Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol In Vitro ; 79: 105280, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34843883

RESUMO

The intestine fulfills roles in the uptake of nutrients and water regulation and acts as a gatekeeper for the intestinal microbiome. For the latter, the intestinal gut barrier system is able to respond to a broad range of bacterial antigens, generally through Toll-like receptor (TLR) signaling pathways. To test the capacity of various in vitro intestinal models, we studied IL-8 secretion, as a marker of pro-inflammatory response through the TLR pathway, in a Caco-2 monoculture, Caco-2/HT29-MTX di-culture, Caco-2/HT29-MTX/HMVEC-d tri-culture and in a HT29-p monoculture in response to exposure to various TLR agonists. Twenty-one-day-old differentiated cells in Transwells were exposed to Pam3CSK4 (TLR1/2), lipopolysaccharide (TLR4), single-stranded RNA (TLR7/8), Poly(i:C) (TLR3) and flagellin (TLR5) for 24 h. In all systems IL-8 secretion was increased in response to flagellin exposure, with HT29-p cells also responding to Poly(I:C) exposure. All other agonists did not induce an IL-8 response in the tested in vitro models, indicating that the specific TLRs are either not present or not functional in these models. This highlights the need for careful selection of in vitro models when studying intestinal immune responses and the need for improved in vitro models that better recapitulate intestinal immune responses.


Assuntos
Interleucina-8/metabolismo , Mucosa Intestinal/imunologia , Receptores Toll-Like/agonistas , Células CACO-2 , Linhagem Celular , Técnicas de Cocultura , Flagelina/toxicidade , Células HT29 , Humanos , Imunidade Inata , Mucosa Intestinal/metabolismo , Lipopeptídeos/toxicidade , Lipopolissacarídeos/toxicidade , Poli I-C/toxicidade , RNA/toxicidade
2.
Hum Mol Genet ; 30(12): 1111-1130, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-33864373

RESUMO

RNA toxicity underlies the pathogenesis of disorders such as myotonic dystrophy type 1 (DM1). Muscular dystrophy is a key element of the pathology of DM1. The means by which RNA toxicity causes muscular dystrophy in DM1 is unclear. Here, we have used the DM200 mouse model of RNA toxicity due to the expression of a mutant DMPK 3'UTR mRNA to model the effects of RNA toxicity on muscle regeneration. Using a BaCl2-induced damage model, we find that RNA toxicity leads to decreased expression of PAX7, and decreased numbers of satellite cells, the stem cells of adult skeletal muscle (also known as MuSCs). This is associated with a delay in regenerative response, a lack of muscle fiber maturation and an inability to maintain a normal number of satellite cells. Repeated muscle damage also elicited key aspects of muscular dystrophy, including fat droplet deposition and increased fibrosis, and the results represent one of the first times to model these classic markers of dystrophic changes in the skeletal muscles of a mouse model of RNA toxicity. Using a ligand-conjugated antisense (LICA) oligonucleotide ASO targeting DMPK sequences for the first time in a mouse model of RNA toxicity in DM1, we find that treatment with IONIS 877864, which targets the DMPK 3'UTR mRNA, is efficacious in correcting the defects in regenerative response and the reductions in satellite cell numbers caused by RNA toxicity. These results demonstrate the possibilities for therapeutic interventions to mitigate the muscular dystrophy associated with RNA toxicity in DM1.


Assuntos
Desenvolvimento Muscular/genética , Distrofia Miotônica/genética , Miotonina Proteína Quinase/genética , Oligonucleotídeos Antissenso/farmacologia , RNA/genética , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Miotônica/patologia , Miotonina Proteína Quinase/antagonistas & inibidores , RNA/toxicidade , RNA Mensageiro/genética , Regeneração/genética
3.
Methods Mol Biol ; 2176: 99-109, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32865785

