Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Brain ; 146(10): 4025-4032, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37337674

RESUMO

Copy number variation (CNV) may lead to pathological traits, and Charcot-Marie-Tooth disease type 1A (CMT1A), the commonest inherited peripheral neuropathy, is due to a genomic duplication encompassing the dosage-sensitive PMP22 gene. MicroRNAs act as repressors on post-transcriptional regulation of gene expression and in rodent models of CMT1A, overexpression of one such microRNA (miR-29a) has been shown to reduce the PMP22 transcript and protein level. Here we present genomic and functional evidence, for the first time in a human CNV-associated phenotype, of the 3' untranslated region (3'-UTR)-mediated role of microRNA repression on gene expression. The proband of the family presented with an early-onset, severe sensorimotor demyelinating neuropathy and harboured a novel de novo deletion in the PMP22 3'-UTR. The deletion is predicted to include the miR-29a seed binding site and transcript analysis of dermal myelinated nerve fibres using a novel platform, revealed a marked increase in PMP22 transcript levels. Functional evidence from Schwann cell lines harbouring the wild-type and mutant 3'-UTR showed significantly increased reporter assay activity in the latter, which was not ameliorated by overexpression of a miR-29a mimic. This shows the importance of miR-29a in regulating PMP22 expression and opens an avenue for therapeutic drug development.


Assuntos
Doença de Charcot-Marie-Tooth , MicroRNAs , Humanos , Doença de Charcot-Marie-Tooth/patologia , MicroRNAs/genética , Variações do Número de Cópias de DNA , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Expressão Gênica
2.
J Biol Chem ; 297(1): 100852, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34090875

RESUMO

The induction of nerve injury response genes in Schwann cells depends on both transcriptional and epigenomic reprogramming. The nerve injury response program is regulated by the repressive histone mark H3K27 trimethylation (H3K27me3), deposited by Polycomb repressive complex 2 (PRC2). Loss of PRC2 function leads to early and augmented induction of the injury response gene network in peripheral nerves, suggesting H3K27 demethylases are required for derepression of Polycomb-regulated nerve injury genes. To determine the function of H3K27 demethylases in nerve injury, we generated Schwann cell-specific knockouts of H3K27 demethylase Kdm6b and double knockouts of Kdm6b/Kdm6a (encoding JMJD3 and UTX). We found that H3K27 demethylases are largely dispensable for Schwann cell development and myelination. In testing the function of H3K27 demethylases after injury, we found early induction of some nerve injury genes was diminished compared with control, but most injury genes were largely unaffected at 1 and 7 days post injury. Although it was proposed that H3K27 demethylases are required to activate expression of the cyclin-dependent kinase inhibitor Cdkn2a in response to injury, Schwann cell-specific deletion of H3K27 demethylases affected neither the expression of this gene nor Schwann cell proliferation after nerve injury. To further characterize the regulation of nerve injury response genes, we found that injury genes are associated with repressive histone H2AK119 ubiquitination catalyzed by PRC1, which declines after injury. Overall, our results indicate H3K27 demethylation is not required for induction of injury response genes and that other mechanisms likely are involved in activating Polycomb-repressed injury genes in peripheral nerve.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Histona Desmetilases/genética , Histona Desmetilases com o Domínio Jumonji/genética , Traumatismos dos Nervos Periféricos/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Código das Histonas/genética , Histonas/genética , Humanos , Metilação , Camundongos , Traumatismos dos Nervos Periféricos/patologia , Complexo Repressor Polycomb 2/genética , Células de Schwann/metabolismo , Células de Schwann/patologia , Ubiquitinação/genética
3.
Hum Mol Genet ; 29(10): 1689-1699, 2020 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-32356557

RESUMO

Copy number variation of the peripheral nerve myelin gene Peripheral Myelin Protein 22 (PMP22) causes multiple forms of inherited peripheral neuropathy. The duplication of a 1.4 Mb segment surrounding this gene in chromosome 17p12 (c17p12) causes the most common form of Charcot-Marie-Tooth disease type 1A, whereas the reciprocal deletion of this gene causes a separate neuropathy termed hereditary neuropathy with liability to pressure palsies (HNPP). PMP22 is robustly induced in Schwann cells in early postnatal development, and several transcription factors and their cognate regulatory elements have been implicated in coordinating the gene's proper expression. We previously found that a distal super-enhancer domain was important for Pmp22 expression in vitro, with particular impact on a Schwann cell-specific alternative promoter. Here, we investigate the consequences of deleting this super-enhancer in vivo. We find that loss of the super-enhancer in mice reduces Pmp22 expression throughout development and into adulthood, with greater impact on the Schwann cell-specific promoter. Additionally, these mice display tomacula formed by excessive myelin folding, a pathological hallmark of HNPP, as have been previously observed in heterozygous Pmp22 mice as well as sural biopsies from patients with HNPP. Our findings demonstrate a mechanism by which smaller copy number variations, not including the Pmp22 gene, are sufficient to reduce gene expression and phenocopy a peripheral neuropathy caused by the HNPP-associated deletion encompassing PMP22.


