Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.082
Filtrar
1.
Bioorg Chem ; 150: 107595, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38968904

RESUMO

Combined therapies play a key role in the fight against complex pathologies, such as cancer and related drug-resistance issues. This is particularly relevant in targeted therapies where inhibition of the drug target can be overcome by cross-activating complementary pathways. Unfortunately, the drug combinations approved to date -mostly based on small molecules- face several problems such as toxicity effects, which limit their clinical use. To address these issues, we have designed a new class of RNase H-sensitive construct (3ASO) that can be disassembled intracellularly upon cell entry, leading to the simultaneous release of three different therapeutic oligonucleotides (ONs), tackling each of them the mRNA of a different protein. Here, we used Escherichia coli RNase H1 as a model to study an unprecedented mode of recognition and cleavage, that is mainly dictated by the topology of our RNA·DNA-based hybrid construct. As a model system for our technology we have created 3ASO constructs designed to specifically inhibit the expression of HER2, Akt and Hsp27 in HER2+ breast cancer cells. These trifunctional ON tools displayed very low toxicity and good levels of antiproliferative activity in HER2+ breast cancer cells. The present study will be of great potential in the fight against complex pathologies involving multiple mRNA targets, as the proposed cleavable designs will allow the efficient single-dose administration of different ON drugs simultaneously.


Assuntos
Proliferação de Células , Oligonucleotídeos Antissenso , Ribonuclease H , Ribonuclease H/metabolismo , Ribonuclease H/antagonistas & inibidores , Humanos , Proliferação de Células/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Estrutura Molecular , Relação Estrutura-Atividade , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Receptor ErbB-2/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Linhagem Celular Tumoral , Escherichia coli/efeitos dos fármacos
2.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38892298

RESUMO

Periostin, a multifunctional 90 kDa protein, plays a pivotal role in the pathogenesis of fibrosis across various tissues, including skeletal muscle. It operates within the transforming growth factor beta 1 (Tgf-ß1) signalling pathway and is upregulated in fibrotic tissue. Alternative splicing of Periostin's C-terminal region leads to six protein-coding isoforms. This study aimed to elucidate the contribution of the isoforms containing the amino acids encoded by exon 17 (e17+ Periostin) to skeletal muscle fibrosis and investigate the therapeutic potential of manipulating exon 17 splicing. We identified distinct structural differences between e17+ Periostin isoforms, affecting their interaction with key fibrotic proteins, including Tgf-ß1 and integrin alpha V. In vitro mouse fibroblast experimentation confirmed the TGF-ß1-induced upregulation of e17+ Periostin mRNA, mitigated by an antisense approach that induces the skipping of exon 17 of the Postn gene. Subsequent in vivo studies in the D2.mdx mouse model of Duchenne muscular dystrophy (DMD) demonstrated that our antisense treatment effectively reduced e17+ Periostin mRNA expression, which coincided with reduced full-length Periostin protein expression and collagen accumulation. The grip strength of the treated mice was rescued to the wild-type level. These results suggest a pivotal role of e17+ Periostin isoforms in the fibrotic pathology of skeletal muscle and highlight the potential of targeted exon skipping strategies as a promising therapeutic approach for mitigating fibrosis-associated complications.


Assuntos
Processamento Alternativo , Moléculas de Adesão Celular , Éxons , Fibrose , Camundongos Endogâmicos mdx , Oligonucleotídeos Antissenso , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Camundongos , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/genética , Fibroblastos/metabolismo , Modelos Animais de Doenças , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Masculino
3.
Molecules ; 29(11)2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38893532