RESUMO

Myotonic dystrophy (DM) types 1 (DM1) and 2 (DM2) are caused by autosomal dominant gain-of-function RNA which are, in turn, created by the expansion of repeat sequences in the DMPK and ZNF9 genes, respectively. The expansions are highly unstable and biased for further expansion in somatic cells and across generations. Despite the different genes involved, DM1 and DM2 share several clinical features due to having the similar underlying mechanism of repetitive RNA-mediated toxicity. Both disorders manifest as multisystemic conditions with features including myotonia, cataract development, and abnormalities in cardiac conduction. At present, there is no cure for DM and treatments mostly aim at symptom management. Among the therapeutics being developed, antisense therapy using gapmers is one of the most promising. Compared to other antisense oligonucleotides, gapmers maintain the ability to induce RNase H cleavage while having enhanced target binding affinity and nuclease resistance. This chapter will consolidate the different strategies studied thus far to develop a treatment for DM1 through the targeting of toxic repetitive RNA using gapmers.


Assuntos
Distrofia Miotônica/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Estabilidade de RNA/efeitos dos fármacos , RNA/metabolismo , Animais , Humanos , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , RNA/genética , RNA/toxicidade , Ribonuclease H/metabolismo
4.
Neurobiol Dis ; 145: 105055, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32829028

RESUMO

A GGGGCC hexanucleotide repeat expansion in the first intron of C9orf72 is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Compelling evidence suggests that gain of toxicity from the bidirectionally transcribed repeat expanded RNAs plays a central role in disease pathogenesis. Two potential mechanisms have been proposed including RNA-mediated toxicity and/or the production of toxic dipeptide repeat proteins. In this review, we focus on the role of RNA mediated toxicity in ALS/FTD caused by the C9orf72 mutation and discuss arguments for and against this mechanism. In addition, we summarize how G4C2 repeat RNAs can elicit toxicity and potential therapeutic strategies to mitigate RNA-mediated toxicity.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Proteína C9orf72/genética , Demência Frontotemporal/patologia , RNA/toxicidade , Esclerose Lateral Amiotrófica/genética , Animais , Expansão das Repetições de DNA , Humanos
5.
Cell Death Dis ; 11(1): 3, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31919387

RESUMO

Polyglutamine diseases are neurodegenerative diseases that occur due to the expansion of CAG repeat regions in coding sequences of genes. Previously, we have shown the formation of large protein aggregates along with activation of the interferon pathway leading to apoptosis in a cellular model of SCA17. Here, we corroborate our previous results in a tetracycline-inducible model of SCA17. Interferon gamma and lambda were upregulated in 59Q-TBP expressing cells as compared to 16Q-TBP expressing cells. Besides interferon-stimulated genes, the SCA17 model and Huntington's mice brain samples showed upregulation of RNA sensors. However, in this improved model interferon pathway activation and apoptosis preceded the formation of large polyglutamine aggregates, suggesting a role for CAG repeat RNA or soluble protein aggregates. A polyglutamine minus mutant of TBP, expressing polyCAG mRNA, was created by site directed mutagenesis of 10 potential start codons. Neither this long CAG embedded mRNA nor short polyCAG RNA could induce interferon pathway genes or cause apoptosis. polyQ-TBP induced the expression of canonical RNA sensors but the downstream transcription factor, IRF3, showed a muted response. We found that expanded CAG repeat RNA is not sufficient to account for the neuronal apoptosis. Neuronal cells sense expanded CAG repeats embedded in messenger RNAs of protein-coding genes. However, polyglutamine containing protein is responsible for the interferon-mediated neuroinflammation and cell death seen in polyglutamine disease. Thus, we delineate the inflammatory role of CAG repeats in the mRNA from the resulting polyglutamine tract in the protein. Embedded in messenger RNAs of protein-coding regions, the cell senses CAG repeat expansion and induces the expression of RNA sensors and interferon-stimulated genes.