Assuntos
Artrogripose/genética , Doença de Charcot-Marie-Tooth/genética , Elementos Facilitadores Genéticos/genética , Neuropatia Hereditária Motora e Sensorial/genética , Proteínas da Mielina/genética , Adulto , Animais , Artrogripose/metabolismo , Artrogripose/patologia , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Variações do Número de Cópias de DNA/genética , Neuropatia Hereditária Motora e Sensorial/metabolismo , Neuropatia Hereditária Motora e Sensorial/patologia , Heterozigoto , Humanos , Camundongos , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Fenótipo , Células de Schwann/metabolismo , Células de Schwann/patologia
4.
Hum Mol Genet ; 27(16): 2830-2839, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29771329

RESUMO

Peripheral nerve myelination is adversely affected in the most common form of the hereditary peripheral neuropathy called Charcot-Marie-Tooth Disease. This form, classified as CMT1A, is caused by a 1.4 Mb duplication on chromosome 17, which includes the abundantly expressed Schwann cell myelin gene, Peripheral Myelin Protein 22 (PMP22). This is one of the most common copy number variants causing neurological disease. Overexpression of Pmp22 in rodent models recapitulates several aspects of neuropathy, and reduction of Pmp22 in such models results in amelioration of the neuropathy phenotype. Recently we identified a potential super-enhancer approximately 90-130 kb upstream of the Pmp22 transcription start sites. This super-enhancer encompasses a cluster of individual enhancers that have the acetylated histone H3K27 active enhancer mark, and coincides with smaller duplications identified in patients with milder CMT1A-like symptoms, where the PMP22 coding region itself was not part of the duplication. In this study, we have utilized genome editing to create a deletion of this super-enhancer to determine its role in Pmp22 regulation. Our data show a significant decrease in Pmp22 transcript expression using allele-specific internal controls. Moreover, the P2 promoter of the Pmp22 gene, which is used in other cell types, is affected, but we find that the Schwann cell-specific P1 promoter is disproportionately more sensitive to loss of the super-enhancer. These data show for the first time the requirement of these upstream enhancers for full Pmp22 expression.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Variações do Número de Cópias de DNA/genética , Elementos Facilitadores Genéticos/genética , Proteínas da Mielina/genética , Animais , Linhagem Celular , Doença de Charcot-Marie-Tooth/patologia , Duplicação Cromossômica/genética , Cromossomos Humanos Par 17/genética , Edição de Genes , Regulação da Expressão Gênica/genética , Humanos , Regiões Promotoras Genéticas , Ratos , Células de Schwann/patologia
5.
Ann Neurol ; 85(6): 887-898, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30945774

RESUMO

OBJECTIVE: Charcot-Marie-Tooth (CMT) disease is most commonly caused by duplication of a chromosomal segment surrounding Peripheral Myelin Protein 22, or PMP22 gene, which is classified as CMT1A. Several candidate therapies reduce Pmp22 mRNA levels in CMT1A rodent models, but development of biomarkers for clinical trials in CMT1A is a challenge given its slow progression and difficulty in obtaining nerve samples. Quantitative PCR measurements of PMP22 mRNA in dermal nerves were performed using skin biopsies in human clinical trials for CMT1A, but this approach did not show increased PMP22 mRNA in CMT1A patients compared to controls. One complicating factor is the variable amounts of Schwann cells (SCs) in skin. The objective of the study was to develop a novel method for precise evaluation of PMP22 levels in skin biopsies that can discriminate CMT1A patients from controls. METHODS: We have developed methods to normalize PMP22 transcript levels to SC-specific genes that are not altered by CMT1A status. Several CMT1A-associated genes were assembled into a custom Nanostring panel to enable precise transcript measurements that can be normalized to variable SC content. RESULTS: The digital expression data from Nanostring analysis showed reproducible elevation of PMP22 levels in CMT1A versus control skin biopsies, particularly after normalization to SC-specific genes. INTERPRETATION: This platform should be useful in clinical trials for CMT1A as a biomarker of target engagement that can be used to optimize dosing, and the same normalization framework is applicable to other types of CMT. ANN NEUROL 2019;85:887-898.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Proteínas da Mielina/genética , Células de Schwann/patologia , Pele/patologia , Adolescente , Adulto , Idoso , Animais , Biomarcadores/metabolismo , Biópsia , Doença de Charcot-Marie-Tooth/metabolismo , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas da Mielina/biossíntese , Células de Schwann/metabolismo , Pele/metabolismo , Adulto Jovem
6.
Ann Neurol ; 85(3): 316-330, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30706531