RESUMO

Spinal muscular atrophy (SMA) is a severe neuromuscular disorder that is caused by mutations in the survival motor neuron 1 (SMN1) gene, hindering the production of functional survival motor neuron (SMN) proteins. Antisense oligonucleotides (ASOs), a versatile DNA-like drug, are adept at binding to target RNA to prevent translation or promote alternative splicing. Nusinersen is an FDA-approved ASO for the treatment of SMA. It effectively promotes alternative splicing in pre-mRNA transcribed from the SMN2 gene, an analog of the SMN1 gene, to produce a greater amount of full-length SMN protein, to compensate for the loss of functional protein translated from SMN1. Despite its efficacy in ameliorating SMA symptoms, the cellular uptake of these ASOs is suboptimal, and their inability to penetrate the CNS necessitates invasive lumbar punctures. Cell-penetrating peptides (CPPs), which can be conjugated to ASOs, represent a promising approach to improve the efficiency of these treatments for SMA and have the potential to transverse the blood-brain barrier to circumvent the need for intrusive intrathecal injections and their associated adverse effects. This review provides a comprehensive analysis of ASO therapies, their application for the treatment of SMA, and the encouraging potential of CPPs as delivery systems to improve ASO uptake and overall efficiency.


Assuntos
Peptídeos Penetradores de Células , Atrofia Muscular Espinal , Oligonucleotídeos Antissenso , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/farmacologia , Humanos , Atrofia Muscular Espinal/tratamento farmacológico , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/farmacologia , Animais , Oligonucleotídeos/química , Oligonucleotídeos/farmacologia , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos
4.
J Mater Chem B ; 12(26): 6492-6499, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38872610

RESUMO

Antisense oligonucleotides (ASOs) are molecules used to regulate RNA expression by targeting specific RNA sequences. One specific type of ASO, known as neutralized DNA (nDNA), contains site-specific methyl phosphotriester (MPTE) linkages on the phosphate backbone, changing the negatively charged DNA phosphodiester into a neutralized MPTE with designed locations. While nDNA has previously been employed as a sensitive nucleotide sequencing probe for the PCR, the potential of nDNA in intracellular RNA regulation and gene therapy remains underexplored. Our study aims to evaluate the regulatory capacity of nDNA as an ASO probe in cellular gene expression. We demonstrated that by tuning MPTE locations, partially and intermediately methylated nDNA loaded onto mesoporous silica nanoparticles (MSNs) can effectively knock down the intracellular miRNA, subsequently resulting in downstream mRNA regulation in colorectal cancer cell HCT116. Additionally, the nDNA ASO-loaded MSNs exhibit superior efficacy in reducing miR-21 levels over 72 hours compared to the efficacy of canonical DNA ASO-loaded MSNs. The reduction in the miR-21 level subsequently resulted in the enhanced mRNA levels of tumour-suppressing genes PTEN and PDCD4. Our findings underscore the potential of nDNA in gene therapies, especially in cancer treatment via a fine-tuned methylation location.


Assuntos
DNA , MicroRNAs , Nanopartículas , Dióxido de Silício , Dióxido de Silício/química , MicroRNAs/genética , MicroRNAs/metabolismo , Humanos , Nanopartículas/química , DNA/química , Porosidade , Células HCT116 , Fosfatos/química , Tamanho da Partícula , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/farmacologia , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Propriedades de Superfície , Proteínas de Ligação a RNA/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Reguladoras de Apoptose/genética
5.
Drugs ; 84(6): 637-643, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38849700

RESUMO

Increasing evidence has implicated lipoprotein(a) [Lp(a)] in the causality of atherosclerosis and calcific aortic stenosis. This has stimulated immense interest in developing novel approaches to integrating Lp(a) into the setting of cardiovascular prevention. Current guidelines advocate universal measurement of Lp(a) levels, with the potential to influence cardiovascular risk assessment and triage of higher-risk patients to use of more intensive preventive therapies. In parallel, considerable activity has been undertaken to develop novel therapeutics with the potential to achieve selective and substantial reductions in Lp(a) levels. Early studies of antisense oligonucleotides (e.g., mipomersen, pelacarsen), RNA interference (e.g., olpasiran, zerlasiran, lepodisiran) and small molecule inhibitors (e.g., muvalaplin) have demonstrated effective Lp(a) lowering and good tolerability. These agents are moving forward in clinical development, in order to determine whether Lp(a) lowering reduces cardiovascular risk. The results of these studies have the potential to transform our approach to the prevention of cardiovascular disease.


