Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 202
Filtrar
1.
Curr Med Sci ; 43(2): 261-267, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36932303

RESUMO

OBJECTIVE: Charcot-Marie-Tooth disease (CMT) severely affects patient activity, and may cause disability. However, no clinical treatment is available to reverse the disease course. The combination of CRISPR/Cas9 and iPSCs may have therapeutic potential against nervous diseases, such as CMT. METHODS: In the present study, the skin fibroblasts of CMT type 2D (CMT2D) patients with the c.880G>A heterozygous nucleotide mutation in the GARS gene were reprogrammed into iPSCs using three plasmids (pCXLE-hSK, pCXLE-hUL and pCXLE-hOCT3/4-shp5-F). Then, CRISPR/Cas9 technology was used to repair the mutated gene sites at the iPSC level. RESULTS: An iPSC line derived from the GARS (G294R) family with fibular atrophy was successfully induced, and the mutated gene loci were repaired at the iPSC level using CRISPR/Cas9 technology. These findings lay the foundation for future research on drug screening and cell therapy. CONCLUSION: iPSCs can differentiate into different cell types, and originate from autologous cells. Therefore, they are promising for the development of autologous cell therapies for degenerative diseases. The combination of CRISPR/Cas9 and iPSCs may open a new avenue for the treatment of nervous diseases, such as CMT.


Assuntos
Doença de Charcot-Marie-Tooth , Células-Tronco Pluripotentes Induzidas , Reparo Gênico Alvo-Dirigido , Humanos , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/terapia , Doença de Charcot-Marie-Tooth/metabolismo , Sistemas CRISPR-Cas/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação , Reparo Gênico Alvo-Dirigido/métodos
2.
RNA Biol ; 19(1): 176-190, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35067193

RESUMO

RNA-based therapeutics have entered the mainstream with seemingly limitless possibilities to treat all categories of neurological disease. Here, common RNA-based drug modalities such as antisense oligonucleotides, small interfering RNAs, RNA aptamers, RNA-based vaccines and mRNA drugs are reviewed highlighting their current and potential applications. Rapid progress has been made across rare genetic diseases and neurodegenerative disorders, but safe and effective delivery to the brain remains a significant challenge for many applications. The advent of individualized RNA-based therapies for ultra-rare diseases is discussed against the backdrop of the emergence of this field into more common conditions such as Alzheimer's disease and ischaemic stroke. There remains significant untapped potential in the use of RNA-based therapeutics for behavioural disorders and tumours of the central nervous system; coupled with the accelerated development expected over the next decade, the true potential of RNA-based therapeutics to transform the therapeutic landscape in neurology remains to be uncovered.


Assuntos
Terapia Genética , Doenças do Sistema Nervoso/terapia , RNA/genética , RNA/uso terapêutico , Animais , Aptâmeros de Nucleotídeos , Gerenciamento Clínico , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Humanos , Doenças do Sistema Nervoso/etiologia , RNA/química , Interferência de RNA , RNA Interferente Pequeno , Terapêutica com RNAi , Reparo Gênico Alvo-Dirigido
3.
Cancer Res Treat ; 54(1): 30-39, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34015890