RESUMO

OBJECTIVE: Genetic modifiers in rare disease have long been suspected to contribute to the considerable variance in disease expression, including Charcot-Marie-Tooth disease type 1A (CMT1A). To address this question, the Inherited Neuropathy Consortium collected a large standardized sample of such rare CMT1A patients over a period of 8 years. CMT1A is caused in most patients by a uniformly sized 1.5 Mb duplication event involving the gene PMP22. METHODS: We genotyped DNA samples from 971 CMT1A patients on Illumina BeadChips. Genome-wide analysis was performed in a subset of 330 of these patients, who expressed the extremes of a hallmark symptom: mild and severe foot dorsiflexion strength impairment. SIPA1L2 (signal-induced proliferation-associated 1 like 2), the top identified candidate modifier gene, was expressed in the peripheral nerve, and our functional studies identified and confirmed interacting proteins using coimmunoprecipitation analysis, mass spectrometry, and immunocytochemistry. Chromatin immunoprecipitation and in vitro siRNA experiments were used to analyze gene regulation. RESULTS: We identified significant association of 4 single nucleotide polymorphisms (rs10910527, rs7536385, rs4649265, rs1547740) in SIPA1L2 with foot dorsiflexion strength (p < 1 × 10-7 ). Coimmunoprecipitation and mass spectroscopy studies identified ß-actin and MYH9 as SIPA1L2 binding partners. Furthermore, we show that SIPA1L2 is part of a myelination-associated coexpressed network regulated by the master transcription factor SOX10. Importantly, in vitro knockdown of SIPA1L2 in Schwannoma cells led to a significant reduction of PMP22 expression, hinting at a potential strategy for drug development. INTERPRETATION: SIPA1L2 is a potential genetic modifier of CMT1A phenotypic expressions and offers a new pathway to therapeutic interventions. ANN NEUROL 2019;85:316-330.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Pé/fisiopatologia , Proteínas Ativadoras de GTPase/genética , Genes Modificadores/genética , Debilidade Muscular/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Doença de Charcot-Marie-Tooth/fisiopatologia , Criança , Pré-Escolar , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Debilidade Muscular/fisiopatologia , Proteínas da Mielina/genética , Neurilemoma/genética , Fenótipo , Polimorfismo de Nucleotídeo Único , Ratos , Índice de Gravidade de Doença , Adulto Jovem
7.
Glia ; 66(11): 2487-2502, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30306639

RESUMO

The transition of differentiated Schwann cells to support of nerve repair after injury is accompanied by remodeling of the Schwann cell epigenome. The EED-containing polycomb repressive complex 2 (PRC2) catalyzes histone H3K27 methylation and represses key nerve repair genes such as Shh, Gdnf, and Bdnf, and their activation is accompanied by loss of H3K27 methylation. Analysis of nerve injury in mice with a Schwann cell-specific loss of EED showed the reversal of polycomb repression is required and a rate limiting step in the increased transcription of Neuregulin 1 (type I), which is required for efficient remyelination. However, mouse nerves with EED-deficient Schwann cells display slow axonal regeneration with significantly low expression of axon guidance genes, including Sema4f and Cntf. Finally, EED loss causes impaired Schwann cell proliferation after injury with significant induction of the Cdkn2a cell cycle inhibitor gene. Interestingly, PRC2 subunits and CDKN2A are commonly co-mutated in the transition from benign neurofibromas to malignant peripheral nerve sheath tumors (MPNST's). RNA-seq analysis of EED-deficient mice identified PRC2-regulated molecular pathways that may contribute to the transition to malignancy in neurofibromatosis.


Assuntos
Proliferação de Células/fisiologia , Regulação da Expressão Gênica/genética , Regeneração Nervosa/efeitos dos fármacos , Complexo Repressor Polycomb 2/metabolismo , Células de Schwann/fisiologia , Neuropatia Ciática/fisiopatologia , Animais , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Regeneração Nervosa/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurregulinas/metabolismo , Proteína Oncogênica v-akt/metabolismo , Complexo Repressor Polycomb 2/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Células de Schwann/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
8.
Hum Mol Genet ; 25(14): 3055-3069, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27288457