Assuntos
Doenças Cardiovasculares , Lipoproteína(a) , Oligonucleotídeos Antissenso , Humanos , Lipoproteína(a)/sangue , Doenças Cardiovasculares/prevenção & controle , Doenças Cardiovasculares/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Oligonucleotídeos Antissenso/farmacologia , Aterosclerose/tratamento farmacológico , Interferência de RNA , Oligonucleotídeos/uso terapêutico , Oligonucleotídeos/farmacologia , Animais
6.
Int J Biol Macromol ; 272(Pt 2): 132890, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38848829

RESUMO

The lack of more effective therapies for breast cancer has enhanced mortality among breast cancer patients. Recent efforts have established efficient treatments to reduce breast cancer-related deaths. The ever-increasing attraction to employing biocompatible polysaccharide-based nanostructures as delivery systems has created interest in various disease therapies, especially breast cancer treatment. A wide range of therapeutic cargo comprising bioactive or chemical drugs, oligonucleotides, peptides, and targeted biomarkers have been considered to comprehend their anti-cancer effects against breast cancer. Some limitations of naked agents or undesired constructs, such as no or low bioavailability, enzymatic digestion, short-range stability, low-cellular uptake, poor solubility, and low surface area, have lessened their effectiveness. However, nanoscale formulations of therapeutic ingredients have provided a promising platform to address the mentioned concerns. For instance, some capable polysaccharides, including cellulose, pectin, chitosan, alginate, and dextran, were developed as breast cancer therapeutics with great nanoparticle structures. This review carefully examines the characteristics of beneficial polysaccharides that are utilized in the formation of nanoparticles (NPs). It also highlights the applications of antisense oligonucleotides (ASOs), and NPs made from polysaccharides in the treatment of breast cancer and suggests ways to enhance these particles for future research.


Assuntos
Neoplasias da Mama , Nanoestruturas , Oligonucleotídeos Antissenso , Polissacarídeos , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/terapia , Polissacarídeos/química , Feminino , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/uso terapêutico , Nanoestruturas/química , Sistemas de Liberação de Medicamentos , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Portadores de Fármacos/química , Nanopartículas/química
7.
Int J Mol Sci ; 25(11)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38891760

RESUMO

Dysferlin is a large transmembrane protein involved in critical cellular processes including membrane repair and vesicle fusion. Mutations in the dysferlin gene (DYSF) can result in rare forms of muscular dystrophy; Miyoshi myopathy; limb girdle muscular dystrophy type 2B (LGMD2B); and distal myopathy. These conditions are collectively known as dysferlinopathies and are caused by more than 600 mutations that have been identified across the DYSF gene to date. In this review, we discuss the key molecular and clinical features of LGMD2B, the causative gene DYSF, and the associated dysferlin protein structure. We also provide an update on current approaches to LGMD2B diagnosis and advances in drug development, including splice switching antisense oligonucleotides. We give a brief update on clinical trials involving adeno-associated viral gene therapy and the current progress on CRISPR/Cas9 mediated therapy for LGMD2B, and then conclude by discussing the prospects of antisense oligomer-based intervention to treat selected mutations causing dysferlinopathies.


Assuntos
Disferlina , Terapia Genética , Distrofia Muscular do Cíngulo dos Membros , Mutação , Humanos , Distrofia Muscular do Cíngulo dos Membros/terapia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Disferlina/genética , Disferlina/metabolismo , Terapia Genética/métodos , Oligonucleotídeos Antissenso/uso terapêutico , Animais
8.
Med ; 5(6): 493-494, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38878767

RESUMO

Reducing the synthesis of apoC-III reduces fasting triglycerides in individuals lacking lipoprotein lipase activity. Recently, Stroes et al.1 published a phase 3 trial on the effects of olezarsen, a third-generation antisense oligonucleotide that blocks apoC-III mRNA, on triglycerides and risk of acute pancreatitis.