Assuntos
Encéfalo/patologia , Inflamação/patologia , Interferons/efeitos adversos , Peptídeos/toxicidade , RNA/toxicidade , Animais , Humanos , Camundongos , Modelos Biológicos , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Proteína de Ligação a TATA-Box/metabolismo , Tetraciclina/farmacologia , Expansão das Repetições de Trinucleotídeos/genética , Regulação para Cima/efeitos dos fármacos
6.
EMBO J ; 39(1): e101112, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31721251

RESUMO

Several neurodegenerative disorders like amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia (SCA) are caused by non-coding nucleotide repeat expansions. Different pathogenic mechanisms may underlie these non-coding repeat expansion disorders. While gain-of-function mechanisms, such as toxicity associated with expression of repeat RNA or toxicity associated with repeat-associated non-ATG (RAN) products, are most frequently connected with these disorders, loss-of-function mechanisms have also been implicated. We review the different pathways that have been linked to non-coding repeat expansion disorders such as C9ORF72-linked ALS/frontotemporal dementia (FTD), myotonic dystrophy, fragile X tremor/ataxia syndrome (FXTAS), SCA, and Huntington's disease-like 2. We discuss modes of RNA toxicity focusing on the identity and the interacting partners of the toxic RNA species. Using the C9ORF72 ALS/FTD paradigm, we further explore the efforts and different methods used to disentangle RNA vs. RAN toxicity. Overall, we conclude that there is ample evidence for a role of RNA toxicity in non-coding repeat expansion diseases.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Ataxia/patologia , Proteína C9orf72/genética , Expansão das Repetições de DNA/genética , Síndrome do Cromossomo X Frágil/patologia , Demência Frontotemporal/patologia , Distrofia Miotônica/patologia , Doenças Neurodegenerativas/patologia , RNA/toxicidade , Tremor/patologia , Esclerose Lateral Amiotrófica/genética , Ataxia/genética , Síndrome do Cromossomo X Frágil/genética , Demência Frontotemporal/genética , Humanos , Mutação , Distrofia Miotônica/genética , Doenças Neurodegenerativas/genética , RNA/genética , Tremor/genética
7.
J Am Chem Soc ; 140(49): 17226-17233, 2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30452248

RESUMO

In this Article, we show that the surface of the bacteriophage Qß is equipped with natural ligands for the synthesis of small gold nanoparticles (AuNPs). By exploiting disulfides in the protein secondary structure and the geometry formed from the capsid quaternary structure, we find that we can produce regularly arrayed patterns of ∼6 nm AuNPs across the surface of the virus-like particle. Experimental and computational analyses provide insight into the formation and stability of this composite. We further show that the entrapped genetic material can hold upward of 500 molecules of the anticancer drug Doxorubicin without leaking and without interfering with the synthesis of the AuNPs. This direct nucleation of nanoparticles on the capsid allows for exceptional conduction of photothermal energy upon nanosecond laser irradiation. As a proof of principle, we demonstrate that this energy is capable of rapidly releasing the drug from the capsid without heating the bulk solution, allowing for highly targeted cell killing in vitro.


Assuntos
Allolevivirus/química , Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Ouro/química , Nanopartículas Metálicas/química , Células A549 , Animais , Antineoplásicos/química , Capsídeo/química , Proteínas do Capsídeo/química , Doxorrubicina/química , Portadores de Fármacos/efeitos da radiação , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Ouro/efeitos da radiação , Ouro/toxicidade , Humanos , Hipertermia Induzida/métodos , Luz , Nanopartículas Metálicas/efeitos da radiação , Nanopartículas Metálicas/toxicidade , Camundongos , Tamanho da Partícula , Fototerapia/métodos , Porosidade , Estudo de Prova de Conceito , Células RAW 264.7 , RNA/química , RNA/toxicidade
8.
Dis Model Mech ; 10(8): 993-1003, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28623239