RESUMO

PURPOSE: K-MASTER project is a Korean national precision medicine platform that screened actionable mutations by analyzing next-generation sequencing (NGS) of solid tumor patients. We compared gene analyses between NGS panel from the K-MASTER project and orthogonal methods. MATERIALS AND METHODS: Colorectal, breast, non-small cell lung, and gastric cancer patients were included. We compared NGS results from K-MASTER projects with those of non-NGS orthogonal methods (KRAS, NRAS, and BRAF mutations in colorectal cancer [CRC]; epidermal growth factor receptor [EGFR], anaplastic lymphoma kinase [ALK] fusion, and reactive oxygen species 1 [ROS1] fusion in non-small cell lung cancer [NSCLC], and Erb-B2 receptor tyrosine kinase 2 (ERBB2) positivity in breast and gastric cancers). RESULTS: In the CRC cohort (n=225), the sensitivity and specificity of NGS were 87.4% and 79.3% (KRAS); 88.9% and 98.9% (NRAS); and 77.8% and 100.0% (BRAF), respectively. In the NSCLC cohort (n=109), the sensitivity and specificity of NGS for EGFR were 86.2% and 97.5%, respectively. The concordance rate for ALK fusion was 100%, but ROS1 fusion was positive in only one of three cases that were positive in orthogonal tests. In the breast cancer cohort (n=260), ERBB2 amplification was detected in 45 by NGS. Compared with orthogonal methods that integrated immunohistochemistry and in situ hybridization, sensitivity and specificity were 53.7% and 99.4%, respectively. In the gastric cancer cohort (n=64), ERBB2 amplification was detected in six by NGS. Compared with orthogonal methods, sensitivity and specificity were 62.5% and 98.2%, respectively. CONCLUSION: The results of the K-MASTER NGS panel and orthogonal methods showed a different degree of agreement for each genetic alteration, but generally showed a high agreement rate.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Medicina de Precisão/normas , Reparo Gênico Alvo-Dirigido/normas , Neoplasias da Mama/genética , Neoplasias Colorretais/genética , Feminino , Humanos , Neoplasias Pulmonares/genética , Masculino , República da Coreia , Sensibilidade e Especificidade , Carcinoma de Pequenas Células do Pulmão/genética , Neoplasias Gástricas/genética
4.
Cancer Res Treat ; 54(1): 40-53, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34044476

RESUMO

PURPOSE: We investigated the feasibility of using an anatomically localized, target-enriched liquid biopsy (TLB) in mouse models of lung cancer. MATERIALS AND METHODS: After irradiating xenograft mouse with human lung cancer cell lines, H1299 (NRAS proto-oncogene, GTPase [NRAS] Q61K) and HCC827 (epidermal growth factor receptor [EGFR] E746-750del), circulating (cell-free) tumor DNA (ctDNA) levels were monitored with quantitative polymerase chain reaction on human long interspersed nuclear element-1 and cell line-specific mutations. We checked dose-dependency at 6, 12, or 18 Gy to each tumor-bearing mouse leg using 6-MV photon beams. We also analyzed ctDNA of lung cancer patients by LiquidSCAN, a targeted deep sequencing to validated the clinical performances of TLB method. RESULTS: Irradiation could enhance the detection sensitivity of NRAS Q61K in the plasma sample of H1299-xenograft mouse to 4.5- fold. While cell-free DNA (cfDNA) level was not changed at 6 Gy, ctDNA level was increased upon irradiation. Using double-xenograft mouse with H1299 and HCC827, ctDNA polymerase chain reaction analysis with local irradiation in each region could specify mutation type matched to transplanted cell types, proposing an anatomically localized, TLB. Furthermore, when we performed targeted deep sequencing of cfDNA to monitor ctDNA level in 11 patients with lung cancer who underwent radiotherapy, the average ctDNA level was increased within a week after the start of radiotherapy. CONCLUSION: TLB using irradiation could temporarily amplify ctDNA release in xenograft mouse and lung cancer patients, which enables us to develop theragnostic method for cancer patients with accurate ctDNA detection.


Assuntos
DNA Tumoral Circulante/sangue , Biópsia Líquida/métodos , Neoplasias Pulmonares/genética , Reparo Gênico Alvo-Dirigido/métodos , Animais , Biomarcadores Tumorais/genética , Estudos de Viabilidade , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Microambiente Tumoral
5.
Cancer Treat Rev ; 101: 102310, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34757307

RESUMO

PURPOSE: Current guidelines recommend somatic genomic sequencing for patients with advanced pancreatic cancer to identify targetable alterations amenable to targeted therapy. The benefit of somatic genomic sequencing in pancreatic cancer remains unclear. This study aims to assess the evidence supporting genomic sequencing to inform treatment selection for patients with advanced pancreatic cancer. METHODS: A systematic review identified prospective studies of exocrine pancreatic cancer patients published before August 2020 which conducted genomic sequencing to inform treatment selection. Outcomes of interest included the proportion of patients with targetable alterations, the proportion that received targeted treatments, and the impact of targeted treatments on overall survival. Meta-analysis for proportions and hazard ratios was performed using Dersimonian and Laird random effect models. RESULTS: 19 studies (representing 2048 pancreatic cancer patients) were included. Sequencing methodologies, definitions of targetable alterations, and approaches treatment selection varied across studies and were incompletely reported. 590 of 1382 sequenced patients harboured a targetable alteration (random effects meta-analysis estimate of the proportion 0.46, 95% confidence interval 0.32-0.61). The proportion of patients with targetable alterations was highly heterogenous between studies (I2 93%, P < 0.001). 91 of 1390 patients received a matched therapy based on their targetable alterations (random effects meta-analysis estimate of the proportion 0.12, 95% CI 0.06-0.23). One observational study reported an overall survival benefit of matched therapy. CONCLUSIONS: Genomic sequencing frequently identifies targetable alterations in pancreatic cancers. Further research is required to standardize the definitions of targetable alterations, the approach to treatment matching, and quantify the benefit of targeted therapy.


Assuntos
Terapia Genética/métodos , Terapia de Alvo Molecular/métodos , Neoplasias Pancreáticas , Análise de Sequência de DNA/métodos , Reparo Gênico Alvo-Dirigido/métodos , Humanos , Estadiamento de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Seleção de Pacientes
6.
Biomed Pharmacother ; 143: 112075, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34488082

RESUMO

Gene therapy may constitute a promising alternative to conventional pharmacological tools and surgeries for epilepsy. For primary epilepsy, a single variant leading to a significant effect is relatively rare, while other forms are considered complex in inheritances with multiple susceptible mutations and impacts from the environment. Gene therapy in preclinical models of epilepsy has attempted to perform antiepileptogenic, anticonvulsant, or disease-modifying effects during epileptogenesis or after establishing the disease. Creating gene vectors tailored for different situations is the key to expanding gene therapy, and choosing the appropriate therapeutic target remains another fundamental problem. A variety of treatment strategies, from overexpressing inhibitory neuropeptides to modulating the expression of neurotransmitters or ion channels, have been tested in animal models. Additionally, emerging new approaches of optogenetics and chemogenetics, as well as genome-editing tools will further boost the prosperity of gene therapy. This review summarizes the experience obtained to date and discusses the challenges and opportunities in clinical translations.


Assuntos
Ondas Encefálicas/genética , Encéfalo/fisiopatologia , Epilepsia/terapia , Terapia Genética , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Edição de Genes , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Reparo Gênico Alvo-Dirigido , Vírus/genética
7.
JCI Insight ; 6(15)2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34369389

RESUMO

Angelman syndrome (AS) is a severe neurodevelopmental disorder for which only symptomatic treatment with limited benefits is available. AS is caused by mutations affecting the maternally inherited ubiquitin protein ligase E3A (UBE3A) gene. Previous studies showed that the silenced paternal Ube3a gene can be activated by targeting the antisense Ube3a-ATS transcript. We investigated antisense oligonucleotide-induced (ASO-induced) Ube3a-ATS degradation and its ability to induce UBE3A reinstatement and rescue of AS phenotypes in an established Ube3a mouse model. We found that a single intracerebroventricular injection of ASOs at postnatal day 1 (P1) or P21 in AS mice resulted in potent and specific UBE3A reinstatement in the brain, with levels up to 74% of WT levels in the cortex and a full rescue of sensitivity to audiogenic seizures. AS mice treated with ASO at P1 also showed rescue of established AS phenotypes, such as open field and forced swim test behaviors, and significant improvement on the reversed rotarod. Hippocampal plasticity of treated AS mice was comparable to WT but not significantly different from PBS-treated AS mice. No rescue was observed for the marble burying and nest building phenotypes. Our findings highlight the promise of ASO-mediated reactivation of UBE3A as a disease-modifying treatment for AS.


Assuntos
Síndrome de Angelman , Oligonucleotídeos Antissenso/uso terapêutico , Ubiquitina-Proteína Ligases/metabolismo , Síndrome de Angelman/genética , Síndrome de Angelman/metabolismo , Animais , Variação Biológica da População , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Inativação Gênica , Camundongos , Reparo Gênico Alvo-Dirigido/métodos , Resultado do Tratamento
8.
Stem Cell Res Ther ; 12(1): 485, 2021 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-34454631

RESUMO

BACKGROUND: Achondroplasia (ACH) is the most common genetic form of dwarfism and belongs to dominant monogenic disorder caused by a gain-of-function point mutation in the transmembrane region of FGFR3. There are no effective treatments for ACH. Stem cells and gene-editing technology provide us with effective methods and ideas for ACH research and treatment. METHODS: We generated non-integrated iPSCs from an ACH girl's skin and an ACH boy's urine by Sendai virus. The mutation of ACH iPSCs was precisely corrected by CRISPR-Cas9. RESULTS: Chondrogenic differentiation ability of ACH iPSCs was confined compared with that of healthy iPSCs. Chondrogenic differentiation ability of corrected ACH iPSCs could be restored. These corrected iPSCs displayed pluripotency, maintained normal karyotype, and demonstrated none of off-target indels. CONCLUSIONS: This study may provide an important theoretical and experimental basis for the ACH research and treatment.


Assuntos
Acondroplasia , Células-Tronco Pluripotentes Induzidas , Acondroplasia/genética , Acondroplasia/terapia , Diferenciação Celular , Feminino , Edição de Genes , Humanos , Masculino , Mutação , Reparo Gênico Alvo-Dirigido
9.
Cell Mol Gastroenterol Hepatol ; 12(4): 1433-1455, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34217895

RESUMO

BACKGROUND & AIMS: Rapid gastric epithelial progenitor cell (EPC) proliferation and inflammatory response inhibition play key roles in promoting the repair of gastric mucosal damage. However, specific targets inducing these effects are unknown. In this study, we explored the effects of a potential target, Ankyrin repeat domain 22 (ANKRD22). METHODS: An acute gastric mucosal injury model was established with Ankrd22-/- and Ankrd22+/+ mice by intragastric administration of acidified ethanol. Organoid culture and flow cytometry were performed to evaluate the effects of ANKRD22 on leucine-rich repeat-containing G-protein-coupled receptor 5-positive (Lgr5+) gastric EPC proliferation. The mechanisms by which ANKRD22 affects gastric EPC proliferation and inflammatory responses were explored by mitochondrial Ca2+ influx and immunoblotting. Candidate ANKRD22 inhibitors then were screened virtually and validated in vitro and in vivo. RESULTS: After acute gastric mucosal injury, the number of Lgr5+ gastric EPCs was increased significantly in Ankrd22-/- mice compared with that in Ankrd22+/+ mice. Moreover, Ankrd22 knockout attenuated inflammatory cell infiltration into damaged gastric tissues. ANKRD22 deletion also reduced mitochondrial Ca2+ influx and cytoplasmic nuclear factor of activated T cells in gastric epithelial cells and macrophages, which further induced Lgr5+ gastric EPC proliferation and decreased macrophage release of tumor necrosis factor-α and interleukin 1α. In addition, a small molecule, AV023, was found to show similar effects to those produced by ANKRD22 deletion in vitro. Intraperitoneal injection of AV023 into the mouse model promoted the repair of gastric mucosal damage, with increased proliferation of Lgr5+ gastric EPCs and visible relief of inflammation. CONCLUSIONS: ANKRD22 inhibition is a potential target-based therapeutic approach for promoting the repair of gastric mucosal damage.


