Using Affymetrix 250K SNP Array genotyping and homozygosity mapping, we mapped an autosomal-recessive LGMD phenotype to the telomeric portion of chromosome 8q in a consanguineous Turkish family with three affected individuals. DNA sequence analysis of PLEC identified a homozygous c.1_9del mutation containing an initiation codon in exon 1f, which is an isoform-specific sequence of plectin isoform 1f. The same homozygous mutation was also detected in two additional families during the analysis of 72 independent LGMD2-affected families. Moreover, we showed that the expression of PLEC was reduced in the patient's muscle and that there was almost no expression for plectin 1f mRNA as a result of the mutation. In addition to dystrophic changes in muscle, ultrastructural alterations, such as membrane duplications, an enlarged space between the membrane and sarcomere, and misalignment of Z-disks, were observed by transmission electron microscopy. Unlike the control skeletal muscle, no sarcolemmal staining of plectin was detected in the patient's muscle. We conclude that as a result of plectin 1f deficiency, the linkage between the sarcolemma and sarcomere is broken, which could affect the structural organization of the myofiber. Our data show that one of the isoforms of plectin plays a key role in skeletal muscle function and that disruption of the plectin 1f can cause the LGMD2 phenotype without any dermatologic component as was previously reported with mutations in constant exons of PLEC.


Subject(s)
Exons , Genes, Recessive , Muscular Dystrophies, Limb-Girdle/genetics , Mutation , Plectin/genetics , Protein Isoforms/genetics , Consanguinity , Female , Humans , Male , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...