RESUMO

The myotonic dystrophies are prototypic toxic RNA gain-of-function diseases. Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are caused by different unstable, noncoding microsatellite repeat expansions - (CTG)DM1 in DMPK and (CCTG)DM2 in CNBP Although transcription of mutant repeats into (CUG)DM1 or (CCUG)DM2 appears to be necessary and sufficient to cause disease, their pathomechanisms remain incompletely understood. To study the mechanisms of (CCUG)DM2 toxicity and develop a convenient model for drug screening, we generated a transgenic DM2 model in the fruit fly Drosophila melanogaster with (CCUG)n repeats of variable length (n=16 and 106). Expression of noncoding (CCUG)106, but not (CCUG)16, in muscle and retinal cells led to the formation of ribonuclear foci and mis-splicing of genes implicated in DM pathology. Mis-splicing could be rescued by co-expression of human MBNL1, but not by CUGBP1 (CELF1) complementation. Flies with (CCUG)106 displayed strong disruption of external eye morphology and of the underlying retina. Furthermore, expression of (CCUG)106 in developing retinae caused a strong apoptotic response. Inhibition of apoptosis rescued the retinal disruption in (CCUG)106 flies. Finally, we tested two chemical compounds that have shown therapeutic potential in DM1 models. Whereas treatment of (CCUG)106 flies with pentamidine had no effect, treatment with a PKR inhibitor blocked both the formation of RNA foci and apoptosis in retinae of (CCUG)106 flies. Our data indicate that expression of expanded (CCUG)DM2 repeats is toxic, causing inappropriate cell death in affected fly eyes. Our Drosophila DM2 model might provide a convenient tool for in vivo drug screening.


Assuntos
Apoptose/efeitos dos fármacos , Expansão das Repetições de DNA/genética , Drosophila melanogaster/genética , Distrofia Miotônica/genética , RNA/toxicidade , Animais , Apoptose/genética , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efeitos dos fármacos , Humanos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Pentamidina/farmacologia , Células Fotorreceptoras de Invertebrados/efeitos dos fármacos , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patologia , Inibidores de Proteínas Quinases/farmacologia , Splicing de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retina/anormalidades , Retina/efeitos dos fármacos , Retina/patologia , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/metabolismo
9.
Nature ; 539(7628): 197-206, 2016 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-27830784

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive and uniformly fatal neurodegenerative disease. A plethora of genetic factors have been identified that drive the degeneration of motor neurons in ALS, increase susceptibility to the disease or influence the rate of its progression. Emerging themes include dysfunction in RNA metabolism and protein homeostasis, with specific defects in nucleocytoplasmic trafficking, the induction of stress at the endoplasmic reticulum and impaired dynamics of ribonucleoprotein bodies such as RNA granules that assemble through liquid-liquid phase separation. Extraordinary progress in understanding the biology of ALS provides new reasons for optimism that meaningful therapies will be identified.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/fisiopatologia , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/terapia , Animais , Transporte Biológico , Proteína C9orf72 , Estresse do Retículo Endoplasmático/genética , Demência Frontotemporal/genética , Humanos , Sistema Nervoso/patologia , Sistema Nervoso/fisiopatologia , Organelas/genética , Organelas/metabolismo , Organelas/patologia , Príons/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteólise , RNA/biossíntese , RNA/genética , RNA/metabolismo , RNA/toxicidade
10.
ACS Comb Sci ; 18(9): 527-47, 2016 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-27509068

RESUMO

RNA nanostructures can be programmed to exhibit defined sizes, shapes and stoichiometries from naturally occurring or de novo designed RNA motifs. These constructs can be used as scaffolds to attach functional moieties, such as ligand binding motifs or gene expression regulators, for nanobiology applications. This review is focused on four areas of importance to RNA nanotechnology: the types of RNAs of particular interest for nanobiology, the assembly of RNA nanoconstructs, the challenges of cellular delivery of RNAs in vivo, and the delivery carriers that aid in the matter. The available strategies for the design of nucleic acid nanostructures, as well as for formulation of their carriers, make RNA nanotechnology an important tool in both basic research and applied biomedical science.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , RNA/administração & dosagem , RNA/química , Animais , Aptâmeros de Nucleotídeos/química , Química Farmacêutica , Biologia Computacional , Desenho de Fármacos , Liberação Controlada de Fármacos , Terapia Genética , Vetores Genéticos , Humanos , Terapia de Alvo Molecular/métodos , Nanotecnologia , RNA/imunologia , RNA/toxicidade , Interferência de RNA , Splicing de RNA , Propriedades de Superfície
11.
Dis Model Mech ; 9(3): 321-34, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26839389