RESUMO

Schwann cells are myelinating glia in the peripheral nervous system that form the myelin sheath. A major cause of peripheral neuropathy is a copy number variant involving the Peripheral Myelin Protein 22 (PMP22) gene, which is located within a 1.4-Mb duplication on chromosome 17 associated with the most common form of Charcot-Marie-Tooth Disease (CMT1A). Rodent models of CMT1A have been used to show that reducing Pmp22 overexpression mitigates several aspects of a CMT1A-related phenotype. Mechanistic studies of Pmp22 regulation identified enhancers regulated by the Sox10 (SRY sex determining region Y-box 10) and Egr2/Krox20 (Early growth response protein 2) transcription factors in myelinated nerves. However, relatively little is known regarding how other transcription factors induce Pmp22 expression during Schwann cell development and myelination. Here, we examined Pmp22 enhancers as a function of cell type-specificity, nerve injury and development. While Pmp22 enhancers marked by active histone modifications were lost or remodeled after injury, we found that these enhancers were permissive in early development prior to Pmp22 upregulation. Pmp22 enhancers contain binding motifs for TEA domain (Tead) transcription factors of the Hippo signaling pathway. We discovered that Tead1 and co-activators Yap and Taz are required for Pmp22 expression, as well as for the expression of Egr2 Tead1 directly binds Pmp22 and Egr2 enhancers early in development and Tead1 binding is induced during myelination, correlating with Pmp22 expression. The data identify Tead1 as a novel regulator of Pmp22 expression during development in concert with Sox10 and Egr2.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas de Ligação a DNA/genética , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteínas da Mielina/genética , Doenças do Sistema Nervoso Periférico/genética , Fatores de Transcrição SOXE/genética , Fatores de Transcrição/genética , Animais , Doença de Charcot-Marie-Tooth/patologia , Variações do Número de Cópias de DNA/genética , Proteínas de Ligação a DNA/biossíntese , Modelos Animais de Doenças , Proteína 2 de Resposta de Crescimento Precoce/biossíntese , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Bainha de Mielina/genética , Bainha de Mielina/patologia , Neurogênese/genética , Doenças do Sistema Nervoso Periférico/patologia , Fenótipo , Células de Schwann/metabolismo , Células de Schwann/patologia , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/biossíntese
9.
Ann Neurol ; 81(5): 690-702, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28393430

RESUMO

OBJECTIVE: Our goal was to define the genetic cause of the profound hypomyelination in the taiep rat model and determine its relevance to human white matter disease. METHODS: Based on previous localization of the taiep mutation to rat chromosome 9, we tested whether the mutation resided within the Tubb4a (ß-tubulin 4A) gene, because mutations in the TUBB4A gene have been described in patients with central nervous system hypomyelination. To determine whether accumulation of microtubules led to progressive demyelination, we analyzed the spinal cord and optic nerves of 2-year-old rats by light and electron microscopy. Cerebral white matter from a patient with TUBB4A Asn414Lys mutation and magnetic resonance imaging evidence of severe hypomyelination were studied similarly. RESULTS: As the taiep rat ages, there is progressive loss of myelin in the brain and dorsal column of the spinal cord associated with increased oligodendrocyte numbers with accumulation of microtubules. This accumulation involved the entire cell body and distal processes of oligodendrocytes, but there was no accumulation of microtubules in axons. A single point mutation in Tubb4a (p.Ala302Thr) was found in homozygous taiep samples. A similar hypomyelination associated with increased oligodendrocyte numbers and arrays of microtubules in oligodendrocytes was demonstrated in the human patient sample. INTERPRETATION: The taiep rat is the first animal model of TUBB4 mutations in humans and a novel system in which to test the mechanism of microtubule accumulation. The finding of microtubule accumulation in a patient with a TUBB4A mutation and leukodystrophy confirms the usefulness of taiep as a model of the human disease. Ann Neurol 2017;81:690-702.


Assuntos
Doenças Desmielinizantes , Modelos Animais de Doenças , Microtúbulos/metabolismo , Nervo Óptico/diagnóstico por imagem , Medula Espinal/diagnóstico por imagem , Tubulina (Proteína)/genética , Substância Branca/diagnóstico por imagem , Animais , Pré-Escolar , Doenças Desmielinizantes/diagnóstico por imagem , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Microscopia Eletrônica , Ratos
10.
Clin Transplant ; 30(5): 606-12, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26928266

RESUMO

BACKGROUND: Although pre-transplant immunization is routinely recommended, this recommendation is based on little data. The primary objective of this study was to compare antibody responses in lung transplant patients who received influenza vaccine before the transplant, within the first six months of transplant, between 13 and 60 months post-transplant, and 110 months or beyond transplant. METHODS: This prospective cohort study included 357 total immunization events performed over five yr to measure H1N1, H3N2, and B antibody responses to the influenza vaccine in pre- and post-lung transplant patients. Geometric mean titers, seroprotection (antibody titer at least 1:40), seroconversion (fourfold increase between pre and post), and mean fold increases were compared. RESULTS: The geometric mean titer distributions were different for H3N2 and B (ANOVA; p = 0.002 for both). Pre-transplant antibody concentrations were higher compared to the 13- to 60-month group for H3N2 (corrected p = 0.002) and the healthy group for B (corrected p = 0.001). The ≥110-month group had higher seroconversion rates compared to the 13- to 60-month group for H3N2 and B viruses. CONCLUSION: Lung pre-transplant patients and the long-term survivors have higher responses to the influenza vaccine than early post-transplant and the transplant control groups.