Assuntos
Apolipoproteína C-III , Hiperlipoproteinemia Tipo I , Oligonucleotídeos , Triglicerídeos , Humanos , Apolipoproteína C-III/genética , Apolipoproteína C-III/sangue , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/sangue , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Triglicerídeos/sangue , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Pancreatite/genética , Benzimidazóis
9.
BioDrugs ; 38(4): 511-526, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38914784

RESUMO

Antisense oligonucleotides (ASOs) are single stranded nucleic acids that target RNA. The US Food and Drug Administration has approved ASOs for several diseases. ASOs utilize three principal modes of action (MOA). The first MOA is initiated by base-pairing between the ASO and its target mRNA, followed by RNase H-dependent mRNA degradation. The second MOA is triggered by ASOs that occlude splice acceptor sites in pre-mRNAs leading to skipping of a mutation-bearing exon. The third MOA involves ASOs that sterically hinder mRNA function, often inhibiting translation. ASOs contain a variety of modifications to the sugar-phosphate backbone and bases that stabilize the ASO or render them resistant to RNase activity. RNase H-dependent ASOs include inotersen and eplontersen (for hereditary transthyretin amyloidosis), fomiversen (for opportunistic cytomegalovirus infection), mipomersen (for familial hypercholesterolemia), and tofersen [for amyotrophic lateral sclerosis (ALS)]. Splice modulating ASOs include nursinersen (for spinal muscular atrophy) and eteplirsen, golodirsen, viltolarsen, and casimersen (all for the treatment of Duchenne muscular dystrophy). In addition, a designer ASO, milasen, was used to treat a single individual afflicted with Batten disease. Since ASO design relies principally upon knowledge of mRNA sequence, the bench to bedside pipeline for ASOs is expedient compared with protein-directed drugs. [Graphical abstract available.].


Assuntos
Aprovação de Drogas , Oligonucleotídeos Antissenso , United States Food and Drug Administration , Humanos , Oligonucleotídeos Antissenso/uso terapêutico , Oligonucleotídeos Antissenso/farmacologia , Estados Unidos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Animais , Splicing de RNA/efeitos dos fármacos
10.
Medicine (Baltimore) ; 103(26): e38767, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38941378

RESUMO

Hereditary transthyretin-mediated amyloidosis (ATTRv amyloidosis), known as Corino de Andrade disease, is a rare neurodegenerative disorder with a significant global impact characterized by the misfolding of transthyretin (TTR) protein leading to amyloid aggregation, ATTRv amyloidosis, especially with polyneuropathy, poses a considerable challenge in managing its rapid progression and debilitating effects. This mini-review focuses on the recent advancements in the treatment landscape for ATTRv amyloidosis with polyneuropathy, specifically the RNA interference therapeutic Vutrisiran and the ligand-conjugated antisense oligonucleotide Eplontersen. We aim to provide a comprehensive overview of the mechanisms, current evidence from clinical trials, and future directions for these novel therapeutic agents. Vutrisiran and Eplontersen have demonstrated significant clinical efficacy in improving neuropathic impairment, quality of life, and serum TTR levels in various trials. The distinct mechanistic approaches of these therapies, coupled with their acceptable safety profiles, offer promising avenues for addressing the complexities of ATTRv amyloidosis with polyneuropathy. The introduction of Vutrisiran and Eplontersen marks a pivotal moment in the quest for effective therapies against ATTRv amyloidosis with polyneuropathy. While clinical evidence is promising, ongoing research is crucial to deepen mechanistic understanding and address research gaps. Future perspectives include the potential expansion of therapeutic options and a more inclusive approach to cater to the diverse needs of individuals globally. This mini-review provides valuable insights into the evolving landscape of ATTRv amyloidosis management and sets the stage for further exploration in this challenging domain.