RESUMO

Polyglutamine (polyQ) diseases represent a group of progressive neurodegenerative disorders that are caused by abnormal expansion of CAG triplet nucleotides in disease genes. Recent evidence indicates that not only mutant polyQ proteins, but also their corresponding mutant RNAs, contribute to the pathogenesis of polyQ diseases. Here, we describe the identification of a 13-amino-acid peptide, P3, which binds directly and preferentially to long-CAG RNA within the pathogenic range. When administered to cell and Drosophila disease models, as well as to patient-derived fibroblasts, P3 inhibited expanded-CAG-RNA-induced nucleolar stress and suppressed neurotoxicity. We further examined the combined therapeutic effect of P3 and polyQ-binding peptide 1 (QBP1), a well-characterized polyQ protein toxicity inhibitor, on neurodegeneration. When P3 and QBP1 were co-administered to disease models, both RNA and protein toxicities were effectively mitigated, resulting in a notable improvement of neurotoxicity suppression compared with the P3 and QBP1 single-treatment controls. Our findings indicate that targeting toxic RNAs and/or simultaneous targeting of toxic RNAs and their corresponding proteins could open up a new therapeutic strategy for treating polyQ degeneration.


Assuntos
Drosophila melanogaster/metabolismo , Peptídeos/farmacologia , RNA/toxicidade , Sequência de Aminoácidos , Animais , Morte Celular/efeitos dos fármacos , Drosophila melanogaster/efeitos dos fármacos , Células HEK293 , Humanos , Modelos Biológicos , Degeneração Neural/patologia , Peptídeos/administração & dosagem , Peptídeos/química , Peptídeos/toxicidade , Fosfoproteínas/metabolismo , RNA Ribossômico/genética , Proteínas de Ligação a RNA/metabolismo , Estresse Fisiológico , Relação Estrutura-Atividade , Transcrição Gênica/efeitos dos fármacos , Transfecção , Expansão das Repetições de Trinucleotídeos/genética , Nucleolina
12.
ACS Chem Biol ; 10(12): 2706-15, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26414664

RESUMO

There are many RNA targets in the transcriptome to which small molecule chemical probes and lead therapeutics are desired. However, identifying compounds that bind and modulate RNA function in cellulo is difficult. Although rational design approaches have been developed, they are still in their infancies and leave many RNAs "undruggable". In an effort to develop a small molecule library that is biased for binding RNA, we computationally identified "drug-like" compounds from screening collections that have favorable properties for binding RNA and for suitability as lead drugs. As proof-of-concept, this collection was screened for binding to and modulating the cellular dysfunction of the expanded repeating RNA (r(CUG)(exp)) that causes myotonic dystrophy type 1. Hit compounds bind the target in cellulo, as determined by the target identification approach Competitive Chemical Cross-Linking and Isolation by Pull-down (C-ChemCLIP), and selectively improve several disease-associated defects. The best compounds identified from our 320-member library are more potent in cellulo than compounds identified by high-throughput screening (HTS) campaigns against this RNA. Furthermore, the compound collection has a higher hit rate (9% compared to 0.01-3%), and the bioactive compounds identified are not charged; thus, RNA can be "drugged" with compounds that have favorable pharmacological properties. Finally, this RNA-focused small molecule library may serve as a useful starting point to identify lead "drug-like" chemical probes that affect the biological (dys)function of other RNA targets by direct target engagement.


Assuntos
Desenho Assistido por Computador , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/fisiopatologia , RNA/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Concentração Inibidora 50 , Estrutura Molecular , RNA/toxicidade , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo
13.
Hum Mol Genet ; 24(1): 251-64, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25168381

RESUMO

RNA toxicity is implicated in a number of disorders; especially those associated with expanded repeat sequences, such as myotonic dystrophy (DM1). Previously, we have shown increased NKX2-5 expression in RNA toxicity associated with DM1. Here, we investigate the relationship between NKX2-5 expression and muscle pathology due to RNA toxicity. In skeletal muscle from mice with RNA toxicity and individuals with DM1, expression of Nkx2-5 or NKX2-5 and its downstream targets are significantly correlated with severity of histopathology. Using C2C12 myoblasts, we show that over-expression of NKX2-5 or mutant DMPK 3'UTR results in myogenic differentiation defects, which can be rescued by knockdown of Nkx2-5, despite continued toxic RNA expression. Furthermore, in a mouse model of NKX2-5 over-expression, we find defects in muscle regeneration after induced damage, similar to those seen in mice with RNA toxicity. Using mouse models of Nkx2-5 over-expression and depletion, we find that NKX2-5 levels modify disease phenotypes in mice with RNA toxicity.


Assuntos
Proteínas de Homeodomínio/genética , Músculo Esquelético/patologia , Distrofias Musculares/genética , RNA/toxicidade , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Genes Modificadores , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Miotonina Proteína Quinase/genética , Fatores de Transcrição/metabolismo
14.
Mol Neurobiol ; 49(3): 1513-31, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24293103

RESUMO

Ataxin-3 is a ubiquitously expressed deubiqutinating enzyme with important functions in the proteasomal protein degradation pathway and regulation of transcription. The C-terminus of the ataxin-3 protein contains a polyglutamine (PolyQ) region that, when mutationally expanded to over 52 glutamines, causes the neurodegenerative disease spinocerebellar ataxia 3 (SCA3). In spite of extensive research, the molecular mechanisms underlying the cellular toxicity resulting from mutant ataxin-3 remain elusive and no preventive treatment is currently available. It has become clear over the last decade that the hallmark intracellular ataxin-3 aggregates are likely not the main toxic entity in SCA3. Instead, the soluble PolyQ containing fragments arising from proteolytic cleavage of ataxin-3 by caspases and calpains are now regarded to be of greater influence in pathogenesis. In addition, recent evidence suggests potential involvement of a RNA toxicity component in SCA3 and other PolyQ expansion disorders, increasing the pathogenic complexity. Herein, we review the functioning of ataxin-3 and the involvement of known protein and RNA toxicity mechanisms of mutant ataxin-3 that have been discovered, as well as future opportunities for therapeutic intervention.


Assuntos
Doença de Machado-Joseph/genética , Doença de Machado-Joseph/terapia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , RNA/toxicidade , Proteínas Repressoras/fisiologia , Animais , Antioxidantes/metabolismo , Antioxidantes/uso terapêutico , Ataxina-3 , Humanos , Doença de Machado-Joseph/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , RNA/genética
15.
Neuron ; 80(2): 257-8, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24139028

RESUMO

Hexanucleotide repeat expansions in C9ORF72 cause neurodegeneration in FTD and ALS by unknown mechanisms. A new report, by Donnelly et al. (2013), finds that these repeats trigger a pathogenic gain-of-function cascade that can be corrected by suppressing expression of the repeat transcript, paving the way for therapeutic strategies aimed at eliminating the toxic RNA.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Demência Frontotemporal/metabolismo , Oligonucleotídeos Antissenso/uso terapêutico , Proteínas/metabolismo , RNA/toxicidade , Proteína C9orf72 , Humanos
16.
Neuron ; 80(2): 415-28, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24139042