Assuntos
Anticorpos Antivirais/sangue , Formação de Anticorpos/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Influenza Humana/prevenção & controle , Pneumopatias/imunologia , Transplante de Pulmão , Anticorpos Antivirais/imunologia , Estudos de Casos e Controles , Feminino , Seguimentos , Sobrevivência de Enxerto , Humanos , Vacinas contra Influenza/administração & dosagem , Influenza Humana/imunologia , Influenza Humana/virologia , Pneumopatias/cirurgia , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Fatores de Risco , Vacinação
11.
Nucleic Acids Res ; 40(14): 6449-60, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22492709

RESUMO

Myelin is essential for the rapidity of saltatory nerve conduction, and also provides trophic support for axons to prevent axonal degeneration. Two critical determinants of myelination are SOX10 and EGR2/KROX20. SOX10 is required for specification of Schwann cells from neural crest, and is required at every stage of Schwann cell development. Egr2/Krox20 expression is activated by axonal signals in myelinating Schwann cells, and is required for cell cycle arrest and myelin formation. To elucidate the integrated function of these two transcription factors during peripheral nerve myelination, we performed in vivo ChIP-Seq analysis of myelinating peripheral nerve. Integration of these binding data with loss-of-function array data identified a range of genes regulated by these factors. In addition, although SOX10 itself regulates Egr2/Krox20 expression, leading to coordinate activation of several major myelin genes by the two factors, there is a large subset of genes that are activated independent of EGR2. Finally, the results identify a set of SOX10-dependent genes that are expressed in early Schwann cell development, but become subsequently repressed by EGR2/KROX20.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Bainha de Mielina/fisiologia , Fatores de Transcrição SOXE/metabolismo , Nervo Isquiático/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Regulação da Expressão Gênica , Genoma , Camundongos , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/fisiologia
12.
Antimicrob Agents Chemother ; 56(8): 4422-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22687502

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) isolates that are susceptible to vancomycin but are tolerant to its killing effect may present a potential challenge for effective treatment. This study compared the microbiologic characteristics of clinical vancomycin-tolerant (VT-MRSA) and vancomycin-susceptible (VS-MRSA) strains using phenotypic and gene regulation studies. MRSA isolates collected from vancomycin-treated patients with bacteremia over a 5-year period were analyzed for vancomycin, daptomycin, and telavancin susceptibility, as well as accessory gene regulator (agr) group and function. Vancomycin tolerance was defined by a minimum bactericidal concentration (MBC)/minimum inhibitor concentration (MIC) ratio of ≥32 mg/liter. VT-MRSA isolates were compared to VS-MRSA isolates for differences in antimicrobial susceptibility, time-kill activity, and gene expression of key cell envelope response genes vraSR, dltA, and mprF. All 115 isolates evaluated were susceptible to vancomycin, daptomycin, and telavancin. Seven isolates (6%) were VT-MRSA. agr group II was more prevalent in isolates with vancomycin MBC/MIC ratios of ≥8. In time-kill analyses, VT-MRSA had reduced vancomycin killing, but daptomycin and telavancin activities were maintained. Significantly greater gene expression was observed in VT-MRSA after 72 h of subinhibitory antibiotic exposures. Vancomycin most notably increased vraSR expression (P = 0.002 versus VS-MRSA strains). Daptomycin and telavancin increased expression of all genes studied, most significantly mprF expression (P < 0.001). Longer durations of antibiotic exposure (72 h versus 24 h) resulted in substantial increases in gene expression in VT-MRSA. Although the clinical impact of VT-MRSA is not fully recognized, these data suggest that VT-MRSA strains, while still susceptible, have altered gene regulation to adapt to the antimicrobial effects of glyco- and lipopeptides that may emerge during prolonged durations of exposure.


Assuntos
Aminoglicosídeos/farmacologia , Daptomicina/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Resistência a Vancomicina/genética , Vancomicina/farmacologia , Aminoaciltransferases/genética , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Tolerância a Medicamentos , Regulação Bacteriana da Expressão Gênica , Humanos , Lipoglicopeptídeos , Staphylococcus aureus Resistente à Meticilina/genética , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Testes de Sensibilidade Microbiana
13.
J Antimicrob Chemother ; 67(1): 123-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21980070

RESUMO

BACKGROUND: Toxins contribute to the pathogenicity of Staphylococcus aureus infections by inducing a dysregulated inflammatory response. This study evaluated the impact of anti-staphylococcal antibiotic exposures over an increasing concentration range on cytokine production from peripheral blood mononuclear cells (PBMCs) after S. aureus toxin exposures. METHODS: Human PBMCs were suspended in complete Roswell Park Memorial Institute (RPMI) 1640 medium with 10% fetal bovine serum at 10(6) cells/mL with 100 ng/mL S. aureus toxic shock syndrome toxin-1 (TSST-1), staphylococcal enterotoxin A (SEA), α-toxin or Panton-Valentine leucocidin (PVL). Vancomycin, trimethoprim/sulfamethoxazole, tigecycline, daptomycin, linezolid, clindamycin and azithromycin were added at a concentration range of 0.5-100 mg/L. Cytokine [interleukin-1ß (IL-1ß), IL-6, IL-8, interferon-γ (IFN-γ) and tumour necrosis factor-α (TNF-α)] concentrations were measured in duplicate by ELISA following exposure and were compared with response with toxin alone. RESULTS: At concentrations approximating serum C(max), tigecycline decreased IL-6 by 52%-57% and IFN-γ production by 43%-53% compared with toxin alone (P ≤ 0.05) and linezolid inhibited TNF-α by 12%-35% and IL-8 by 25%-42% (P ≤ 0.02). However, trimethoprim/sulfamethoxazole increased TNF-α and IL-8 production (P = 0.002). Clindamycin, daptomycin, vancomycin and azithromycin had no consistent significant effect at approximate serum C(max) concentrations. All antibiotics had a concentration-dependent effect on cytokine production, with tigecycline, clindamycin and trimethoprim/sulfamethoxazole being the most potent inhibitors of cytokine production at concentrations exceeding 25 mg/L. CONCLUSIONS: S. aureus toxins stimulate production of inflammatory cytokines in PBMCs. Antimicrobials with high tissue penetration, including tigecycline, clindamycin, trimethoprim/sulfamethoxazole and linezolid, reduced cytokine production, which, along with their antimicrobial effects, may have importance in the therapeutic outcome of severe infections.


Assuntos
Anti-Infecciosos/farmacologia , Toxinas Bacterianas/imunologia , Citocinas/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Sangue/imunologia , Células Cultivadas , Humanos , Staphylococcus aureus/imunologia
14.
iScience ; 25(4): 104132, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35434551

RESUMO

Cells elaborate transcriptional programs in response to external signals. In the peripheral nerves, Schwann cells (SC) sort axons of given caliber and start the process of wrapping their membrane around them. We identify Actin-like protein 6a (ACTL6a), part of SWI/SNF chromatin remodeling complex, as critical for the integration of axonal caliber recognition with the transcriptional program of myelination. Nuclear levels of ACTL6A in SC are increased by contact with large caliber axons or nanofibers, and result in the eviction of repressive histone marks to facilitate myelination. Without Actl6a the SC are unable to coordinate caliber recognition and myelin production. Peripheral nerves in knockout mice display defective radial sorting, hypo-myelination of large caliber axons, and redundant myelin around small caliber axons, resulting in a clinical motor phenotype. Overall, this suggests that ACTL6A is a key component of the machinery integrating external signals for proper myelination of the peripheral nerve.

15.
J Virol ; 84(18): 9390-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20631142

RESUMO

The 9-aminoacridine (9AA) derivative quinacrine (QC) has a long history of safe human use as an antiprotozoal and antirheumatic agent. QC intercalates into DNA and RNA and can inhibit DNA replication, RNA transcription, and protein synthesis. The extent of QC intercalation into RNA depends on the complexity of its secondary and tertiary structure. Internal ribosome entry sites (IRESs) that are required for initiation of translation of some viral and cellular mRNAs typically have complex structures. Recent work has shown that some intercalating drugs, including QC, are capable of inhibiting hepatitis C virus IRES-mediated translation in a cell-free system. Here, we show that QC suppresses translation directed by the encephalomyocarditis virus (EMCV) and poliovirus IRESs in a cell-free system and in virus-infected HeLa cells. In contrast, IRESs present in the mammalian p53 transcript that are predicted to have less-complex structures were not sensitive to QC. Inhibition of IRES-mediated translation by QC correlated with the affinity of binding between QC and the particular IRES. Expression of viral capsid proteins, replication of viral RNAs, and production of virus were all strongly inhibited by QC (and 9AA). These results suggest that QC and similar intercalating drugs could potentially be used for treatment of viral infections.


Assuntos
Antivirais/farmacologia , Vírus da Encefalomiocardite/efeitos dos fármacos , Poliovirus/efeitos dos fármacos , Quinacrina/farmacologia , Replicação Viral/efeitos dos fármacos , Sítios de Ligação , Vírus da Encefalomiocardite/fisiologia , Células HeLa , Humanos , Conformação de Ácido Nucleico , Poliovirus/fisiologia , Biossíntese de Proteínas/efeitos dos fármacos , RNA Viral/metabolismo , Proteínas Virais/biossíntese
16.
Respiration ; 82(5): 458-67, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21846958

RESUMO

BACKGROUND: Xanthine oxidase is a major source of superoxide in the vascular endothelium. Previous work in humans demonstrated improved conduit artery function following xanthine oxidase inhibition in patients with obstructive sleep apnea. OBJECTIVES: To determine whether impairments in endothelium-dependent vasodilation produced by exposure to chronic intermittent hypoxia are prevented by in vivo treatment with allopurinol, a xanthine oxidase inhibitor. METHODS: Sprague-Dawley rats received allopurinol (65 mg/kg/day) or vehicle via oral gavage. Half of each group was exposed to intermittent hypoxia (FIO(2) = 0.10 for 1 min, 15×/h, 12 h/day) and the other half to normoxia. After 14 days, gracilis arteries were isolated, cannulated with micropipettes, and perfused and superfused with physiological salt solution. Diameters were measured before and after exposure to acetylcholine (10(-6)M) and nitroprusside (10(-4)M). RESULTS: In vehicle-treated rats, intermittent hypoxia impaired acetylcholine-induced vasodilation compared to normoxia (+4 ± 4 vs. +21 ± 6 µm, p = 0.01). Allopurinol attenuated this impairment (+26 ± 6 vs. +34 ± 9 µm for intermittent hypoxia and normoxia groups treated with allopurinol, p = 0.55). In contrast, nitroprusside-induced vasodilation was similar in all rats (p = 0.43). Neither allopurinol nor intermittent hypoxia affected vessel morphometry or systemic markers of oxidative stress. Urinary uric acid concentrations were reduced in allopurinol- versus vehicle-treated rats (p = 0.02). CONCLUSIONS: These data confirm previous findings that exposure to intermittent hypoxia impairs endothelium-dependent vasodilation in skeletal muscle resistance arteries and extend them by demonstrating that this impairment can be prevented with allopurinol. Thus, xanthine oxidase appears to play a key role in mediating intermittent hypoxia-induced vascular dysfunction.


Assuntos
Alopurinol/farmacologia , Endotélio Vascular/efeitos dos fármacos , Hipóxia/prevenção & controle , Resistência Vascular/efeitos dos fármacos , Xantina Oxidase/antagonistas & inibidores , Acetilcolina/farmacologia , Animais , Endotélio Vascular/fisiopatologia , Sequestradores de Radicais Livres/farmacologia , Hipóxia/complicações , Masculino , Nitroprussiato/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
17.
ACS Pharmacol Transl Sci ; 4(4): 1422-1436, 2021 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-34423274

RESUMO

Charcot-Marie-Tooth 1A (CMT1A) is the most common form of hereditary peripheral neuropathies, characterized by genetic duplication of the critical myelin gene Peripheral Myelin Protein 22 (PMP22). PMP22 overexpression results in abnormal Schwann cell differentiation, leading to axonal loss and muscle wasting. Since regulation of PMP22 expression is a major target of therapeutic discovery for CMT1A, we sought to establish unbiased approaches that allow the identification of therapeutic agents for this disease. Using genome editing, we generated a coincidence reporter assay that accurately monitors Pmp22 transcript levels in the S16 rat Schwann cell line, while reducing reporter-based false positives. A quantitative high-throughput screen (qHTS) of 42 577 compounds using this assay revealed diverse novel chemical classes that reduce endogenous Pmp22 transcript levels. Moreover, some of these classes show pharmacological specificity in reducing Pmp22 over another major myelin-associated gene, Mpz (Myelin protein zero). Finally, to investigate whether compound-mediated reduction of Pmp22 transcripts translates to reduced PMP22 protein levels, we edited the S16 genome to generate a reporter assay that expresses a PMP22-HiBiT fusion protein using CRISPR/Cas9. Overall, we present a screening platform that combines genome edited cell lines encoding reporters that monitor transcriptional and post-translational regulation of PMP22 with titration-based screening (e.g., qHTS), which could be efficiently incorporated into drug discovery campaigns for CMT1A.

18.
PLoS One ; 15(12): e0244075, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33382735

RESUMO

Anterior cruciate ligament (ACL) rupture is an important condition of the human knee. Second ruptures are common and societal costs are substantial. Canine cranial cruciate ligament (CCL) rupture closely models the human disease. CCL rupture is common in the Labrador Retriever (5.79% prevalence), ~100-fold more prevalent than in humans. Labrador Retriever CCL rupture is a polygenic complex disease, based on genome-wide association study (GWAS) of single nucleotide polymorphism (SNP) markers. Dissection of genetic variation in complex traits can be enhanced by studying structural variation, including copy number variants (CNVs). Dogs are an ideal model for CNV research because of reduced genetic variability within breeds and extensive phenotypic diversity across breeds. We studied the genetic etiology of CCL rupture by association analysis of CNV regions (CNVRs) using 110 case and 164 control Labrador Retrievers. CNVs were called from SNPs using three different programs (PennCNV, CNVPartition, and QuantiSNP). After quality control, CNV calls were combined to create CNVRs using ParseCNV and an association analysis was performed. We found no strong effect CNVRs but found 46 small effect (max(T) permutation P<0.05) CCL rupture associated CNVRs in 22 autosomes; 25 were deletions and 21 were duplications. Of the 46 CCL rupture associated CNVRs, we identified 39 unique regions. Thirty four were identified by a single calling algorithm, 3 were identified by two calling algorithms, and 2 were identified by all three algorithms. For 42 of the associated CNVRs, frequency in the population was <10% while 4 occurred at a frequency in the population ranging from 10-25%. Average CNVR length was 198,872bp and CNVRs covered 0.11 to 0.15% of the genome. All CNVRs were associated with case status. CNVRs did not overlap previous canine CCL rupture risk loci identified by GWAS. Associated CNVRs contained 152 annotated genes; 12 CNVRs did not have genes mapped to CanFam3.1. Using pathway analysis, a cluster of 19 homeobox domain transcript regulator genes was associated with CCL rupture (P = 6.6E-13). This gene cluster influences cranial-caudal body pattern formation during embryonic limb development. Clustered genes were found in 3 CNVRs on chromosome 14 (HoxA), 28 (NKX6-2), and 36 (HoxD). When analysis was limited to deletion CNVRs, the association was strengthened (P = 8.7E-16). This study suggests a component of the polygenic risk of CCL rupture in Labrador Retrievers is associated with small effect CNVs and may include aspects of stifle morphology regulated by homeobox domain transcript regulator genes.


Assuntos
Lesões do Ligamento Cruzado Anterior/genética , Ligamento Cruzado Anterior , Variações do Número de Cópias de DNA , Polimorfismo de Nucleotídeo Único , Animais , Cães , Estudo de Associação Genômica Ampla
19.
Ann Clin Transl Neurol ; 7(1): 69-82, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31833243

RESUMO

OBJECTIVE: Development of biomarkers for Charcot-Marie-Tooth (CMT) disease is critical for implementing effective clinical trials. The most common form of CMT, type 1A, is caused by a genomic duplication surrounding the PMP22 gene. A recent report (Neurology 2018;90:e518-3524) showed elevation of neurofilament light (NfL) in plasma of CMT1A disease patients, which correlated with disease severity. However, no plasma/serum biomarker has been identified that is specific to Schwann cells, the most directly affected cells in CMT1A. METHODS: We used the Olink immuno PCR platform to profile CMT1A patient (n = 47, 2 cohorts) and normal control plasma (n = 41, two cohorts) on five different Olink panels to screen 398 unique proteins. RESULTS: The TMPRSS5 protein (Transmembrane protease serine 5) was elevated 2.07-fold (P = <0.0001) in two independent cohorts of CMT1A samples relative to controls. TMPRSS5 is most highly expressed in Schwann cells of peripheral nerve. Consistent with early myelination deficits in CMT1A, TMPRSS5 was not significantly correlated with disease score (CMTES-R, CMTNS-R), nerve conduction velocities (Ulnar CMAP, Ulnar MNCV), or with age. TMPRSS5 was not significantly elevated in smaller sample sets from patients with CMT2A, CMT2E, CMT1B, or CMT1X. The Olink immuno PCR assays confirmed elevated levels of NfL (average 1.58-fold, P < 0.0001), which correlated with CMT1A patient disease score. INTERPRETATION: These data identify the first Schwann cell-specific protein that is elevated in plasma of CMT1A patients, and may provide a disease marker and a potentially treatment-responsive biomarker with good disease specificity for clinical trials.


Assuntos
Doença de Charcot-Marie-Tooth/sangue , Doença de Charcot-Marie-Tooth/diagnóstico , Proteínas de Membrana/sangue , Proteínas Mitocondriais/sangue , Células de Schwann , Serina Endopeptidases/sangue , Adulto , Animais , Biomarcadores/sangue , Células Cultivadas , Doença de Charcot-Marie-Tooth/fisiopatologia , Estudos de Coortes , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Condução Nervosa/fisiologia , Reação em Cadeia da Polimerase , Ratos
20.
Prog Transplant ; 19(2): 153-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19588665

RESUMO

BACKGROUND: Influenza viral infections cause significant morbidity and mortality each season. Lung transplant patients may be at higher risk because of their underlying pathophysiology. Although annual immunization is the standard of care, its efficacy remains largely unproven. Previous studies showed poor antibody response to influenza vaccine in lung transplant patients, but no data on the antibody response in consecutive seasons have been published. METHODS: We studied antibody responses to influenza vaccine in 122 subjects: 66 lung transplant recipients, 28 control subjects, and 28 patients awaiting lung transplantation. We compared antibody response rates to individual viruses contained in influenza vaccines in consecutive years within the 3 groups. Serum antibody concentrations were measured at baseline and 2 to 4 weeks after vaccination by using the hemagglutination inhibition assay. Log-transformed antibody concentrations and incidence of serconversion and seroprotection were calculated. RESULTS: Median log-transformed antibody responses were similar in consecutive seasons in lung transplant subjects. Incidences of seroprotection and seroconversion did not differ between consecutive seasons in lung transplant recipients. CONCLUSIONS: Antibody responses were similar in consecutively measured years in lung transplant subjects. Annual influenza vaccination in lung transplant subjects produces similar immune responses in 2 consecutive years, indicating that these patients are not at significantly increased risk of vaccine failure.


Assuntos
Anticorpos Antivirais , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza B/imunologia , Vacinas contra Influenza/imunologia , Transplante de Pulmão/imunologia , Adulto , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Estudos de Casos e Controles , Criança , Feminino , Humanos , Terapia de Imunossupressão/efeitos adversos , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Transplante de Pulmão/efeitos adversos , Masculino , Pessoa de Meia-Idade , Estatísticas não Paramétricas , Fatores de Tempo , Imunologia de Transplantes , Resultado do Tratamento , Vacinação , Wisconsin
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa