Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
1.
ACS Chem Biol ; 15(7): 1820-1825, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32551539

RESUMO

Expanded RNA repeats cause more than 30 incurable diseases. One approach to mitigate their toxicity is by using small molecules that assemble into potent, oligomeric species upon binding to the disease-causing RNA in cells. Herein, we show that the expanded repeat [r(CUG)exp] that causes myotonic dystrophy type 1 (DM1) catalyzes the in situ synthesis of its own inhibitor using an RNA-templated tetrazine ligation in DM1 patient-derived cells. The compound synthesized on-site improved DM1-associated defects at picomolar concentrations, enhancing potency by 10 000-fold, compared to its parent compounds that cannot undergo oligomerization. A fluorogenic reaction is also described where r(CUG)exp templates the synthesis of its own imaging probe to enable visualization of the repeat in its native context in live cells and muscle tissue.


Assuntos
Fluoresceínas/farmacologia , Corantes Fluorescentes/farmacologia , Compostos Heterocíclicos com 1 Anel/farmacologia , Miotonina Proteína Quinase/genética , RNA/antagonistas & inibidores , Animais , Química Click , Fluoresceínas/síntese química , Corantes Fluorescentes/síntese química , Compostos Heterocíclicos com 1 Anel/síntese química , Humanos , Camundongos , Distrofia Miotônica/enzimologia , Distrofia Miotônica/genética , RNA/genética , Sequências de Repetição em Tandem , Transcrição Gênica/efeitos dos fármacos
2.
Proc Natl Acad Sci U S A ; 116(42): 20991-21000, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31570586

RESUMO

A CTG repeat expansion in the DMPK gene is the causative mutation of myotonic dystrophy type 1 (DM1). Transcription of the expanded CTG repeat produces toxic gain-of-function CUG RNA, leading to disease symptoms. A screening platform that targets production or stability of the toxic CUG RNA in a selective manner has the potential to provide new biological and therapeutic insights. A DM1 HeLa cell model was generated that stably expresses a toxic r(CUG)480 and an analogous r(CUG)0 control from DMPK and was used to measure the ratio-metric level of r(CUG)480 versus r(CUG)0. This DM1 HeLa model recapitulates pathogenic hallmarks of DM1, including CUG ribonuclear foci and missplicing of pre-mRNA targets of the muscleblind (MBNL) alternative splicing factors. Repeat-selective screening using this cell line led to the unexpected identification of multiple microtubule inhibitors as hits that selectively reduce r(CUG)480 levels and partially rescue MBNL-dependent missplicing. These results were validated by using the Food and Drug Administration-approved clinical microtubule inhibitor colchicine in DM1 mouse and primary patient cell models. The mechanism of action was found to involve selective reduced transcription of the CTG expansion that we hypothesize to involve the LINC (linker of nucleoskeleton and cytoskeleton) complex. The unanticipated identification of microtubule inhibitors as selective modulators of toxic CUG RNA opens research directions for this form of muscular dystrophy and may shed light on the biology of CTG repeat expansion and inform therapeutic avenues. This approach has the potential to identify modulators of expanded repeat-containing gene expression for over 30 microsatellite expansion disorders.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Microtúbulos/efeitos dos fármacos , Distrofia Miotônica/genética , RNA/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Expansão das Repetições de Trinucleotídeos/efeitos dos fármacos , Animais , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Microtúbulos/genética , Microtúbulos/metabolismo , Distrofia Miotônica/enzimologia , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , RNA/química , RNA/metabolismo
4.
Biochim Biophys Acta Gene Regul Mech ; 1860(6): 740-749, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28435090

RESUMO

In yeast and higher eukaryotes nuclear retention of transcripts may serve in control over RNA decay, nucleocytoplasmic transport and premature cytoplasmic appearance of mRNAs. Hyperadenylation of RNA is known to be associated with nuclear retention, but the cause-consequence relationship between hyperadenylation and regulation of RNA nuclear export is still unclear. We compared polyadenylation status between normal and expanded DMPK transcripts in muscle cells and tissues derived from unaffected individuals and patients with myotonic dystrophy type 1 (DM1). DM1 is an autosomal dominant disorder caused by (CTG)n repeat expansion in the DMPK gene. DM1 etiology is characterized by an almost complete block of nuclear export of DMPK transcripts carrying a long (CUG)n repeat, including aberrant sequestration of RNA-binding proteins. We show here by use of cell fractionation, RNA size separation and analysis of poly(A) tail length that a considerable fraction of transcripts from the normal DMPK allele is also retained in the nucleus (~30%). They carry poly(A) tails with an unusually broad length distribution, ranging between a few dozen to >500 adenosine residues. Remarkably, expanded DMPK (CUG)n transcripts from the mutant allele, almost exclusively nuclear, carry equally long poly(A) tails. Our findings thus suggest that nuclear retention may be a common feature of regulation of DMPK RNA expression. The typical forced nuclear residence of expanded DMPK transcripts affects this regulation in tissues of DM1 patients, but not through hyperadenylation.


Assuntos
Núcleo Celular/metabolismo , Regulação Enzimológica da Expressão Gênica , Miotonina Proteína Quinase/biossíntese , Poli A , Expansão das Repetições de Trinucleotídeos , Alelos , Linhagem Celular Transformada , Núcleo Celular/genética , Núcleo Celular/patologia , Humanos , Distrofia Miotônica/enzimologia , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Miotonina Proteína Quinase/genética
5.
J Clin Invest ; 127(2): 549-563, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28067669

RESUMO

Myotonic dystrophy type I (DM1) is a disabling multisystemic disease that predominantly affects skeletal muscle. It is caused by expanded CTG repeats in the 3'-UTR of the dystrophia myotonica protein kinase (DMPK) gene. RNA hairpins formed by elongated DMPK transcripts sequester RNA-binding proteins, leading to mis-splicing of numerous pre-mRNAs. Here, we have investigated whether DM1-associated muscle pathology is related to deregulation of central metabolic pathways, which may identify potential therapeutic targets for the disease. In a well-characterized mouse model for DM1 (HSALR mice), activation of AMPK signaling in muscle was impaired under starved conditions, while mTORC1 signaling remained active. In parallel, autophagic flux was perturbed in HSALR muscle and in cultured human DM1 myotubes. Pharmacological approaches targeting AMPK/mTORC1 signaling greatly ameliorated muscle function in HSALR mice. AICAR, an AMPK activator, led to a strong reduction of myotonia, which was accompanied by partial correction of misregulated alternative splicing. Rapamycin, an mTORC1 inhibitor, improved muscle relaxation and increased muscle force in HSALR mice without affecting splicing. These findings highlight the involvement of AMPK/mTORC1 deregulation in DM1 muscle pathophysiology and may open potential avenues for the treatment of this disease.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Complexos Multiproteicos/antagonistas & inibidores , Fibras Musculares Esqueléticas/enzimologia , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/enzimologia , Ribonucleotídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Adulto , Aminoimidazol Carboxamida/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Mutantes , Pessoa de Meia-Idade , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Relaxamento Muscular/efeitos dos fármacos , Relaxamento Muscular/genética , Distrofia Miotônica/genética , Distrofia Miotônica/fisiopatologia , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , Transdução de Sinais/genética , Sirolimo/farmacocinética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
6.
Neuromuscul Disord ; 26(6): 378-85, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27133661

RESUMO

Sarcoplasmic/endoplasmic reticulum Ca(2+) ATPase (SERCA) pumps play the major role in lowering cytoplasmic calcium concentration in skeletal muscle by catalyzing the ATP-dependent transport of Ca(2+) from the cytosol to the lumen of the sarcoplasmic reticulum (SR). Although SERCA abnormalities have been hypothesized to contribute to the dysregulation of intracellular Ca(2+) homeostasis and signaling in muscle of patients with myotonic dystrophy (DM) and hypothyroid myopathy, the characterization of SERCA pumps remains elusive and their impairment is still unclear. We assessed the activity of SR Ca(2+)-ATPase, expression levels and fiber distribution of SERCA1 and SERCA2, and oligomerization of SERCA1 protein in muscle of patients with DM type 1 and 2, and with hypothyroid myopathy. Our data provide evidence that SR Ca(2+) ATPase activity, protein levels and muscle fiber distribution of total SERCA1 and SERCA2, and SERCA1 oligomerization pattern are similar in patients with both DM1 and DM2, hypothyroid myopathy and in control subjects. We prove that SERCA1b, the neonatal isoform of SERCA1, is expressed at protein level in muscle of patients with DM2 and, in lower amount, of patients with DM1. Our present study demonstrates that SERCA function is not altered in muscle of patients with DM and with hypothyroid myopathy.


Assuntos
Hipotireoidismo/enzimologia , Músculo Esquelético/enzimologia , Distrofia Miotônica/enzimologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Adulto , Feminino , Humanos , Hipotireoidismo/patologia , Isoenzimas , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/patologia , Distrofia Miotônica/patologia , Adulto Jovem
7.
Neuromuscul Disord ; 26(6): 370-3, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27132119

RESUMO

Myotonic dystrophy type 2 (DM2) is a rare, autosomal dominant, multisystem disorder with proximal weakness, myotonia, pain and cataract as important symptoms. Given the assumed underreporting of DM2 in the Netherlands combined with the predominant role of pain in DM2 as well as in fibromyalgia syndrome (FMS), we hypothesized there will be an excess prevalence of DM2 in patients with (suspected) FMS. Our objective was to determine the prevalence of DM2 in patients with suspected FMS. A prevalence of 2% was considered a relevant excess frequency. Between November 2011 and April 2014, 398 patients with suspected FMS who had been assessed by a rheumatologist participated in this cross-sectional study. 95% of the study population was female, with a mean age of 42 years. The final ICD-9 diagnoses were collected, in 96% the diagnosis was FMS. 92% met the 2010 American College of Rheumatology (ACR) diagnostic criteria for FMS. A questionnaire including neuromuscular symptoms was completed. Creatine kinase was determined, and genetic testing for DM2 was conducted in all patients. DM2 was established in only one patient (0.25%, 95% CI 0.04-1.4%), thus disapproving our hypothesis of a relevant prevalence of 2%. Our results suggest that patients with suspected FMS should not routinely be tested for DM2.


Assuntos
Fibromialgia/complicações , Fibromialgia/epidemiologia , Distrofia Miotônica/complicações , Distrofia Miotônica/epidemiologia , Adolescente , Adulto , Idoso , Creatina Quinase/metabolismo , Estudos Transversais , Feminino , Fibromialgia/enzimologia , Fibromialgia/genética , Humanos , Masculino , Pessoa de Meia-Idade , Distrofia Miotônica/enzimologia , Distrofia Miotônica/genética , Prevalência , Proteínas de Ligação a RNA/genética , Adulto Jovem
8.
J Muscle Res Cell Motil ; 36(2): 195-203, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25487304

RESUMO

The sarcoplasmic/endoplasmic reticulum calcium ATPase 1 (SERCA1) has two muscle specific splice isoforms; SERCA1a in fast-type adult and SERCA1b in neonatal and regenerating skeletal muscles. At the protein level the only difference between these two isoforms is that SERCA1a has C-terminal glycine while SERCA1b has an octapeptide tail instead. This makes the generation of a SERCA1a specific antibody not feasible. The switch between the two isoforms is a hallmark of differentiation so we describe here a method based on the signal ratios of the SERCA1b specific and pan SERCA1 antibodies to estimate the SERCA1b/SERCA1a dominance on immunoblot of human muscles. Using this method we showed that unlike in mouse and rat, SERCA1b was only expressed in pre-matured infant leg and arm muscles; it was replaced by SERCA1a in more matured neonatal muscles and was completely absent in human foetal and neonatal diaphragms. Interestingly, only SERCA1a and no SERCA1b were detected in muscles of 7-12 years old boys with Duchenne, a degenerative-regenerative muscular dystrophy. However, in adult patients with myotonic dystrophy type 2 (DM2), the SERCA1b dominated over SERCA1a. Thus the human SERCA1b has a different expression pattern from that of rodents and it is associated with DM2.


Assuntos
Processamento Alternativo , Regulação Enzimológica da Expressão Gênica , Músculo Esquelético/enzimologia , Distrofia Muscular de Duchenne/enzimologia , Distrofia Miotônica/enzimologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , Adulto , Animais , Criança , Feminino , Humanos , Recém-Nascido , Isoenzimas/biossíntese , Isoenzimas/genética , Masculino , Camundongos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética
9.
RNA Biol ; 11(6): 742-54, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24824895

RESUMO

Expandable (CTG)n repeats in the 3' UTR of the DMPK gene are a cause of myotonic dystrophy type 1 (DM1), which leads to a toxic RNA gain-of-function disease. Mutant RNAs with expanded CUG repeats are retained in the nucleus and aggregate in discrete inclusions. These foci sequester splicing factors of the MBNL family and trigger upregulation of the CUGBP family of proteins resulting in the mis-splicing of their target transcripts. To date, many efforts to develop novel therapeutic strategies have been focused on disrupting the toxic nuclear foci and correcting aberrant alternative splicing via targeting mutant CUG repeats RNA; however, no effective treatment for DM1 is currently available. Herein, we present results of culturing of human DM1 myoblasts and fibroblasts with two small-molecule ATP-binding site-specific kinase inhibitors, C16 and C51, which resulted in the alleviation of the dominant-negative effects of CUG repeat expansion. Reversal of the DM1 molecular phenotype includes a reduction of the size and number of foci containing expanded CUG repeat transcripts, decreased steady-state levels of CUGBP1 protein, and consequent improvement of the aberrant alternative splicing of several pre-mRNAs misregulated in DM1.


Assuntos
Inibidores Enzimáticos/farmacologia , Distrofia Miotônica/genética , Fosfotransferases/antagonistas & inibidores , Processamento Alternativo , Proteínas CELF1 , Células Cultivadas , Inibidores Enzimáticos/química , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/enzimologia , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Precursores de RNA , Splicing de RNA , RNA Mensageiro , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica
10.
Nucleic Acids Res ; 42(11): 7186-200, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24792155

RESUMO

Myotonic dystrophy type 1 (DM1) is caused by CUG triplet expansions in the 3' UTR of dystrophia myotonica protein kinase (DMPK) messenger ribonucleic acid (mRNA). The etiology of this multi-systemic disease involves pre-mRNA splicing defects elicited by the ability of the CUG-expanded mRNA to 'sponge' splicing factors of the muscleblind family. Although nuclear aggregation of CUG-containing mRNPs in distinct foci is a hallmark of DM1, the mechanisms of their homeostasis have not been completely elucidated. Here we show that a DEAD-box helicase, DDX6, interacts with CUG triplet-repeat mRNA in primary fibroblasts from DM1 patients and with CUG-RNA in vitro. DDX6 overexpression relieves DM1 mis-splicing, and causes a significant reduction in nuclear DMPK-mRNA foci. Conversely, knockdown of endogenous DDX6 leads to a significant increase in DMPK-mRNA foci count and to increased sequestration of MBNL1 in the nucleus. While the level of CUG-expanded mRNA is unaffected by increased DDX6 expression, the mRNA re-localizes to the cytoplasm and its interaction partner MBNL1 becomes dispersed and also partially re-localized to the cytoplasm. Finally, we show that DDX6 unwinds CUG-repeat duplexes in vitro in an adenosinetriphosphate-dependent manner, suggesting that DDX6 can remodel and release nuclear DMPK messenger ribonucleoprotein foci, leading to normalization of pathogenic alternative splicing events.


Assuntos
Regiões 3' não Traduzidas , RNA Helicases DEAD-box/metabolismo , Distrofia Miotônica/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/metabolismo , Núcleo Celular/química , Células Cultivadas , Citoplasma/química , RNA Helicases DEAD-box/antagonistas & inibidores , Fibroblastos/química , Fibroblastos/metabolismo , Humanos , Distrofia Miotônica/enzimologia , Distrofia Miotônica/metabolismo , Miotonina Proteína Quinase , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Splicing de RNA , RNA Mensageiro/análise , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/análise , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Sequências Repetitivas de Ácido Nucleico , Expansão das Repetições de Trinucleotídeos
11.
Mol Ther ; 22(2): 312-320, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24196578

RESUMO

Myotonic dystrophy type 1 (DM1) is caused by the expansion of (CTG)n in the 3' untranslated region of the dystrophia myotonica-protein kinase (DMPK) gene, which is transcribed as (CUG)n repeats that accumulate in the nucleus. The RNA repeats specifically sequester or change the expression levels of several RNA-binding proteins, leading to aberrant splicing of many target genes. In this study, we developed artificial site-specific RNA endonucleases (ASREs) that specifically bind and cleave (CUG)n repeats RNA. We have generated one ASRE that can target the expanded RNA repeats in DM1 patient cells and specifically degrade the pathogenic DMPK messenger RNAs with minimal effect on wild-type alleles. Such ASRE treatment significantly decreased the number of nuclear foci in DM1 patient cells and can reverse the missplicing of many genes affected in DM1 patients. Taken together, the application of ASRE provides a new route of gene therapy for DM1 treatment.


Assuntos
Endorribonucleases/metabolismo , Distrofia Miotônica/genética , Repetições de Trinucleotídeos , Processamento Alternativo , Animais , Domínio Catalítico , Linhagem Celular , Endorribonucleases/química , Endorribonucleases/genética , Expressão Gênica , Humanos , Hidrólise , Distrofia Miotônica/enzimologia , Distrofia Miotônica/terapia , Ligação Proteica , Repetições de Trinucleotídeos/genética
12.
Proc Natl Acad Sci U S A ; 110(33): 13570-5, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23901116

RESUMO

Myotonic dystrophy type 1 (DM1) is caused by expansion of CTG repeats in the 3' UTR of the DMPK gene. Expression of CUG expansion (CUG(exp)) RNA produces a toxic gain of function by disrupting the functions of RNA splicing factors, such as MBNL1 and CELF1, leading to splicing changes associated with clinical abnormalities. Progressive skeletal muscle weakness and wasting is one of the most prominent clinical features in DM1; however, the underlying mechanisms remain unclear. Here we report that the embryonic M2 isoform of pyruvate kinase (PKM2), a key enzyme contributing to the Warburg effect in cancer, is significantly induced in DM1 tissue and mouse models owing to aberrant splicing. Expression of PKM2 in DM1 skeletal muscle is restricted to the type 1 fibers, which are particularly susceptible to wasting in DM1. Using antisense oligonucleotides to shift PKM splicing toward increased PKM2 expression, we observed increased glucose consumption with reduced oxidative metabolism in cell culture and increased respiratory exchange ratio in mice, suggesting defects in energy metabolism conferred by PKM2 expression. We propose that PKM2 expression induces changes in type 1 fibers associated with muscle atrophy and muscle weakness in DM1.


Assuntos
Processamento Alternativo/genética , Metabolismo Energético/genética , Glucose/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Distrofia Miotônica/enzimologia , Piruvato Quinase/metabolismo , Animais , Western Blotting , Calorimetria Indireta , Linhagem Celular , Humanos , Imuno-Histoquímica , Ácido Láctico/metabolismo , Camundongos , Fibras Musculares de Contração Lenta/patologia , Oligorribonucleotídeos Antissenso/genética , Consumo de Oxigênio/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Clin Invest ; 122(12): 4461-72, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23160194

RESUMO

Myotonic dystrophy type 1 (DM1) is a complex neuromuscular disease characterized by skeletal muscle wasting, weakness, and myotonia. DM1 is caused by the accumulation of CUG repeats, which alter the biological activities of RNA-binding proteins, including CUG-binding protein 1 (CUGBP1). CUGBP1 is an important skeletal muscle translational regulator that is activated by cyclin D3-dependent kinase 4 (CDK4). Here we show that mutant CUG repeats suppress Cdk4 signaling by increasing the stability and activity of glycogen synthase kinase 3ß (GSK3ß). Using a mouse model of DM1 (HSA(LR)), we found that CUG repeats in the 3' untranslated region (UTR) of human skeletal actin increase active GSK3ß in skeletal muscle of mice, prior to the development of skeletal muscle weakness. Inhibition of GSK3ß in both DM1 cell culture and mouse models corrected cyclin D3 levels and reduced muscle weakness and myotonia in DM1 mice. Our data predict that compounds normalizing GSK3ß activity might be beneficial for improvement of muscle function in patients with DM1.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Músculo Esquelético/enzimologia , Distrofia Miotônica/enzimologia , Adulto , Animais , Células CHO , Cricetinae , Ciclina D3/metabolismo , Estabilidade Enzimática , Feminino , Regulação Enzimológica da Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Lítio/farmacologia , Lítio/uso terapêutico , Masculino , Camundongos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Força Muscular/efeitos dos fármacos , Músculo Esquelético/patologia , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , Tiadiazóis/farmacologia , Tiadiazóis/uso terapêutico
14.
Biochem Biophys Res Commun ; 423(2): 212-7, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22609207

RESUMO

Myotonic dystrophy type 1 (DM1) is a multi-systemic disease with no established treatment to date. Small, cell-permeable molecules hold the potential to treat DM1. In this study, we investigated the association between protein kinase C (PKC) signaling and splicing of sarcoplasmic reticulum Ca(2+)-ATPase1 (SERCA1). Our aim was to clarify the mechanisms underlying the regulation of alternative splicing, in order to explore new therapeutic strategies for DM1. By assessing the splicing pattern of the endogenous SERCA1 gene in HEK293 cells, we found that treatment with phorbol 12-myristate 13-acetate (PMA) regulated SERCA1 splicing. Interestingly, treatment with PMA for 48 h normalized SERCA1 splicing, while treatment for 1.5h promoted aberrant splicing. These two responses showed dose dependency and were completely abolished by the PKC inhibitor Ro 31-8220. Furthermore, repression of PKCßII and PKCθ by RNAi mimicked prolonged PMA treatment. These results indicate that PKC signaling is involved in the splicing of SERCA1 and provide new evidence for a link between alternative splicing and PKC signaling.


Assuntos
Processamento Alternativo/fisiologia , Distrofia Miotônica/enzimologia , Proteína Quinase C-delta/metabolismo , Proteína Quinase C/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Processamento Alternativo/efeitos dos fármacos , Éxons/genética , Células HEK293 , Humanos , Indóis/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C beta , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia
15.
FASEB J ; 26(7): 3042-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22459146

RESUMO

Phosphoinositide-phospholipase C ß1 (PLCß1) plays a crucial role in the initiation of the genetic program responsible for muscle differentiation. We previously demonstrated that nuclear PLCß1 activates the cyclin D3 promoter during the differentiation of myoblasts to myotubes, indicating that PLCß1 is essential for cyclin D3 promoter activation and gene transcription, through c-jun/AP1. Myotonic dystrophy (DM) is the most prevalent form of muscular dystrophy in adults. DM type 1 (DM1) and type 2 (DM2) are dominantly inherited multisystem disorders. DM1 is triggered by the pathological expansion of a (CTG)(n) triplet repeat in the gene coding for DMPK, the dystrophia myotonica-protein kinase, whereas a (CCTG)(n) tetranucleotide repeat expansion in the ZNF9 gene, encoding a CCHC-type zinc finger protein, causes DM2. We found that, unlike in normal myotubes, the level of expression of PLCß1 in DM1 and DM2 cells was already elevated in proliferating cells. Treatment with insulin induced a dramatic decrease in the amount of PLCß1. During differentiation, cyclin D3 and myogenin were elevated in normal myotubes, whereas differentiating DM1 and DM2 cells did not increase these proteins. Forced expression of PLCß1 in DM1 and DM2 cells increased the expression of differentiation markers, myogenin and cyclin D3, and enhanced fusion of DM myoblasts. These results highlight again that PLCß1 expression is a key player in myoblast differentiation, functioning as a positive regulator in the correction of delayed differentiation of skeletal muscle in DM human myoblasts.


Assuntos
Transtornos Miotônicos/enzimologia , Transtornos Miotônicos/genética , Distrofia Miotônica/enzimologia , Distrofia Miotônica/genética , Fosfolipase C beta/genética , Fosfolipase C beta/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Ciclina D3/genética , Ciclina D3/metabolismo , Perfilação da Expressão Gênica , Humanos , Insulina/farmacologia , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/enzimologia , Mioblastos Esqueléticos/patologia , Miogenina/genética , Miogenina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção , Regulação para Cima
16.
J Biol Chem ; 286(46): 40296-306, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21949239

RESUMO

Myotonic dystrophy 1 (DM1) is a multisystemic disease caused by a triplet nucleotide repeat expansion in the 3' untranslated region of the gene coding for myotonic dystrophy protein kinase (DMPK). DMPK is a nuclear envelope (NE) protein that promotes myogenic gene expression in skeletal myoblasts. Muscular dystrophy research has revealed the NE to be a key determinant of nuclear structure, gene regulation, and muscle function. To investigate the role of DMPK in NE stability, we analyzed DMPK expression in epithelial and myoblast cells. We found that DMPK localizes to the NE and coimmunoprecipitates with Lamin-A/C. Overexpression of DMPK in HeLa cells or C2C12 myoblasts disrupts Lamin-A/C and Lamin-B1 localization and causes nuclear fragmentation. Depletion of DMPK also disrupts NE lamina, showing that DMPK is required for NE stability. Our data demonstrate for the first time that DMPK is a critical component of the NE. These novel findings suggest that reduced DMPK may contribute to NE instability, a common mechanism of skeletal muscle wasting in muscular dystrophies.


Assuntos
Células Epiteliais/enzimologia , Proteínas Musculares/metabolismo , Mioblastos Esqueléticos/enzimologia , Distrofia Miotônica/enzimologia , Membrana Nuclear/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Células Epiteliais/patologia , Regulação da Expressão Gênica/genética , Células HeLa , Humanos , Laminas/genética , Laminas/metabolismo , Proteínas Musculares/genética , Mioblastos Esqueléticos/patologia , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Miotonina Proteína Quinase , Membrana Nuclear/genética , Membrana Nuclear/patologia , Proteínas Serina-Treonina Quinases/genética
17.
Clin Biochem ; 43(15): 1246-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20691173

RESUMO

OBJECTIVES: Elevation of serum gamma-glutamyltransferase (GGT), in absence of a clinically significant liver damage, is often found in Myotonic Dystrophy type-1 (DM1). In this study we investigated if a specific GGT fraction pattern is present in DM1. DESIGNS AND METHODS: We compared total and fractional GGT values (b-, m-, s-, f-GGT) among patients with DM1 or liver disease (LD) and healthy subjects (HS). RESULTS: The increase of GGT in DM1 and LD, vs HS, was mainly due to s-GGT (median: 32.7; 66.7; and 7.9 U/L, respectively), and b-GGT (8.5; 18.9; and 2.1 U/L). The subset of DM1 patients matched with HS with corresponding serum GGT showed higher b-GGT (6.0 vs 4.2 U/L). CONCLUSIONS: DM1 patients with normal total GGT values showed an alteration of the production and release in the blood of GGT fractions. Since increased s-GGT is also found in LD, a sub-clinical liver damage likely occurs in DM1 subjects apparently free of liver disease.


Assuntos
Saúde , Hepatopatias/sangue , Hepatopatias/enzimologia , Distrofia Miotônica/sangue , Distrofia Miotônica/enzimologia , gama-Glutamiltransferase/sangue , Adulto , Estudos de Casos e Controles , Fracionamento Químico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
18.
Muscle Nerve ; 41(6): 758-62, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20513102

RESUMO

The purpose of this study was to describe the frequency of absent, unrecognized, or minimal myotonic discharges (MDs) in myotonic dystrophy type 2 (DM2). We performed a retrospective review of needle electromyography (EMG) data prior to genetic diagnosis in 49 DM2 patients at the Mayo Clinic. MDs were not reported on first or repeat EMG studies (n = 8) and not found in archived recordings of 4 patients (8%); archived EMG recordings (n = 4) confirmed the absence of MDs (n = 2), including 1 patient with normal insertional activity in all muscles, and misinterpretation of MDs as slow fibrillation potentials (n = 1) and complex repetitive discharge (CRD) activity (n = 1). Eight (16%) patients had minimal classic MDs with diffusely increased insertional activity, including waning-only MDs in all patients in this group with archived EMG recordings (n = 5). Diffuse MDs were found in 33 (67%) patients. Absent or minimal MDs do not exclude DM2. Over-reliance on diffuse MDs in patients who present with myopathy may lead to delay in genetic diagnosis of DM2.


Assuntos
Transtornos Miotônicos/genética , Distrofia Miotônica/fisiopatologia , Adulto , Idoso , Cromossomos Humanos Par 3 , Creatina Quinase/genética , Creatina Quinase/metabolismo , Expansão das Repetições de DNA , Eletromiografia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Contração Muscular , Músculo Esquelético/fisiopatologia , Transtornos Miotônicos/enzimologia , Transtornos Miotônicos/fisiopatologia , Distrofia Miotônica/classificação , Distrofia Miotônica/enzimologia , Distrofia Miotônica/genética , Estudos Retrospectivos
20.
Biochim Biophys Acta ; 1804(6): 1334-41, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20188867

RESUMO

Myotonic dystrophy type 1 (DM1) is caused by an expansion of CTG repeats at the 3'-UTR of the serine/threonine protein kinase DMPK. Expanded CTG repeats are toxic since they are transcribed into an RNA molecule which is then sequestered within the nucleus in the form of foci. RNA cytotoxicity is linked to the aberrant splicing of several developmentally regulated genes. DMPK transcripts undergo alternative splicing giving rise to many isoforms but do not seem to be involved in the splicing dysregulation of DM1. However, decreased levels of DMPK in DM1 patients and DMPK involvement in muscle weakness and cardiac dysfunction in animal models have been reported. The variability in phenotypic expression of DMPK together with its differential subcellular targeting, suggests that different splicing isoforms may be involved in different signalling pathways, possibly through DMPK-interacting proteins. To gain better insight into the DMPK function, we used mass spectrometry to identify proteins co-segregating with DMPK in soluble complexes isolated from high-speed supernatant of rat muscles. We carried out experiments with native DMPK to preserve the physiological stoichiometry with potential partners. DMPK-containing complexes were isolated and immuno-detected by non-denaturing electrophoresis, gel filtration, ionic-exchange chromatography and immunoprecipitation. DMPK peptides were identified by high-resolution mass spectrometry together with several putative DMPK-binding proteins, including several heat shock proteins such as HSP20/HSPB6, HSP60/CPN60, HSP70 and HSP90. We also obtained evidence of a direct interaction of DMPK with alphaB-crystallin/HSPB5 and HSP25/HSPB1.


Assuntos
Proteínas de Choque Térmico/metabolismo , Complexos Multienzimáticos/metabolismo , Músculo Esquelético/enzimologia , Peptídeos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Regiões 3' não Traduzidas/genética , Processamento Alternativo/genética , Animais , Núcleo Celular/enzimologia , Núcleo Celular/genética , Núcleo Celular/patologia , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Corpos de Inclusão Intranuclear/genética , Corpos de Inclusão Intranuclear/metabolismo , Corpos de Inclusão Intranuclear/patologia , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Espectrometria de Massas , Complexos Multienzimáticos/química , Complexos Multienzimáticos/genética , Músculo Esquelético/patologia , Distrofia Miotônica/enzimologia , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Miotonina Proteína Quinase , Peptídeos/química , Peptídeos/genética , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Ratos , Ratos Wistar , Transcrição Gênica/genética , Expansão das Repetições de Trinucleotídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...