RESUMO

A hexanucleotide GGGGCC repeat expansion in the noncoding region of the C9ORF72 gene is the most common genetic abnormality in familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The function of the C9ORF72 protein is unknown, as is the mechanism by which the repeat expansion could cause disease. Induced pluripotent stem cell (iPSC)-differentiated neurons from C9ORF72 ALS patients revealed disease-specific (1) intranuclear GGGGCCexp RNA foci, (2) dysregulated gene expression, (3) sequestration of GGGGCCexp RNA binding protein ADARB2, and (4) susceptibility to excitotoxicity. These pathological and pathogenic characteristics were confirmed in ALS brain and were mitigated with antisense oligonucleotide (ASO) therapeutics to the C9ORF72 transcript or repeat expansion despite the presence of repeat-associated non-ATG translation (RAN) products. These data indicate a toxic RNA gain-of-function mechanism as a cause of C9ORF72 ALS and provide candidate antisense therapeutics and candidate human pharmacodynamic markers for therapy.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Demência Frontotemporal/metabolismo , Oligonucleotídeos Antissenso/uso terapêutico , Proteínas/metabolismo , RNA/toxicidade , Adenosina Desaminase/metabolismo , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Proteína C9orf72 , Contagem de Células , Relação Dose-Resposta a Droga , Demência Frontotemporal/tratamento farmacológico , Demência Frontotemporal/genética , Ácido Glutâmico/toxicidade , Humanos , Células-Tronco Pluripotentes Induzidas , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Proteínas/genética , RNA/genética , RNA/metabolismo , Proteínas de Ligação a RNA , Sequências Repetitivas de Ácido Nucleico
17.
J Mol Med (Berl) ; 91(6): 683-91, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23512265

RESUMO

In Huntington's disease and other polyglutamine (polyQ) disorders, mutant proteins containing a long polyQ stretch are well documented as the trigger of numerous aberrant cellular processes that primarily lead to degeneration and, ultimately, the death of neuronal cells. However, mutant transcripts containing expanded CAG repeats may also be toxic and contribute to cellular dysfunction. The exact nature and importance of RNA toxicity in polyQ diseases are only beginning to be recognized, and the first insights have mainly resulted from studies using simple model systems. In this review, we briefly present the basic mechanisms of protein toxicity in polyQ disorders and RNA toxicity in myotonic dystrophy type 1 and discuss recent results suggesting that the pathogenesis of polyQ diseases may also be mediated by mutant transcripts. This review is focused on the experimental systems used thus far to demonstrate RNA toxicity in polyQ disorders and the design of new systems that will be more relevant to the human disease situation and capable of separating RNA toxicity from protein toxicity.


Assuntos
Doenças Neurodegenerativas/genética , Peptídeos/genética , RNA/toxicidade , Animais , Pesquisa Biomédica , Humanos , Modelos Genéticos
18.
Biochim Biophys Acta ; 1832(9): 1390-409, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23500957

RESUMO

Mutant ribonucleic acid (RNA) molecules can be toxic to the cell, causing human disease through trans-acting dominant mechanisms. RNA toxicity was first described in myotonic dystrophy type 1, a multisystemic disorder caused by the abnormal expansion of a non-coding trinucleotide repeat sequence. The development of multiple and complementary animal models of disease has greatly contributed to clarifying the complex disease pathways mediated by toxic RNA molecules. RNA toxicity is not limited to myotonic dystrophy and spreads to an increasing number of human conditions, which share some unifying pathogenic events mediated by toxic RNA accumulation and disruption of RNA-binding proteins. The remarkable progress in the dissection of disease pathobiology resulted in the rational design of molecular therapies, which have been successfully tested in animal models. Toxic RNA diseases, and in particular myotonic dystrophy, clearly illustrate the critical contribution of animal models of disease in translational research: from gene mutation to disease mechanisms, and ultimately to therapy development. This article is part of a Special Issue entitled: Animal Models of Disease.


Assuntos
Modelos Animais de Doenças , Distrofia Miotônica/etiologia , Distrofia Miotônica/terapia , RNA/toxicidade , Repetições de Trinucleotídeos/genética , Animais , Descoberta de Drogas , Humanos , RNA/genética
19.
Nucleic Acid Ther ; 23(2): 109-17, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23308382

RESUMO

Expansions of CUG trinucleotide sequences in RNA transcripts provide the basis for toxic RNA gain-of-function that leads to detrimental changes in RNA metabolism. A CTG repeat element normally resides in the 3' untranslated region of the dystrophia myotonica-protein kinase (DMPK) gene, but when expanded it is the genetic lesion of myotonic dystrophy type 1 (DM1), a hereditary neuromuscular disease. The pathogenic DMPK transcript containing the CUG expansion is retained in ribonuclear foci as part of a complex with RNA-binding proteins such as muscleblind-like 1 (MBNL1), resulting in aberrant splicing of numerous RNA transcripts and consequent physiological abnormalities including myotonia. Herein, we demonstrate molecular and physiological amelioration of the toxic effects of mutant RNA in the HSA(LR) mouse model of DM1 by systemic administration of peptide-linked morpholino (PPMO) antisense oligonucleotides bearing a CAG repeat sequence. Intravenous administration of PPMO conjugates to HSA(LR) mice led to redistribution of Mbnl1 protein in myonuclei and corrections in abnormal RNA splicing. Additionally, myotonia was completely eliminated in PPMO-treated HSA(LR) mice. These studies provide proof of concept that neutralization of RNA toxicity by systemic delivery of antisense oligonucleotides that target the CUG repeat is an effective therapeutic approach for treating the skeletal muscle aspects of DM1 pathology.


Assuntos
Morfolinos/administração & dosagem , Distrofia Miotônica/genética , Peptídeos/administração & dosagem , Proteínas de Ligação a RNA/genética , Regiões 3' não Traduzidas/genética , Animais , Humanos , Camundongos , Morfolinos/química , Mutação , Distrofia Miotônica/metabolismo , Distrofia Miotônica/patologia , Miotonina Proteína Quinase , Oligonucleotídeos Antissenso/administração & dosagem , Peptídeos/química , Proteínas Serina-Treonina Quinases/genética , RNA/genética , RNA/toxicidade , Splicing de RNA/genética , Expansão das Repetições de Trinucleotídeos/genética , Repetições de Trinucleotídeos/genética
20.
Hum Mol Genet ; 20(24): 4810-21, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21933837

RESUMO

Spinocerebellar ataxia type 3 is one of the polyglutamine (polyQ) diseases, which are caused by a CAG-repeat expansion within the coding region of the associated genes. The CAG repeat specifies glutamine, and the expanded polyQ domain mutation confers dominant toxicity on the protein. Traditionally, studies have focused on protein toxicity in polyQ disease mechanisms. Recent findings, however, demonstrate that the CAG-repeat RNA, which encodes the toxic polyQ protein, also contributes to the disease in Drosophila. To provide insights into the nature of the RNA toxicity, we extracted brain-enriched RNA from flies expressing a toxic CAG-repeat mRNA (CAG100) and a non-toxic interrupted CAA/G mRNA repeat (CAA/G105) for microarray analysis. This approach identified 160 genes that are differentially expressed specifically in CAG100 flies. Functional annotation clustering analysis revealed several broad ontologies enriched in the CAG100 gene list, including iron ion binding and nucleotide binding. Intriguingly, transcripts for the Hsp70 genes, a powerful suppressor of polyQ and other human neurodegenerative diseases, were also upregulated. We therefore tested and showed that upregulation of heat shock protein 70 mitigates CAG-repeat RNA toxicity. We then assessed whether other modifiers of the pathogenic, expanded Ataxin-3 polyQ protein could also modify the CAG-repeat RNA toxicity. This approach identified the co-chaperone Tpr2, the transcriptional regulator Dpld, and the RNA-binding protein Orb2 as modifiers of both polyQ protein toxicity and CAG-repeat RNA-based toxicity. These findings suggest an overlap in the mechanisms of RNA and protein-based toxicity, providing insights into the pathogenicity of the RNA in polyQ disease.


Assuntos
Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Redes Reguladoras de Genes/genética , Genes de Insetos/genética , RNA/toxicidade , Expansão das Repetições de Trinucleotídeos/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Testes Genéticos , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Degeneração Neural/genética , Degeneração Neural/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/toxicidade , RNA não Traduzido/genética , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...