Assuntos
Biomarcadores , Mucosa Gástrica/metabolismo , Proteínas de Membrana/genética , Receptores Acoplados a Proteínas G/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Desenvolvimento de Medicamentos , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Regulação da Expressão Gênica , Imuno-Histoquímica , Imunofenotipagem , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Modelos Moleculares , Receptores Acoplados a Proteínas G/metabolismo , Gastropatias/tratamento farmacológico , Gastropatias/etiologia , Gastropatias/metabolismo , Gastropatias/patologia , Relação Estrutura-Atividade , Reparo Gênico Alvo-Dirigido , Via de Sinalização Wnt
10.
Int J Biol Sci ; 17(8): 1940-1952, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34131397

RESUMO

There is a continued need for investigating the roles of microRNAs (miRNAs) and their targets on the progression of gastric cancer (GC), especially metastasis. Here, we performed an integrated study to identify dysregulated miRNAs critical for GC development and progression. miR-135b was determined as a promising biomarker for GC. The expression level of miR-135b was increased among GC cell lines, patient tumor tissues, serum samples, and correlation with aggravation of the GC patients. The in vitro functional assays demonstrated overexpression of miR-135b promoted cell proliferation, migration and invasion in GC, while miR-135b inhibition led to the opposite results. CAMK2D was found to be the direct target of miR-135b, serving as a tumor suppressor in GC cells. Based on our and public datasets, we confirmed the attenuation of CAMK2D expression in GC tissues. And, the expression levels of miR-135b and CAMK2D were closely associated with prognosis of GC patients. Ectopic expression of miR-135b resulted in the down-regulation of CAMK2D. Additionally, CAMK2D was a prerequisite for miR-135b to promote GC cells proliferation and migration by regulating the EMT process, which was confirmed by the in vivo experiments. Importantly, in vivo injection of miR-135b antagomir significantly repressed the tumor growth and metastasis of xenograft models, which suggested that the miR-135b antagomir were promising for clinical applications. Taken together, these results indicate that miR-135b/CAMK2D axis drives GC progression by EMT process remodeling, suggesting that miR-135b may be utilized as a new therapeutic target and prognostic marker for GC patients.


Assuntos
Antagomirs/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Transição Epitelial-Mesenquimal/genética , MicroRNAs , Neoplasias Gástricas , Reparo Gênico Alvo-Dirigido/métodos , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Estudos de Associação Genética , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Prognóstico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia
11.
Med Sci (Paris) ; 37(4): 413-416, 2021 Apr.
Artigo em Francês | MEDLINE | ID: mdl-33908864

RESUMO

Hutchinson-Gilford Progeria (acute premature aging) is caused by a de novo point mutation in the lamin A gene. Recently, this mutation has been accurately corrected by base editing in patient cell lines and in a mouse model, resulting in nearly complete reversal to a normal phenotype. This success opens the perspective for clinical applications in Progeria and other diseases.


Assuntos
Lamina Tipo A/genética , Mutação Puntual , Progéria/terapia , Reparo Gênico Alvo-Dirigido , Animais , Modelos Animais de Doenças , Imunofluorescência , Edição de Genes/métodos , Humanos , Lamina Tipo A/metabolismo , Camundongos , Progéria/genética
12.
Nat Commun ; 12(1): 472, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33473139

RESUMO

Targeted DNA correction of disease-causing mutations in hematopoietic stem and progenitor cells (HSPCs) may enable the treatment of genetic diseases of the blood and immune system. It is now possible to correct mutations at high frequencies in HSPCs by combining CRISPR/Cas9 with homologous DNA donors. Because of the precision of gene correction, these approaches preclude clonal tracking of gene-targeted HSPCs. Here, we describe Tracking Recombination Alleles in Clonal Engraftment using sequencing (TRACE-Seq), a methodology that utilizes barcoded AAV6 donor template libraries, carrying in-frame silent mutations or semi-randomized nucleotides outside the coding region, to track the in vivo lineage contribution of gene-targeted HSPC clones. By targeting the HBB gene with an AAV6 donor template library consisting of ~20,000 possible unique exon 1 in-frame silent mutations, we track the hematopoietic reconstitution of HBB targeted myeloid-skewed, lymphoid-skewed, and balanced multi-lineage repopulating human HSPC clones in mice. We anticipate this methodology could potentially be used for HSPC clonal tracking of Cas9 RNP and AAV6-mediated gene targeting outcomes in translational and basic research settings.


Assuntos
Alelos , Células Clonais , Marcação de Genes/métodos , Células-Tronco Hematopoéticas , Recombinação Genética , Animais , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Feminino , Edição de Genes/métodos , Terapia Genética/métodos , Humanos , Camundongos , Mutação , Reparo Gênico Alvo-Dirigido/métodos
13.
Int J Mol Sci ; 21(23)2020 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-33266216

RESUMO

Using nanoparticles to carry and delivery anticancer drugs holds much promise in cancer therapy, but nanoparticles per se are lacking specificity. Active targeting, that is, using specific ligands to functionalize nanoparticles, is attracting much attention in recent years. Aptamers, with their several favorable features like high specificity and affinity, small size, very low immunogenicity, relatively low cost for production, and easiness to store, are one of the best candidates for the specific ligands of nanoparticle functionalization. This review discusses the benefits and challenges of using aptamers to functionalize nanoparticles for active targeting and especially presents nearly all of the published works that address the topic of using aptamers to functionalize nanoparticles for targeted drug delivery and cancer therapy.


Assuntos
Antineoplásicos/administração & dosagem , Aptâmeros de Nucleotídeos/administração & dosagem , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Nanopartículas , Animais , Antineoplásicos/química , Aptâmeros de Nucleotídeos/química , Portadores de Fármacos/química , Avaliação Pré-Clínica de Medicamentos , Humanos , Ligantes , Terapia de Alvo Molecular , Nanopartículas/química , Neoplasias/etiologia , Neoplasias/patologia , Neoplasias/terapia , Reparo Gênico Alvo-Dirigido , Nanomedicina Teranóstica
14.
Nat Commun ; 11(1): 5352, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-33097693

RESUMO

Prime editing is a recent genome editing technology using fusion proteins of Cas9-nickase and reverse transcriptase, that holds promise to correct the vast majority of genetic defects. Here, we develop prime editing for primary adult stem cells grown in organoid culture models. First, we generate precise in-frame deletions in the gene encoding ß-catenin (CTNNB1) that result in proliferation independent of Wnt-stimuli, mimicking a mechanism of the development of liver cancer. Moreover, prime editing functionally recovers disease-causing mutations in intestinal organoids from patients with DGAT1-deficiency and liver organoids from a patient with Wilson disease (ATP7B). Prime editing is as efficient in 3D grown organoids as in 2D grown cell lines and offers greater precision than Cas9-mediated homology directed repair (HDR). Base editing remains more reliable than prime editing but is restricted to a subgroup of pathogenic mutations. Whole-genome sequencing of four prime-edited clonal organoid lines reveals absence of genome-wide off-target effects underscoring therapeutic potential of this versatile and precise gene editing strategy.


Assuntos
Edição de Genes/métodos , Organoides/metabolismo , beta Catenina/genética , Sistemas CRISPR-Cas , Linhagem Celular , Proliferação de Células , ATPases Transportadoras de Cobre/genética , Desoxirribonuclease I/metabolismo , Diacilglicerol O-Aciltransferase/genética , Células HEK293 , Degeneração Hepatolenticular/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação , Reparo de DNA por Recombinação , Células-Tronco , Reparo Gênico Alvo-Dirigido/métodos
15.
Int J Gynecol Cancer ; 30(10): 1608-1618, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32928926

RESUMO

The clinical development of poly-(ADP)-ribose polymerase inhibitors (PARPi) began with the treatment of ovarian cancer patients harboring BRCA1/2 mutations and continues to be expanded to other gynecological cancers. Furthermore, The Cancer Genome Atlas (TCGA) analysis of endometrial and cervical cancers offered rationale that PARPi may be an option for treatment based on the molecular profiles of these cancer types. This review summarizes the current indications of PARPi, such as its role in the treatment and maintenance of recurrent ovarian cancer and for first-line maintenance therapy in advanced ovarian cancer. We also outline new concepts for PARPi therapy in other gynecological cancers such as endometrial and cervical cancers based on recent clinical data. Finally, we present potential future directions to continue exploring the world of PARPi resistance and combining PARPi with other therapies.


Assuntos
Neoplasias do Endométrio/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Neoplasias do Colo do Útero/tratamento farmacológico , Dano ao DNA/efeitos dos fármacos , Feminino , Humanos , Recidiva Local de Neoplasia/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Intervalo Livre de Progressão , Reparo Gênico Alvo-Dirigido/métodos
16.
J Autoimmun ; 114: 102529, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32782117

RESUMO

The identification of RNA interference (RNAi) has caused a growing interest in harnessing its potential in the treatment of different diseases. Modulation of dysregulated genes through targeting by RNAi represents a potential approach with which to alter the biological pathways at a post-transcriptional level, especially as it pertains to autoimmunity and malignancy. Short hairpin RNAs (shRNA), short interfering RNAs (siRNA), and microRNAs (miRNA) are mainly involved as effector mechanisms in the targeting of RNAi biological pathways. The manipulation and delivery of these molecules in an efficient way promotes the specificity and stability of RNAi-based systems, while minimizing the unwanted adverse reactions by the immune system and reducing cytotoxicity and off-target effects. Advances made to date in identifying the etiopathogenesis of autoimmune diseases has prompted the utilization of RNAi-based systems in vitro and in vivo. Future investigations aimed at deciphering the molecular basis of RNAi and optimizing the delivery of RNAi-based targeting systems will hopefully promote the applicability of such regulatory mechanisms and, ultimately, transfer the acquired knowledge from bench-to-bedside to ameliorate human diseases. In this review, we seek to clarify the potential of RNAi, with a focus on siRNAs, in designing therapeutics for potential treatment of human autoimmune disorders.


Assuntos
Doenças Autoimunes/etiologia , Doenças Autoimunes/terapia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Animais , Regulação da Expressão Gênica , Terapia Genética/métodos , Humanos , MicroRNAs/genética , Reparo Gênico Alvo-Dirigido , Pesquisa Translacional Biomédica
17.
Nat Biotechnol ; 38(9): 1037-1043, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32632303

RESUMO

Base editors, including adenine base editors (ABEs)1 and cytosine base editors (CBEs)2,3, are widely used to induce point mutations. However, determining whether a specific nucleotide in its genomic context can be edited requires time-consuming experiments. Furthermore, when the editable window contains multiple target nucleotides, various genotypic products can be generated. To develop computational tools to predict base-editing efficiency and outcome product frequencies, we first evaluated the efficiencies of an ABE and a CBE and the outcome product frequencies at 13,504 and 14,157 target sequences, respectively, in human cells. We found that there were only modest asymmetric correlations between the activities of the base editors and Cas9 at the same targets. Using deep-learning-based computational modeling, we built tools to predict the efficiencies and outcome frequencies of ABE- and CBE-directed editing at any target sequence, with Pearson correlations ranging from 0.50 to 0.95. These tools and results will facilitate modeling and therapeutic correction of genetic diseases by base editing.


Assuntos
Adenina , Citosina , Edição de Genes/métodos , Reparo Gênico Alvo-Dirigido/métodos , Aminoidrolases/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas , Citosina Desaminase/metabolismo , Engenharia Genética , Genoma Humano/genética , Células HEK293 , Humanos , Mutação Puntual , RNA Guia de Cinetoplastídeos/genética
18.
Int J Mol Sci ; 21(11)2020 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-32471232

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease caused by the death of motor neurons in the spinal cord and brainstem. ALS has a diverse genetic origin; at least 20 genes have been shown to be related to ALS. Most familial and sporadic cases of ALS are caused by variants of the SOD1, C9orf72, FUS, and TARDBP genes. Genome editing using clustered regularly interspaced short palindromic repeats/CRISPR-associated system 9 (CRISPR/Cas9) can provide insights into the underlying genetics and pathophysiology of ALS. By correcting common mutations associated with ALS in animal models and patient-derived induced pluripotent stem cells (iPSCs), CRISPR/Cas9 has been used to verify the effects of ALS-associated mutations and observe phenotype differences between patient-derived and gene-corrected iPSCs. This technology has also been used to create mutations to investigate the pathophysiology of ALS. Here, we review recent studies that have used CRISPR/Cas9 to understand the genetic underpinnings of ALS.


Assuntos
Esclerose Lateral Amiotrófica/terapia , Sistemas CRISPR-Cas , Reparo Gênico Alvo-Dirigido/métodos , Esclerose Lateral Amiotrófica/genética , Animais , Proteína C9orf72/genética , Proteínas de Ligação a DNA/genética , Humanos , Proteína FUS de Ligação a RNA/genética , Superóxido Dismutase-1/genética
19.
Int J Mol Sci ; 21(8)2020 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-32340368

RESUMO

RNA-binding proteins (RBPs) are involved in regulating all aspects of RNA metabolism, including processing, transport, translation, and degradation. Dysregulation of RNA metabolism is linked to a plethora of diseases, such as cancer, neurodegenerative diseases, and neuromuscular disorders. Recent years have seen a dramatic shift in the knowledge base, with RNA increasingly being recognised as an attractive target for precision medicine therapies. In this article, we are going to review current RNA-targeted therapies. Furthermore, we will scrutinise a range of drug discoveries targeting protein-RNA interactions. In particular, we will focus on the interplay between Lin28 and let-7, splicing regulatory proteins and survival motor neuron (SMN) pre-mRNA, as well as HuR, Musashi, proteins and their RNA targets. We will highlight the mechanisms RBPs utilise to modulate RNA metabolism and discuss current high-throughput screening strategies. This review provides evidence that we are entering a new era of RNA-targeted medicine.


Assuntos
Descoberta de Drogas , Terapia Genética , Ensaios de Triagem em Larga Escala , Terapia de Alvo Molecular , RNA/genética , Animais , Biomarcadores , Estudos Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Terapia de Alvo Molecular/métodos , RNA/química , RNA/metabolismo , Interferência de RNA , Proteínas de Ligação a RNA/metabolismo , Reparo Gênico Alvo-Dirigido , Resultado do Tratamento
20.
Wiley Interdiscip Rev RNA ; 11(5): e1594, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32233021

RESUMO

Antisense oligonucleotides (ASOs) represent a new and highly promising class of drugs for personalized medicine. In the last decade, major chemical developments and improvements of the backbone structure of ASOs have transformed them into true approved and commercialized drugs. ASOs target both DNA and RNA, including pre-mRNA, mRNA, and ncRDA, based on sequence complementary. They are designed to be specific for each identified molecular and genetic alteration to restore a normal, physiological situation. Thus, the characterization of the underpinning mechanisms and alterations that sustain pathology is critical for accurate ASO-design. ASOs can be used to cure both rare and common diseases, such as orphan genetic alterations and cancer. Through pioneering examples, this review shows the versatility of the mechanisms of action that provide ASOs with the potential capacity to achieve custom treatment, revolutionizing personalized medicine. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Interactions with Proteins and Other Molecules > Small Molecule-RNA Interactions.


Assuntos
Terapia Genética , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Medicina de Precisão , Animais , Desenvolvimento de Medicamentos , Regulação da Expressão Gênica , Inativação Gênica , Terapia Genética/métodos , Humanos , Oligonucleotídeos Antissenso/química , Medicina de Precisão/métodos , Biossíntese de Proteínas , Interferência de RNA , Estabilidade de RNA , Elementos de Resposta , Reparo Gênico Alvo-Dirigido , Pesquisa Translacional Biomédica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA