Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Front Genet ; 14: 1216066, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37576554

RESUMO

Muscle damage and fibro-fatty replacement of skeletal muscles is a main pathologic feature of Duchenne muscular dystrophy (DMD) with more proximal muscles affected earlier and more distal affected later in the disease course, suggesting that different skeletal muscle groups possess distinctive characteristics that influence their susceptibility to disease. To explore transcriptomic factors driving differential gene expression and modulating DMD skeletal muscle severity, we characterized the transcriptome of vastus lateralis (VL), a more proximal and susceptible muscle, relative to tibialis anterior (TA), a more distal and protected muscle, in 15 healthy individuals using bulk RNA sequencing to identify gene expression differences that may mediate their relative susceptibility to damage with loss of dystrophin. Matching single nuclei RNA sequencing data was generated for 3 of the healthy individuals, to infer cell composition in the bulk RNA sequencing dataset and to improve mapping of differentially expressed genes to their cell source of expression. A total of 3,410 differentially expressed genes were identified and mapped to cell type using single nuclei RNA sequencing of muscle, including long non-coding RNAs and protein coding genes. There was an enrichment of genes involved in calcium release from the sarcoplasmic reticulum, particularly in the myofibers and these myofiber genes were higher in the VL. There was an enrichment of genes in "Collagen-Containing Extracellular Matrix" expressed by fibroblasts, endothelial, smooth muscle and pericytes, with most genes higher in the TA, as well as genes in "Regulation Of Apoptotic Process" expressed across all cell types. Previously reported genetic modifiers were also enriched within the differentially expressed genes. We also identify 6 genes with differential isoform usage between the VL and TA. Lastly, we integrate our findings with DMD RNA sequencing data from the TA, and identify "Collagen-Containing Extracellular Matrix" and "Negative Regulation Of Apoptotic Process" as differentially expressed between DMD compared to healthy. Collectively, these findings propose novel candidate mechanisms that may mediate differential muscle susceptibility in muscular dystrophies and provide new insight into potential therapeutic targets.

2.
Commun Biol ; 5(1): 989, 2022 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-36123393

RESUMO

In Duchenne muscular dystrophy, dystrophin loss leads to chronic muscle damage, dysregulation of repair, fibro-fatty replacement, and weakness. We develop methodology to efficiently isolate individual nuclei from minute quantities of frozen skeletal muscle, allowing single nuclei sequencing of irreplaceable archival samples and from very small samples. We apply this method to identify cell and gene expression dynamics within human DMD and mdx mouse muscle, characterizing effects of dystrophin rescue by exon skipping therapy at single nuclei resolution. DMD exon 23 skipping events are directly observed and increased in myonuclei from treated mice. We describe partial rescue of type IIa and IIx myofibers, expansion of an MDSC-like myeloid population, recovery of repair/remodeling M2-macrophage, and repression of inflammatory POSTN1 + fibroblasts in response to exon skipping and partial dystrophin restoration. Use of this method enables exploration of cellular and transcriptomic mechanisms of dystrophin loss and repair within an intact muscle environment. Our initial findings will scaffold our future work to more directly examine muscular dystrophies and putative recovery pathways.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Animais , Distrofina/genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Transcriptoma
3.
Front Cell Dev Biol ; 10: 830415, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35465312

RESUMO

In vitro models of patient-derived muscle allow for more efficient development of genetic medicines for the muscular dystrophies, which often present mutation-specific pathologies. One popular strategy to generate patient-specific myotubes involves reprogramming dermal fibroblasts to a muscle lineage through MyoD induction. However, creating physiologically relevant, reproducible tissues exhibiting multinucleated, aligned myotubes with organized striations is dependent on the introduction of physicochemical cues that mimic the native muscle microenvironment. Here, we engineered patient-specific control and dystrophic muscle tissues in vitro by culturing and differentiating MyoD-directly reprogrammed fibroblasts isolated from one healthy control subject, three patients with Duchenne muscular dystrophy (DMD), and two Limb Girdle 2A/R1 (LGMD2A/R1) patients on micromolded gelatin hydrogels. Engineered DMD and LGMD2A/R1 tissues demonstrated varying levels of defects in α-actinin expression and organization relative to control, depending on the mutation. In genetically relevant DMD tissues amenable to mRNA reframing by targeting exon 44 or 45 exclusion, exposure to exon skipping antisense oligonucleotides modestly increased myotube coverage and alignment and rescued dystrophin protein expression. These findings highlight the value of engineered culture substrates in guiding the organization of reprogrammed patient fibroblasts into aligned muscle tissues, thereby extending their value as tools for exploration and dissection of the cellular and molecular basis of genetic muscle defects, rescue, and repair.

4.
Sci Rep ; 11(1): 1128, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441839

RESUMO

Emerging and promising therapeutic interventions for Duchenne muscular dystrophy (DMD) are confounded by the challenges of quantifying dystrophin. Current approaches have poor precision, require large amounts of tissue, and are difficult to standardize. This paper presents an immuno-mass spectrometry imaging method using gadolinium (Gd)-labeled anti-dystrophin antibodies and laser ablation-inductively coupled plasma-mass spectrometry to simultaneously quantify and localize dystrophin in muscle sections. Gd is quantified as a proxy for the relative expression of dystrophin and was validated in murine and human skeletal muscle sections following k-means clustering segmentation, before application to DMD patients with different gene mutations where dystrophin expression was measured up to 100 µg kg-1 Gd. These results demonstrate that immuno-mass spectrometry imaging is a viable approach for pre-clinical to clinical research in DMD. It rapidly quantified relative dystrophin in single tissue sections, efficiently used valuable patient resources, and may provide information on drug efficacy for clinical translation.


Assuntos
Distrofina/análise , Distrofia Muscular de Duchenne/metabolismo , Músculo Quadríceps/química , Adolescente , Idoso de 80 Anos ou mais , Animais , Criança , Distrofina/genética , Distrofina/imunologia , Feminino , Imunofluorescência , Gadolínio , Humanos , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Camundongos , Fibras Musculares Esqueléticas/química , Distrofia Muscular de Duchenne/genética , Mutação
5.
Muscle Nerve ; 62(6): 688-698, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32820569

RESUMO

Serial muscle biopsies within clinical trials for Duchenne muscular dystrophy (DMD) are critical to document therapeutic responses. Less invasive means of sampling muscle are needed. We analyzed a retrospective consecutive case-series cohort of vacuum-assisted core needle muscle biopsy procedures performed on healthy and dystrophic individuals at a single institution assessing for safety and reliability of obtaining sufficient high-quality biopsy tissue for histologic assessment in adult and pediatric subjects. Of 471 muscle cores from 128 biopsy procedures, 377-550 mg of total muscle tissue was obtained per procedure with mean core weight of 129 mg (SD, 25.1 mg). All biopsies were adequate for histological assessment. There were no significant adverse events. This core needle biopsy approach, when combined with improved sample processing, provides a safe means to consistently obtain muscle samples for diagnostic and clinical trial applications.


Assuntos
Biópsia com Agulha de Grande Calibre/métodos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Adolescente , Adulto , Idoso , Anestésicos Locais/uso terapêutico , Biópsia com Agulha de Grande Calibre/instrumentação , Estudos de Casos e Controles , Criança , Pré-Escolar , Sedação Consciente , Feminino , Humanos , Biópsia Guiada por Imagem , Masculino , Pessoa de Meia-Idade , Dor Processual/prevenção & controle , Reprodutibilidade dos Testes , Manejo de Espécimes/métodos , Preservação de Tecido/métodos , Ultrassonografia , Vácuo , Adulto Jovem
6.
Neuromuscul Disord ; 29(11): 863-873, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31672265

RESUMO

Duchenne muscular dystrophy is caused by mutations in the dystrophin-encoding DMD gene. While Duchenne is most commonly caused by large intragenic deletions that cause frameshift and complete loss of dystrophin expression, in-frame deletions in DMD can result in the expression of internally truncated dystrophin proteins and may be associated with a milder phenotype. In this study, we describe two individuals with large in-frame 5' deletions (exon 3-23 and exon 3-28) that remove the majority of the N-terminal region, including part of the actin binding and central rod domains. Both patients had progressive muscle weakness during childhood but are observed to have a relatively mild disease course compared to typical Duchenne. We show that in muscle biopsies from both patients, truncated dystrophin is expressed at the sarcolemma. We have additionally developed a patient-specific fibroblast-derived cell model, which can be inducibly reprogrammed to form myotubes that largely recapitulate biopsy findings for the patient with the exon 3-23 deletion, providing a culture model for future investigation of this unusual case. We discuss these mutations in the context of previously reported 5' in-frame DMD deletions and relevant animal models, and review the spectrum of phenotypes associated with these deletions.


Assuntos
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Deleção de Sequência , Adolescente , Células Cultivadas , Criança , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Fenótipo , Índice de Gravidade de Doença
7.
Mol Ther Nucleic Acids ; 18: 580-589, 2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-31678734

RESUMO

Systemic delivery of antisense oligonucleotides (AO) for DMD exon skipping has proven effective for reframing DMD mRNA, rescuing dystrophin expression, and slowing disease progression in animal models. In humans with Duchenne muscular dystrophy treated with AOs, low levels of dystrophin have been induced, and modest slowing of disease progression has been observed, highlighting the need for improved efficiency of human skipping drugs. Here, we demonstrate that dantrolene and Rycals S107 and ARM210 potentiate AO-mediated exon skipping of exon 44 or exon 45 in patient-derived myotube cultures with appropriate mutations. Further, dantrolene is shown to boost AO-mediated exon skipping in patient-derived, induced cardiomyocyte cultures. Our findings further validate the ryanodine receptors (RyR) as the likely target responsible for exon skip boosting and demonstrate potential applicability beyond exon 51 skipping. These data provide preclinical support of dantrolene trial as an adjuvant to AO-mediated exon-skipping therapy in humans and identify a novel Rycal, ARM210, for development as a potential exon-skipping booster. Further, they highlight the value of mutation-specific DMD culture models for basic discovery, preclinical drug screening and translation of personalized genetic medicines.

8.
Methods Mol Biol ; 1828: 309-326, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171550

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene. Most deletions, duplications, or indels lead to shift of mRNA reading frame, which prevent the production of dystrophin protein. DMD is the leading fatal genetic disorder in childhood. One therapeutic strategy aims to skip one or more exons to restore reading frame to enable the production of internally truncated proteins with partial functionality. However, to date the efficiency of this strategy is suboptimal. Here we present methods for assessing exon skipping using AON alone or in combination with skip booster in the context of human DMD patient fibroblast derived myotubes and in the mdx mouse model of DMD.


Assuntos
Distrofina/genética , Éxons , Distrofia Muscular de Duchenne/genética , Splicing de RNA , Animais , Reprogramação Celular/genética , Modelos Animais de Doenças , Fibroblastos/metabolismo , Terapia Genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos mdx , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/terapia , Mutação , Proteína MyoD/genética , Proteína MyoD/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Hum Mutat ; 39(9): 1193-1202, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29907980

RESUMO

Antisense oligonucleotide (AON)-mediated exon skipping is an emerging therapeutic for individuals with Duchenne muscular dystrophy (DMD). Skipping of exons adjacent to common exon deletions in DMD using AONs can produce in-frame transcripts and functional protein. Targeted skipping of DMD exons 8, 44, 45, 50, 51, 52, 53, and 55 is predicted to benefit 47% of affected individuals. We observed a correlation between mutation subgroups and age at loss of ambulation in the Duchenne Registry, a large database of phenotypic and genetic data for DMD (N = 765). Males amenable to exon 44 (N = 74) and exon 8 skipping (N = 18) showed prolonged ambulation compared to other exon skip groups and nonsense mutations (P = 0.035 and P < 0.01, respectively). In particular, exon 45 deletions were associated with prolonged age at loss of ambulation relative to the rest of the exon 44 skip amenable cohort and other DMD mutations. Exon 3-7 deletions also showed prolonged ambulation relative to all other exon 8 skippable mutations. Cultured myotubes from DMD patients with deletions of exons 3-7 or exon 45 showed higher endogenous skipping than other mutations, providing a potential biological rationale for our observations. These results highlight the utility of aggregating phenotypic and genotypic data for rare pediatric diseases to reveal progression differences, identify potentially confounding factors, and probe molecular mechanisms that may affect disease severity.


Assuntos
Distrofina/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Oligodesoxirribonucleotídeos Antissenso/genética , Adolescente , Adulto , Fatores Etários , Biópsia , Códon sem Sentido/genética , Distrofina/antagonistas & inibidores , Éxons/genética , Feminino , Fibroblastos/patologia , Genótipo , Humanos , Estimativa de Kaplan-Meier , Tempo de Internação , Masculino , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Mioblastos/patologia , Oligodesoxirribonucleotídeos Antissenso/uso terapêutico , Cultura Primária de Células , Sistema de Registros , Deleção de Sequência/genética , Caracteres Sexuais , Adulto Jovem
10.
Mol Ther Nucleic Acids ; 11: 180-191, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29858053

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutations in DMD, resulting in loss of dystrophin, which is essential to muscle health. DMD "exon skipping" uses anti-sense oligo-nucleotides (AONs) to force specific exon exclusion during mRNA processing to restore reading frame and rescue of partially functional dystrophin protein. Although exon-skipping drugs in humans show promise, levels of rescued dystrophin protein remain suboptimal. We previously identified dantrolene as a skip booster when combined with AON in human DMD cultures and short-term mdx dystrophic mouse studies. Here, we assess the effect of dantrolene/AON combination on DMD exon-23 skipping over long-term mdx treatment under conditions that better approximate potential human dosing. To evaluate the dantrolene/AON combination treatment effect on dystrophin induction, we assayed three AON doses, with and without oral dantrolene, to assess multiple outcomes across different muscles. Meta-analyses of the results of statistical tests from both the quadriceps and diaphragm assessing contributions of dantrolene beyond AON, across all AON treatment groups, provide strong evidence that dantrolene modestly boosts exon skipping and dystrophin rescue while reducing muscle pathology in mdx mice (p < 0.0087). These findings support a trial of combination dantrolene/AON to increase exon-skipping efficacy and highlight the value of combinatorial approaches and Food and Drug Administration (FDA) drug re-purposing for discovery of unsuspected therapeutic application and rapid translation.

11.
Neurology ; 89(17): 1811-1820, 2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-28972192

RESUMO

OBJECTIVE: To conduct a randomized trial to test the primary hypothesis that once-daily tadalafil, administered orally for 48 weeks, lessens the decline in ambulatory ability in boys with Duchenne muscular dystrophy (DMD). METHODS: Three hundred thirty-one participants with DMD 7 to 14 years of age taking glucocorticoids were randomized to tadalafil 0.3 mg·kg-1·d-1, tadalafil 0.6 mg·kg-1·d-1, or placebo. The primary efficacy measure was 6-minute walk distance (6MWD) after 48 weeks. Secondary efficacy measures included North Star Ambulatory Assessment and timed function tests. Performance of Upper Limb (PUL) was a prespecified exploratory outcome. RESULTS: Tadalafil had no effect on the primary outcome: 48-week declines in 6MWD were 51.0 ± 9.3 m with placebo, 64.7 ± 9.8 m with low-dose tadalafil (p = 0.307 vs placebo), and 59.1 ± 9.4 m with high-dose tadalafil (p = 0.538 vs placebo). Tadalafil also had no effect on secondary outcomes. In boys >10 years of age, total PUL score and shoulder subscore declined less with low-dose tadalafil than placebo. Adverse events were consistent with the known safety profile of tadalafil and the DMD disease state. CONCLUSIONS: Tadalafil did not lessen the decline in ambulatory ability in boys with DMD. Further studies should be considered to confirm the hypothesis-generating upper limb data and to determine whether ambulatory decline can be slowed by initiation of tadalafil before 7 years of age. CLINICALTRIALSGOV IDENTIFIER: NCT01865084. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that tadalafil does not slow ambulatory decline in 7- to 14-year-old boys with Duchenne muscular dystrophy.


Assuntos
Distrofia Muscular de Duchenne/tratamento farmacológico , Tadalafila/uso terapêutico , Vasodilatadores/uso terapêutico , Adolescente , Área Sob a Curva , Criança , Relação Dose-Resposta a Droga , Método Duplo-Cego , Seguimentos , Glucocorticoides/uso terapêutico , Frequência Cardíaca/fisiologia , Humanos , Cooperação Internacional , Masculino , Distrofia Muscular de Duchenne/psicologia , Qualidade de Vida , Testes de Função Respiratória , Resultado do Tratamento , Função Ventricular Esquerda , Caminhada/fisiologia
12.
Glycobiology ; 27(12): 1134-1143, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973355

RESUMO

Our understanding of muscle glycosylation to date has derived from studies in mouse models and a limited number of human lectin histochemistry studies. As various therapeutic approaches aimed at treating patients with muscular dystrophies are being translated from rodent models to human, it is critical to better understand human muscle glycosylation and relevant disease-specific differences between healthy and dystrophic muscle. Here, we report the first quantitative characterization of human muscle glycosylation, and identify differentiation- and disease-specific differences in human muscle glycosylation. Utilizing a panel of 13 lectins with varying glycan specificities, we surveyed lectin binding to primary and immortalized myoblasts and myotubes from healthy and dystrophic sources. Following differentiation of primary and immortalized healthy human muscle cells, we observed increased binding of Narcissus pseudonarcissus agglutinin (NPA), PNA, MAA-II and WFA to myotubes compared to myoblasts. Following differentiation of immortalized healthy and dystrophic human muscle cells, we observed disease-specific differences in binding of NPA, Jac and Tricosanthes japonica agglutinin-I (TJA-I) to differentiated myotubes. We also observed differentiation- and disease-specific differences in binding of NPA, Jac, PNA, TJA-I and WFA to glycoprotein receptors in muscle cells. Additionally, Jac, PNA and WFA precipitated functionally glycosylated α-DG, that bound laminin, while NPA and TJA-I did not. Lectin histochemistry of healthy and dystrophic human muscle sections identified disease-specific differences in binding of O-glycan and sialic acid-specific lectins between healthy and dystrophic muscle. These results indicate that specific and discrete changes in glycosylation occur following differentiation, and identify specific lectins as potential biomarkers sensitive to changes in healthy human muscle glycosylation.


Assuntos
Glicoproteínas/metabolismo , Proteínas Musculares/metabolismo , Distrofias Musculares/metabolismo , Mioblastos Esqueléticos/metabolismo , Narcissus/química , Lectinas de Plantas/farmacologia , Linhagem Celular Transformada , Glicoproteínas/química , Humanos , Proteínas Musculares/química , Distrofias Musculares/patologia , Mioblastos Esqueléticos/química , Mioblastos Esqueléticos/patologia , Lectinas de Plantas/química
13.
JAMA ; 317(14): 1480, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28399245
14.
J Cell Biol ; 213(2): 275-88, 2016 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-27091452

RESUMO

In the degenerative disease Duchenne muscular dystrophy, inflammatory cells enter muscles in response to repetitive muscle damage. Immune factors are required for muscle regeneration, but chronic inflammation creates a profibrotic milieu that exacerbates disease progression. Osteopontin (OPN) is an immunomodulator highly expressed in dystrophic muscles. Ablation of OPN correlates with reduced fibrosis and improved muscle strength as well as reduced natural killer T (NKT) cell counts. Here, we demonstrate that the improved dystrophic phenotype observed with OPN ablation does not result from reductions in NKT cells. OPN ablation skews macrophage polarization toward a pro-regenerative phenotype by reducing M1 and M2a and increasing M2c subsets. These changes are associated with increased expression of pro-regenerative factors insulin-like growth factor 1, leukemia inhibitory factor, and urokinase-type plasminogen activator. Furthermore, altered macrophage polarization correlated with increases in muscle weight and muscle fiber diameter, resulting in long-term improvements in muscle strength and function in mdx mice. These findings suggest that OPN ablation promotes muscle repair via macrophage secretion of pro-myogenic growth factors.


Assuntos
Macrófagos/metabolismo , Distrofia Muscular Animal/patologia , Osteopontina/fisiologia , Animais , Polaridade Celular , Macrófagos/citologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculos/metabolismo , Músculos/patologia , Músculos/fisiologia , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/fisiologia , Osteopontina/genética , Osteopontina/metabolismo , Fenótipo , Regeneração
16.
Cell Stem Cell ; 18(4): 533-40, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26877224

RESUMO

Mutations in DMD disrupt the reading frame, prevent dystrophin translation, and cause Duchenne muscular dystrophy (DMD). Here we describe a CRISPR/Cas9 platform applicable to 60% of DMD patient mutations. We applied the platform to DMD-derived hiPSCs where successful deletion and non-homologous end joining of up to 725 kb reframed the DMD gene. This is the largest CRISPR/Cas9-mediated deletion shown to date in DMD. Use of hiPSCs allowed evaluation of dystrophin in disease-relevant cell types. Cardiomyocytes and skeletal muscle myotubes derived from reframed hiPSC clonal lines had restored dystrophin protein. The internally deleted dystrophin was functional as demonstrated by improved membrane integrity and restoration of the dystrophin glycoprotein complex in vitro and in vivo. Furthermore, miR31 was reduced upon reframing, similar to observations in Becker muscular dystrophy. This work demonstrates the feasibility of using a single CRISPR pair to correct the reading frame for the majority of DMD patients.


Assuntos
Sistemas CRISPR-Cas/genética , Distrofina/metabolismo , Deleção de Genes , Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Animais , Distrofina/deficiência , Distrofina/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Camundongos SCID , Músculo Esquelético/citologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia
17.
PLoS One ; 10(7): e0133353, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26186728

RESUMO

Functionally diverse CD8+ T cells develop in response to antigenic stimulation with differing capacities to couple TCR engagement to downstream signals and functions. However, mechanisms of diversifying TCR signaling are largely uncharacterized. Here we identified two alternative splice variants of scaffold protein Dlg1, Dlg1AB and Dlg1B, that diversify signaling to regulate p38-dependent and -independent effector functions in CD8+ T cells. Dlg1AB, but not Dlg1B associated with Lck, coupling TCR stimulation to p38 activation and proinflammatory cytokine production. Conversely, both Dlg1AB and Dlg1B mediated p38-independent degranulation. Degranulation depended on a Dlg1 fragment containing an intact Dlg1SH3-domain and required the SH3-ligand WASp. Further, Dlg1 controlled WASp activation by promoting TCR-triggered conformational opening of WASp. Collectively, our data support a model where Dlg1 regulates p38-dependent proinflammatory cytokine production and p38-independent cytotoxic granule release through the utilization of alternative splice variants, providing a mechanism whereby TCR engagement couples downstream signals to unique effector functions in CD8+ T cells.


Assuntos
Processamento Alternativo/genética , Linfócitos T CD8-Positivos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Actinas/metabolismo , Animais , Linfócitos T CD8-Positivos/enzimologia , Degranulação Celular , Citocinas/genética , Proteína 1 Homóloga a Discs-Large , Ativação Enzimática , Técnicas de Silenciamento de Genes , Granzimas/genética , Mediadores da Inflamação/metabolismo , Interferon gama/genética , Interleucina-2/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Camundongos , Fatores de Transcrição NFATC/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Polimerização , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Associadas SAP90-PSD95 , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
18.
Am J Hum Genet ; 96(3): 498-506, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25728775

RESUMO

Chromatin remodeling through histone acetyltransferase (HAT) and histone deactylase (HDAC) enzymes affects fundamental cellular processes including the cell-cycle, cell differentiation, metabolism, and apoptosis. Nonsense mutations in genes that are involved in histone acetylation and deacetylation result in multiple congenital anomalies with most individuals displaying significant developmental delay, microcephaly and dysmorphism. Here, we report a syndrome caused by de novo heterozygous nonsense mutations in KAT6A (a.k.a., MOZ, MYST3) identified by clinical exome sequencing (CES) in four independent families. The same de novo nonsense mutation (c.3385C>T [p.Arg1129∗]) was observed in three individuals, and the fourth individual had a nearby de novo nonsense mutation (c.3070C>T [p.Arg1024∗]). Neither of these variants was present in 1,815 in-house exomes or in public databases. Common features among all four probands include primary microcephaly, global developmental delay including profound speech delay, and craniofacial dysmorphism, as well as more varied features such as feeding difficulties, cardiac defects, and ocular anomalies. We further demonstrate that KAT6A mutations result in dysregulation of H3K9 and H3K18 acetylation and altered P53 signaling. Through histone and non-histone acetylation, KAT6A affects multiple cellular processes and illustrates the complex role of acetylation in regulating development and disease.


Assuntos
Códon sem Sentido/genética , Deficiências do Desenvolvimento/genética , Histona Acetiltransferases/genética , Microcefalia/genética , Anormalidades Múltiplas/genética , Acetilação , Pré-Escolar , Exoma , Feminino , Heterozigoto , Histona Acetiltransferases/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Mutação , Linhagem
19.
J Immunol ; 193(6): 2651-60, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25098293

RESUMO

CD8(+) T cells respond to TCR stimulation by producing proinflammatory cytokines, and destroying infected or malignant cells through the production and release of cytotoxic granules. Scaffold protein Discs large homolog 1 (Dlg1) specifies TCR-dependent functions by channeling proximal signals toward the activation of p38-dependent proinflammatory cytokine gene expression and/or p38-independent cytotoxic granule release. Two Dlg1 variants are expressed in CD8(+) T cells via alternative splicing, Dlg1AB and Dlg1B, which have differing abilities coordinate TCR-dependent functions. Although both variants facilitate p38-independent cytotoxicity, only Dlg1AB coordinates p38-dependent proinflammatory cytokine expression. In this study, we identify TCR-induced Dlg1 tyrosine phosphorylation as a key regulatory step required for Dlg1AB-mediated p38-dependent functions, including proinflammatory cytokine expression. We find that Dlg1AB but not Dlg1B is tyrosine phosphorylated by proximal tyrosine kinase Lck in response to TCR stimulation. Furthermore, we identify Dlg1 tyrosine 222 (Y222) as a major site of Dlg1 phosphorylation required for TCR-triggered p38 activation and NFAT-dependent expression of proinflammatory cytokines, but not for p38-independent cytotoxicity. Taken together, our data support a model where TCR-induced phosphorylation of Dlg1 Y222 is a key point of control that endows Dlg1AB with the ability to coordinate p38 activation and proinflammatory cytokine production. We propose blocking Dlg1AB phosphorylation as a novel therapeutic target to specifically block proinflammatory cytokine production but not cytotoxicity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citocinas/biossíntese , Proteínas do Tecido Nervoso/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Processamento Alternativo , Sequência de Aminoácidos , Animais , Comunicação Celular/imunologia , Proteína 1 Homóloga a Discs-Large , Ativação Enzimática/imunologia , Inflamação/imunologia , Ativação Linfocitária/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Fosforilação , Isoformas de Proteínas/imunologia , Proteínas Associadas SAP90-PSD95 , Alinhamento de Sequência , Transdução de Sinais/imunologia , Tirosina/química
20.
Neurology ; 82(23): 2085-91, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24808022

RESUMO

OBJECTIVE: To determine whether phosphodiesterase type 5 (PDE5) inhibition can alleviate exercise-induced skeletal muscle ischemia in boys with Duchenne muscular dystrophy (DMD). METHODS: In 10 boys with DMD and 10 healthy age-matched male controls, we assessed exercise-induced attenuation of reflex sympathetic vasoconstriction, i.e., functional sympatholysis, a protective mechanism that matches oxygen delivery to metabolic demand. Reflex vasoconstriction was induced by simulated orthostatic stress, measured as the decrease in forearm muscle oxygenation with near-infrared spectroscopy, and performed when the forearm muscles were rested or lightly exercised with rhythmic handgrip exercise. Then, the patients underwent an open-label, dose-escalation, crossover trial with single oral doses of tadalafil or sildenafil. RESULTS: The major new findings are 2-fold: first, sympatholysis is impaired in boys with DMD-producing functional muscle ischemia-despite contemporary background therapy with corticosteroids alone or in combination with cardioprotective medication. Second, PDE5 inhibition with standard clinical doses of either tadalafil or sildenafil alleviates this ischemia in a dose-dependent manner. Furthermore, PDE5 inhibition also normalizes the exercise-induced increase in skeletal muscle blood flow (measured by Doppler ultrasound), which is markedly blunted in boys with DMD. CONCLUSIONS: These data provide in-human proof of concept for PDE5 inhibition as a putative new therapeutic strategy for DMD. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that in patients with DMD, PDE5 inhibition restores functional sympatholysis.


Assuntos
Carbolinas/farmacologia , Isquemia/tratamento farmacológico , Músculo Esquelético , Distrofia Muscular de Duchenne/tratamento farmacológico , Inibidores da Fosfodiesterase 5/farmacologia , Piperazinas/farmacologia , Sulfonas/farmacologia , Sistema Nervoso Simpático/fisiopatologia , Adolescente , Carbolinas/administração & dosagem , Carbolinas/efeitos adversos , Carbolinas/farmacocinética , Criança , Exercício Físico/fisiologia , Antebraço/irrigação sanguínea , Antebraço/diagnóstico por imagem , Humanos , Isquemia/etiologia , Masculino , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/fisiopatologia , Inibidores da Fosfodiesterase 5/administração & dosagem , Inibidores da Fosfodiesterase 5/efeitos adversos , Inibidores da Fosfodiesterase 5/farmacocinética , Piperazinas/administração & dosagem , Piperazinas/efeitos adversos , Piperazinas/farmacocinética , Purinas/administração & dosagem , Purinas/efeitos adversos , Purinas/farmacocinética , Purinas/farmacologia , Citrato de Sildenafila , Espectroscopia de Luz Próxima ao Infravermelho , Sulfonas/administração & dosagem , Sulfonas/efeitos adversos , Sulfonas/farmacocinética , Sistema Nervoso Simpático/efeitos dos fármacos , Tadalafila , Ultrassonografia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...