Assuntos
Neuropatias Amiloides Familiares , Polineuropatias , Humanos , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/terapia , Polineuropatias/genética , Oligonucleotídeos/uso terapêutico , Oligonucleotídeos Antissenso/uso terapêutico , Pré-Albumina/genética , Qualidade de Vida
11.
Kidney Int ; 106(1): 21-23, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38906653

RESUMO

Chronic hemodialysis patients exhibit an excessive cardiovascular risk and a marked increase in both thromboembolism and bleeding episodes. Factor XI inhibition may provide anticoagulation, with a low risk of bleeding, and several factor XI inhibitors, including fesomersen, an antisense oligonucleotide, are under development. Recently, a phase 2 study of fesomersen showed a good safety profile in chronic hemodialysis patients and suggested that clotting rates of the arteriovenous fistula and the dialysis circuit are lower.


Assuntos
Anticoagulantes , Fator XI , Hemorragia , Diálise Renal , Humanos , Diálise Renal/efeitos adversos , Anticoagulantes/efeitos adversos , Anticoagulantes/uso terapêutico , Hemorragia/induzido quimicamente , Hemorragia/prevenção & controle , Fator XI/antagonistas & inibidores , Fator XI/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Oligonucleotídeos Antissenso/uso terapêutico , Oligonucleotídeos Antissenso/efeitos adversos , Oligonucleotídeos Antissenso/administração & dosagem , Tromboembolia/prevenção & controle , Tromboembolia/etiologia , Derivação Arteriovenosa Cirúrgica/efeitos adversos
12.
Nat Commun ; 15(1): 4756, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38834544

RESUMO

Given the absence of approved treatments for pathogenic variants in Peripherin-2 (PRPH2), it is imperative to identify a universally effective therapeutic target for PRPH2 pathogenic variants. To test the hypothesis that formation of the elongated discs in presence of PRPH2 pathogenic variants is due to the presence of the full complement of rhodopsin in absence of the required amounts of functional PRPH2. Here we demonstrate the therapeutic potential of reducing rhodopsin levels in ameliorating disease phenotype in knockin models for p.Lys154del (c.458-460del) and p.Tyr141Cys (c.422 A > G) in PRPH2. Reducing rhodopsin levels improves physiological function, mitigates the severity of disc abnormalities, and decreases retinal gliosis. Additionally, intravitreal injections of a rhodopsin-specific antisense oligonucleotide successfully enhance the physiological function of photoreceptors and improves the ultrastructure of discs in mutant mice. Presented findings shows that reducing rhodopsin levels is an effective therapeutic strategy for the treatment of inherited retinal degeneration associated with PRPH2 pathogenic variants.


Assuntos
Periferinas , Rodopsina , Periferinas/genética , Periferinas/metabolismo , Animais , Rodopsina/genética , Rodopsina/metabolismo , Camundongos , Humanos , Modelos Animais de Doenças , Regulação para Baixo , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Degeneração Retiniana/terapia , Oligonucleotídeos Antissenso/genética , Retina/metabolismo , Retina/patologia , Doenças Retinianas/genética , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Doenças Retinianas/terapia , Camundongos Endogâmicos C57BL , Mutação , Feminino , Técnicas de Introdução de Genes , Masculino
13.
Curr Neurol Neurosci Rep ; 24(8): 255-264, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38861215

RESUMO

PURPOSE OF REVIEW: Huntington's disease (HD) is an autosomal-dominant disorder caused by a pathological expansion of a trinucleotide repeat (CAG) on exon 1 of the huntingtin (HTT) gene. HD is characterized by the presence of chorea, alongside other hyperkinesia, parkinsonism and a combination of cognitive and behavioural features. Currently, there are no disease-modifying therapies (DMTs) for HD, and the only intervention(s) with approved indication target the treatment of chorea. This article reviews recent research on the clinical development of DMTs and newly developed tools that enhance clinical trial design towards a successful DMT in the future. RECENT FINDINGS: HD is living in an era of target-specific drug development with emphasis on the mechanisms related to mutant Huntingtin (HTT) protein. Examples include antisense oligonucleotides (ASO), splicing modifiers and microRNA molecules that aim to reduce the levels of mutant HTT protein. After initial negative results with ASO molecules Tominersen and WVE-120101/ WVE-120102, the therapeutic landscape continues to expand, with various trials currently under development to document proof-of-concept and safety/tolerability. Immune-targeted therapies have also been evaluated in early-phase clinical trials, with promising preliminary findings. The possibility of quantifying mHTT in CSF, along with the development of an integrated biological staging system in HD are important innovations applicable to clinical trial design that enhance the drug development process. Although a future in HD with DMTs remains a hope for those living with HD, care partners and care providers, the therapeutic landscape is promising, with various drug development programs underway following a targeted approach supported by disease-specific biomarkers and staging frameworks.


Assuntos
Doença de Huntington , Humanos , Doença de Huntington/terapia , Doença de Huntington/genética , Doença de Huntington/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Proteína Huntingtina/genética , Animais , Desenvolvimento de Medicamentos
15.
Antiviral Res ; 228: 105946, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38925369

RESUMO

SARS-CoV-2 is a betacoronavirus that causes COVID-19, a global pandemic that has resulted in many infections, deaths, and socio-economic challenges. The virus has a large positive-sense, single-stranded RNA genome of ∼30 kb, which produces subgenomic RNAs (sgRNAs) through discontinuous transcription. The most abundant sgRNA is sgRNA N, which encodes the nucleocapsid (N) protein. In this study, we probed the secondary structure of sgRNA N and a shorter model without a 3' UTR in vitro, using the SHAPE (selective 2'-hydroxyl acylation analyzed by a primer extension) method and chemical mapping with dimethyl sulfate and 1-cyclohexyl-(2-morpholinoethyl) carbodiimide metho-p-toluene sulfonate. We revealed the secondary structure of sgRNA N and its shorter variant for the first time and compared them with the genomic RNA N structure. Based on the structural information, we designed gapmers, siRNAs and antisense oligonucleotides (ASOs) to target the N protein coding region of sgRNA N. We also generated eukaryotic expression vectors containing the complete sequence of sgRNA N and used them to screen for new SARS-CoV-2 gene N expression inhibitors. Our study provides novel insights into the structure and function of sgRNA N and potential therapeutic tools against SARS-CoV-2.


Assuntos
Conformação de Ácido Nucleico , RNA Viral , SARS-CoV-2 , Replicação Viral , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/genética , Replicação Viral/efeitos dos fármacos , RNA Viral/genética , Humanos , Antivirais/farmacologia , Antivirais/química , Proteínas do Nucleocapsídeo de Coronavírus/genética , Proteínas do Nucleocapsídeo de Coronavírus/antagonistas & inibidores , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Proteínas do Nucleocapsídeo de Coronavírus/química , Ésteres do Ácido Sulfúrico/farmacologia , Ésteres do Ácido Sulfúrico/química , COVID-19/virologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/química , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/química , Genoma Viral , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/química
16.
Neuron ; 112(11): 1730-1732, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38843779

RESUMO

In a recent issue of Nature, Chen and colleagues1 reveal the potential for antisense oligonucleotides (ASOs) to rescue the neuropathological mechanisms underlying Timothy syndrome (TS) using three-dimensional neuronal models. Combining in vitro and in vivo approaches, the authors present a strategy to translate disease biology findings into potential therapeutics.


Assuntos
Transtorno Autístico , Síndrome do QT Longo , Neurônios , Sindactilia , Humanos , Transtorno Autístico/genética , Transtorno Autístico/patologia , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Sindactilia/genética , Oligonucleotídeos Antissenso/farmacologia , Animais
17.
Inflamm Res ; 73(7): 1123-1135, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38698180

RESUMO

OBJECTIVE: Immunotherapy targeting programmed cell death 1 (PDCD1 or PD-1) and its ligands has shown remarkable promise and the regulation mechanism of PD-1 expression has received arising attention in recent years. PDCD1 exon 3 encodes the transmembrane domain and the deletion of exon 3 produces a soluble protein isoform of PD-1 (sPD-1), which can enhance immune response by competing with full-length PD-1 protein (flPD-1 or surface PD-1) on T cell surface. However, the mechanism of PDCD1 exon 3 skipping is unclear. METHODS: The online SpliceAid program and minigene expression system were used to analyze potential splicing factors involved in the splicing event of PDCD1 exon 3. The potential binding motifs of heterogeneous nuclear ribonucleoprotein K (HNRNPK) on exon 3 predicted by SpliceAid were mutated by site-directed mutagenesis technology, which were further verified by pulldown assay. Antisense oligonucleotides (ASOs) targeting the exonic splicing silencer (ESS) on PDCD1 exon 3 were synthesized and screened to suppress the skipping of exon 3. The alternative splicing of PDCD1 exon 3 was analyzed by semiquantitative reverse transcription PCR. Western blot and flow cytometry were performed to detect the surface PD-1 expression in T cells. RESULTS: HNRNPK was screened as a key splicing factor that promoted PDCD1 exon 3 skipping, causing a decrease in flPD-1 expression on T cell membrane and an increase in sPD-1 expression. Mechanically, a key ESS has been identified on exon 3 and can be bound by HNRNPK protein to promote exon 3 skipping. Blocking the interaction between ESS and HNRNPK with an ASO significantly reduced exon 3 skipping. Importantly, HNRNPK can promote exon 3 skipping of mouse Pdcd1 gene as well. CONCLUSIONS: Our study revealed a novel evolutionarily conserved regulatory mechanism of PD-1 expression. The splicing factor HNRNPK markedly promoted PDCD1 exon 3 skipping by binding to the ESS on PDCD1 exon 3, resulting in decreased expression of flPD-1 and increased expression of sPD-1 in T cells.


Assuntos
Éxons , Ribonucleoproteínas Nucleares Heterogêneas Grupo K , Receptor de Morte Celular Programada 1 , Linfócitos T , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Animais , Humanos , Linfócitos T/metabolismo , Linfócitos T/imunologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Processamento Alternativo , Camundongos Endogâmicos C57BL , Camundongos , Oligonucleotídeos Antissenso/genética
18.
Arch Pharm Res ; 47(6): 571-595, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38764004

RESUMO

Huntington's disease (HD) is a paradigm of a genetic neurodegenerative disorder characterized by the expansion of CAG repeats in the HTT gene. This extensive review investigates the molecular complexities of HD by highlighting the pathogenic mechanisms initiated by the mutant huntingtin protein. Adverse outcomes of HD include mitochondrial dysfunction, compromised protein clearance, and disruption of intracellular signaling, consequently contributing to the gradual deterioration of neurons. Numerous therapeutic strategies, particularly precision medicine, are currently used for HD management. Antisense oligonucleotides, such as Tominersen, play a leading role in targeting and modulating the expression of mutant huntingtin. Despite the promise of these therapies, challenges persist, particularly in improving delivery systems and the necessity for long-term safety assessments. Considering the future landscape, the review delineates promising directions for HD research and treatment. Innovations such as Clustered regularly interspaced short palindromic repeats associated system therapies (CRISPR)-based genome editing and emerging neuroprotective approaches present unprecedented opportunities for intervention. Collaborative interdisciplinary endeavors and a more insightful understanding of HD pathogenesis are on the verge of reshaping the therapeutic landscape. As we navigate the intricate landscape of HD, this review serves as a guide for unraveling the intricacies of this disease and progressing toward transformative treatments.


Assuntos
Proteína Huntingtina , Doença de Huntington , Doença de Huntington/terapia , Doença de Huntington/genética , Humanos , Animais , Proteína Huntingtina/genética , Proteína Huntingtina/antagonistas & inibidores , Proteína Huntingtina/metabolismo , Oligonucleotídeos Antissenso/uso terapêutico , Terapia Genética/métodos , Edição de Genes/métodos , Fármacos Neuroprotetores/uso terapêutico
19.
BMC Oral Health ; 24(1): 552, 2024 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-38735923

RESUMO

Patients who suffer from myofascial orofacial pain could affect their quality of life deeply. The pathogenesis of pain is still unclear. Our objective was to assess Whether Voltage-gated calcium channel α2δ-1(Cavα2δ-1) is related to myofascial orofacial pain. Rats were divided into the masseter tendon ligation group and the sham group. Compared with the sham group, the mechanical pain threshold of the masseter tendon ligation group was reduced on the 4th, 7th, 10th and 14th day after operation(P < 0.05). On the 14th day after operation, Cavα2δ-1 mRNA expression levels in trigeminal ganglion (TG) and the trigeminal spinal subnucleus caudalis and C1-C2 spinal cervical dorsal horn (Vc/C2) of the masseter tendon ligation group were increased (PTG=0.021, PVc/C2=0.012). Rats were divided into three groups. On the 4th day after ligating the superficial tendon of the left masseter muscle of the rats, 10 ul Cavα2δ-1 antisense oligonucleotide, 10 ul Cavα2δ-1 mismatched oligonucleotides and 10 ul normal saline was separately injected into the left masseter muscle of rats in Cavα2δ-1 antisense oligonucleotide group, Cavα2δ-1 mismatched oligonucleotides group and normal saline control group twice a day for 4 days. The mechanical pain threshold of the Cavα2δ-1 antisense oligonucleotides group was higher than Cavα2δ-1 mismatched oligonucleotides group on the 7th and 10th day after operation (P < 0.01). After PC12 cells were treated with lipopolysaccharide, Cavα2δ-1 mRNA expression level increased (P < 0.001). Cavα2δ-1 may be involved in the occurrence and development in myofascial orofacial pain.


Assuntos
Canais de Cálcio Tipo L , Dor Facial , Músculo Masseter , Gânglio Trigeminal , Animais , Masculino , Ratos , Canais de Cálcio/metabolismo , Dor Facial/metabolismo , Músculo Masseter/metabolismo , Síndromes da Dor Miofascial , Oligonucleotídeos Antissenso/farmacologia , Limiar da Dor , Ratos Sprague-Dawley , RNA Mensageiro/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Gânglio Trigeminal/metabolismo
20.
JCI Insight ; 9(12)2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38743496

RESUMO

Diabetes increases the risk of both cardiovascular disease and kidney disease. Notably, most of the excess cardiovascular risk in people with diabetes is in those with kidney disease. Apolipoprotein C3 (APOC3) is a key regulator of plasma triglycerides, and it has recently been suggested to play a role in both type 1 diabetes-accelerated atherosclerosis and kidney disease progression. To investigate if APOC3 plays a role in kidney disease in people with type 2 diabetes, we analyzed plasma levels of APOC3 from the Veterans Affairs Diabetes Trial. Elevated baseline APOC3 levels predicted a greater loss of renal function. To mechanistically test if APOC3 plays a role in diabetic kidney disease and associated atherosclerosis, we treated black and tan, brachyury, WT and leptin-deficient (OB; diabetic) mice, a model of type 2 diabetes, with an antisense oligonucleotide (ASO) to APOC3 or a control ASO, all in the setting of human-like dyslipidemia. Silencing APOC3 prevented diabetes-augmented albuminuria, renal glomerular hypertrophy, monocyte recruitment, and macrophage accumulation, partly driven by reduced ICAM1 expression. Furthermore, reduced levels of APOC3 suppressed atherosclerosis associated with diabetes. This suggests that targeting APOC3 might benefit both diabetes-accelerated atherosclerosis and kidney disease.


Assuntos
Apolipoproteína C-III , Aterosclerose , Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Apolipoproteína C-III/genética , Apolipoproteína C-III/sangue , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Animais , Aterosclerose/metabolismo , Aterosclerose/etiologia , Camundongos , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Oligonucleotídeos Antissenso/farmacologia , Modelos Animais de Doenças
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA