Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
N Engl J Med ; 379(1): 22-31, 2018 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-29972757

RESUMO

BACKGROUND: Hereditary transthyretin amyloidosis is caused by pathogenic single-nucleotide variants in the gene encoding transthyretin ( TTR) that induce transthyretin misfolding and systemic deposition of amyloid. Progressive amyloid accumulation leads to multiorgan dysfunction and death. Inotersen, a 2'- O-methoxyethyl-modified antisense oligonucleotide, inhibits hepatic production of transthyretin. METHODS: We conducted an international, randomized, double-blind, placebo-controlled, 15-month, phase 3 trial of inotersen in adults with stage 1 (patient is ambulatory) or stage 2 (patient is ambulatory with assistance) hereditary transthyretin amyloidosis with polyneuropathy. Patients were randomly assigned, in a 2:1 ratio, to receive weekly subcutaneous injections of inotersen (300 mg) or placebo. The primary end points were the change in the modified Neuropathy Impairment Score+7 (mNIS+7; range, -22.3 to 346.3, with higher scores indicating poorer function; minimal clinically meaningful change, 2 points) and the change in the score on the patient-reported Norfolk Quality of Life-Diabetic Neuropathy (QOL-DN) questionnaire (range, -4 to 136, with higher scores indicating poorer quality of life). A decrease in scores indicated improvement. RESULTS: A total of 172 patients (112 in the inotersen group and 60 in the placebo group) received at least one dose of a trial regimen, and 139 (81%) completed the intervention period. Both primary efficacy assessments favored inotersen: the difference in the least-squares mean change from baseline to week 66 between the two groups (inotersen minus placebo) was -19.7 points (95% confidence interval [CI], -26.4 to -13.0; P<0.001) for the mNIS+7 and -11.7 points (95% CI, -18.3 to -5.1; P<0.001) for the Norfolk QOL-DN score. These improvements were independent of disease stage, mutation type, or the presence of cardiomyopathy. There were five deaths in the inotersen group and none in the placebo group. The most frequent serious adverse events in the inotersen group were glomerulonephritis (in 3 patients [3%]) and thrombocytopenia (in 3 patients [3%]), with one death associated with one of the cases of grade 4 thrombocytopenia. Thereafter, all patients received enhanced monitoring. CONCLUSIONS: Inotersen improved the course of neurologic disease and quality of life in patients with hereditary transthyretin amyloidosis. Thrombocytopenia and glomerulonephritis were managed with enhanced monitoring. (Funded by Ionis Pharmaceuticals; NEURO-TTR ClinicalTrials.gov number, NCT01737398 .).


Assuntos
Neuropatias Amiloides Familiares/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Pré-Albumina/antagonistas & inibidores , Terapêutica com RNAi , Adulto , Idoso , Idoso de 80 Anos ou mais , Neuropatias Amiloides Familiares/sangue , Neuropatias Amiloides Familiares/complicações , Progressão da Doença , Método Duplo-Cego , Feminino , Glomerulonefrite/induzido quimicamente , Humanos , Injeções Subcutâneas , Análise dos Mínimos Quadrados , Masculino , Pessoa de Meia-Idade , Oligonucleotídeos Antissenso/efeitos adversos , Polineuropatias/etiologia , Polineuropatias/terapia , Pré-Albumina/análise , Pré-Albumina/genética , Qualidade de Vida , Índice de Gravidade de Doença , Trombocitopenia/induzido quimicamente
2.
Exp Cell Res ; 395(2): 112217, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32768500

RESUMO

Hereditary transthyretin amyloidosis (ATTR) is caused by amyloid deposition of misfolded transthyretin (TTR) in various tissues. Recently, reduction of circulating serum TTR, achieved via silencing oligonucleotides, was introduced as therapy of ATTR amyloidosis. We explored the impact of Serpin Family A Member 1 (SERPINA1) on TTR mRNA and protein expression. Oncostatin M (OSM) induced SERPINA1 in hepatoma cells and mice, while concomitantly TTR expression was significantly reduced. SERPINA1 knockdown resulted in specific elevated TTR expression in hepatoma cells; however other genes belonging to the group of acute phase proteins were unaffected. In mice, serum TTR was elevated after mSERPINA1 knockdown throughout antisense treatment. Following SERPINA1 knockdown, TTR deposition in several tissues, including dorsal root ganglia and intestine, was found to be increased, however numbers did not exceed significance levels. The data suggest that SERPINA1 is a co-factor of TTR expression. Our findings provide novel insight in the regulation of TTR and reveal a role of SERPINA1 in the pathogenesis of ATTR amyloidosis.


Assuntos
Neuropatias Amiloides Familiares/metabolismo , Pré-Albumina/metabolismo , alfa 1-Antitripsina/metabolismo , Animais , Humanos , Camundongos , RNA Mensageiro/genética , alfa 1-Antitripsina/genética
3.
Proc Natl Acad Sci U S A ; 115(43): 11066-11071, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30291191

RESUMO

Centronuclear myopathies (CNM) are a group of severe muscle diseases for which no effective therapy is currently available. We have previously shown that reduction of the large GTPase DNM2 in a mouse model of the X-linked form, due to loss of myotubularin phosphatase MTM1, prevents the development of the skeletal muscle pathophysiology. As DNM2 is mutated in autosomal dominant forms, here we tested whether DNM2 reduction can rescue DNM2-related CNM in a knock-in mouse harboring the p.R465W mutation (Dnm2RW/+) and displaying a mild CNM phenotype similar to patients with the same mutation. A single intramuscular injection of adeno-associated virus-shRNA targeting Dnm2 resulted in reduction in protein levels 5 wk post injection, with a corresponding improvement in muscle mass and fiber size distribution, as well as an improvement in histopathological CNM features. To establish a systemic treatment, weekly i.p. injections of antisense oligonucleotides targeting Dnm2 were administered to Dnm2RW/+mice for 5 wk. While muscle mass, histopathology, and muscle ultrastructure were perturbed in Dnm2RW/+mice compared with wild-type mice, these features were indistinguishable from wild-type mice after reducing DNM2. Therefore, DNM2 knockdown via two different strategies can efficiently correct the myopathy due to DNM2 mutations, and it provides a common therapeutic strategy for several forms of centronuclear myopathy. Furthermore, we provide an example of treating a dominant disease by targeting both alleles, suggesting that this strategy may be applied to other dominant diseases.


Assuntos
Dinamina II/genética , Miopatias Congênitas Estruturais/genética , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Mutação/genética , Proteínas Tirosina Fosfatases não Receptoras/genética
4.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502397

RESUMO

Transthyretin (TTR) proteolysis has been recognized as a complementary mechanism contributing to transthyretin-related amyloidosis (ATTR amyloidosis). Accordingly, amyloid deposits can be composed mainly of full-length TTR or contain a mixture of both cleaved and full-length TTR, particularly in the heart. The fragmentation pattern at Lys48 suggests the involvement of a serine protease, such as plasmin. The most common TTR variant, TTR V30M, is susceptible to plasmin-mediated proteolysis, and the presence of TTR fragments facilitates TTR amyloidogenesis. Recent studies revealed that the serine protease inhibitor, SerpinA1, was differentially expressed in hepatocyte-like cells (HLCs) from ATTR patients. In this work, we evaluated the effects of SerpinA1 on in vitro and in vivo modulation of TTR V30M proteolysis, aggregation, and deposition. We found that plasmin-mediated TTR proteolysis and aggregation are partially inhibited by SerpinA1. Furthermore, in vivo downregulation of SerpinA1 increased TTR levels in mice plasma and deposition in the cardiac tissue of older animals. The presence of TTR fragments was observed in the heart of young and old mice but not in other tissues following SerpinA1 knockdown. Increased proteolytic activity, particularly plasmin activity, was detected in mice plasmas. Overall, our results indicate that SerpinA1 modulates TTR proteolysis and aggregation in vitro and in vivo.


Assuntos
Pré-Albumina/metabolismo , alfa 1-Antitripsina/metabolismo , Fatores Etários , Amiloide/metabolismo , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/fisiopatologia , Amiloidose/genética , Amiloidose/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Fibrinolisina , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pré-Albumina/genética , Pré-Albumina/fisiologia , Proteólise , alfa 1-Antitripsina/fisiologia
5.
Am J Respir Cell Mol Biol ; 63(1): 46-56, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32176858

RESUMO

Goblet cell metaplasia, excessive mucus production, and inadequate mucus clearance accompany and exacerbate multiple chronic respiratory disorders, such as asthma and chronic obstructive pulmonary disease. Notch signaling plays a central role in controlling the fate of multiple cell types in the lung, including goblet cells. In the present study, we explored the therapeutic potential of modulating the Notch pathway in the adult murine lung using chemically modified antisense oligonucleotides (ASOs). To this end, we designed and characterized ASOs targeting the Notch receptors Notch1, Notch2, and Notch3 and the Notch ligands Jag1 (Jagged 1) and Jag2 (Jagged 2). Pulmonary delivery of ASOs in healthy mice or mice exposed to house dust mite, a commonly used mouse model of asthma, resulted in a significant reduction of the respective mRNAs in the lung. Furthermore, ASO-mediated knockdown of Jag1 or Notch2 in the lungs of healthy adult mice led to the downregulation of the club cell marker Scgb1a1 and the concomitant upregulation of the ciliated cell marker FoxJ1 (forkhead box J1). Similarly, ASO-mediated knockdown of Jag1 or Notch2 in the house dust mite disease model led to reduced goblet cell metaplasia and decreased mucus production. Because goblet cell metaplasia and excessive mucus secretion are a common basis for many lung pathologies, we propose that ASO-mediated inhibition of JAG1 could provide a novel therapeutic path for the treatment of multiple chronic respiratory diseases.


Assuntos
Células Caliciformes/efeitos dos fármacos , Células Caliciformes/metabolismo , Proteína Jagged-1/metabolismo , Pulmão/efeitos dos fármacos , Metaplasia/tratamento farmacológico , Metaplasia/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Animais , Asma/metabolismo , Biomarcadores/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pyroglyphidae , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
6.
Nature ; 508(7495): 258-62, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24717514

RESUMO

In obesity and type 2 diabetes, Glut4 glucose transporter expression is decreased selectively in adipocytes. Adipose-specific knockout or overexpression of Glut4 alters systemic insulin sensitivity. Here we show, using DNA array analyses, that nicotinamide N-methyltransferase (Nnmt) is the most strongly reciprocally regulated gene when comparing gene expression in white adipose tissue (WAT) from adipose-specific Glut4-knockout or adipose-specific Glut4-overexpressing mice with their respective controls. NNMT methylates nicotinamide (vitamin B3) using S-adenosylmethionine (SAM) as a methyl donor. Nicotinamide is a precursor of NAD(+), an important cofactor linking cellular redox states with energy metabolism. SAM provides propylamine for polyamine biosynthesis and donates a methyl group for histone methylation. Polyamine flux including synthesis, catabolism and excretion, is controlled by the rate-limiting enzymes ornithine decarboxylase (ODC) and spermidine-spermine N(1)-acetyltransferase (SSAT; encoded by Sat1) and by polyamine oxidase (PAO), and has a major role in energy metabolism. We report that NNMT expression is increased in WAT and liver of obese and diabetic mice. Nnmt knockdown in WAT and liver protects against diet-induced obesity by augmenting cellular energy expenditure. NNMT inhibition increases adipose SAM and NAD(+) levels and upregulates ODC and SSAT activity as well as expression, owing to the effects of NNMT on histone H3 lysine 4 methylation in adipose tissue. Direct evidence for increased polyamine flux resulting from NNMT inhibition includes elevated urinary excretion and adipocyte secretion of diacetylspermine, a product of polyamine metabolism. NNMT inhibition in adipocytes increases oxygen consumption in an ODC-, SSAT- and PAO-dependent manner. Thus, NNMT is a novel regulator of histone methylation, polyamine flux and NAD(+)-dependent SIRT1 signalling, and is a unique and attractive target for treating obesity and type 2 diabetes.


Assuntos
Dieta , Nicotinamida N-Metiltransferase/deficiência , Nicotinamida N-Metiltransferase/metabolismo , Obesidade/enzimologia , Obesidade/prevenção & controle , Acetiltransferases/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/enzimologia , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/enzimologia , Tecido Adiposo Branco/metabolismo , Animais , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético , Fígado Gorduroso , Técnicas de Silenciamento de Genes , Intolerância à Glucose , Transportador de Glucose Tipo 4/deficiência , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Resistência à Insulina , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NAD/metabolismo , Niacinamida/metabolismo , Nicotinamida N-Metiltransferase/genética , Obesidade/etiologia , Obesidade/genética , Ornitina Descarboxilase/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , S-Adenosilmetionina/metabolismo , Sirtuína 1/metabolismo , Espermina/análogos & derivados , Espermina/metabolismo , Magreza/enzimologia , Magreza/metabolismo , Poliamina Oxidase
7.
Mol Ther ; 27(10): 1749-1757, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31351782

RESUMO

Cystic fibrosis (CF) is an autosomal recessive monogenic disease caused by mutations in the CFTR gene. Therapeutic approaches that are focused on correcting CFTR protein face the challenge of the heterogeneity in CFTR mutations and resulting defects. Thus, while several small molecules directed at CFTR show benefit in the clinic for subsets of CF patients, these drugs cannot treat all CF patients. Additionally, the clinical benefit from treatment with these modulators could be enhanced with novel therapies. To address this unmet need, we utilized an approach to increase CFTR protein levels through antisense oligonucleotide (ASO)-mediated steric inhibition of 5' UTR regulatory elements. We identified ASOs to upregulate CFTR protein expression and confirmed the regulatory role of the sites amenable to ASO-mediated upregulation. Two ASOs were investigated further, and both increased CFTR protein expression and function in cell lines and primary human bronchial epithelial cells with distinct CF genotypes. ASO treatment further increased CFTR function in almost all CF genotypes tested on top of treatment with the FDA approved drug Symdeko (ivacaftor and tezacaftor). Thus, we present a novel approach to CFTR therapeutic intervention, through ASO-mediated modulation of translation.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Regulação para Cima , Regiões 5' não Traduzidas , Aminofenóis/farmacologia , Animais , Benzodioxóis/farmacologia , Linhagem Celular , Combinação de Medicamentos , Sinergismo Farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Genótipo , Células HeLa , Humanos , Indóis/farmacologia , Conformação de Ácido Nucleico , Oligonucleotídeos Antissenso/química , Quinolonas/farmacologia
8.
Am J Respir Cell Mol Biol ; 61(3): 290-300, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30836009

RESUMO

The recessive genetic disease cystic fibrosis (CF) is caused by loss-of-function mutations in the CFTR (CF transmembrane conductance regulator) gene. Approximately 10% of patients with CF have at least one allele with a nonsense mutation in CFTR. Nonsense mutations generate premature termination codons that can subject mRNA transcripts to rapid degradation through the nonsense-mediated mRNA decay (NMD) pathway. Currently, there are no approved therapies that specifically target nonsense mutations in CFTR. Here, we identified antisense oligonucleotides (ASOs) that target the NMD factor SMG1 to inhibit the NMD pathway, and determined their effects on the W1282X CFTR mutation. First, we developed and validated two in vitro models of the W1282X CFTR mutation. Next, we treated these cells with antisense oligonucleotides to inhibit NMD and measured the effects of these treatments on W1282X expression and function. SMG1-ASO-mediated NMD inhibition upregulated the RNA, protein, and surface-localized protein expression of the truncated W1282X gene product. Additionally, these ASOs increased the CFTR chloride channel function in cells homozygous for the W1282X mutation. Our approach suggests a new therapeutic strategy for patients harboring nonsense mutations and may be beneficial as a single agent in patients with CF and the W1282X mutation.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Degradação do RNAm Mediada por Códon sem Sentido/efeitos dos fármacos , RNA/genética , Aminofenóis/farmacologia , Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Homozigoto , Humanos , Mutação/genética , Quinolonas/farmacologia , RNA/metabolismo
9.
Hum Mol Genet ; 25(23): 5178-5187, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28007899

RESUMO

ISCU myopathy is an inherited disease that primarily affects individuals of northern Swedish descent who share a single point mutation in the fourth intron of the ISCU gene. The current study shows correction of specific phenotypes associated with disease following treatment with an antisense oligonucleotide (ASO) targeted to the site of the mutation. We have shown that ASO treatment diminished aberrant splicing and increased ISCU protein levels in both patient fibroblasts and patient myotubes in a concentration dependent fashion. Upon ASO treatment, levels of SDHB in patient myotubular cell lines increased to levels observed in control myotubular cell lines. Additionally, we have shown that both patient fibroblast and myotubular cell lines displayed an increase in complex II activity with a concomitant decrease in succinate levels in patient myotubular cell lines after ASO treatment. Mitochondrial and cytosolic aconitase activities increased significantly following ASO treatment in patient myotubes. The current study suggests that ASO treatment may serve as a viable approach to correcting ISCU myopathy in patients.


Assuntos
Acidose Láctica/congênito , Proteínas Ferro-Enxofre/genética , Doenças Musculares/congênito , Oligonucleotídeos Antissenso/genética , Succinato Desidrogenase/genética , Acidose Láctica/genética , Acidose Láctica/patologia , Acidose Láctica/terapia , Feminino , Humanos , Íntrons/genética , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Doenças Musculares/genética , Doenças Musculares/patologia , Doenças Musculares/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Fenótipo , Mutação Puntual , Splicing de RNA/efeitos dos fármacos , Splicing de RNA/genética , Succinato Desidrogenase/biossíntese
10.
N Engl J Med ; 372(3): 232-40, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25482425

RESUMO

BACKGROUND: Experimental data indicate that reducing factor XI levels attenuates thrombosis without causing bleeding, but the role of factor XI in the prevention of postoperative venous thrombosis in humans is unknown. FXI-ASO (ISIS 416858) is a second-generation antisense oligonucleotide that specifically reduces factor XI levels. We compared the efficacy and safety of FXI-ASO with those of enoxaparin in patients undergoing total knee arthroplasty. METHODS: In this open-label, parallel-group study, we randomly assigned 300 patients who were undergoing elective primary unilateral total knee arthroplasty to receive one of two doses of FXI-ASO (200 mg or 300 mg) or 40 mg of enoxaparin once daily. The primary efficacy outcome was the incidence of venous thromboembolism (assessed by mandatory bilateral venography or report of symptomatic events). The principal safety outcome was major or clinically relevant nonmajor bleeding. RESULTS: Around the time of surgery, the mean (±SE) factor XI levels were 0.38±0.01 units per milliliter in the 200-mg FXI-ASO group, 0.20±0.01 units per milliliter in the 300-mg FXI-ASO group, and 0.93±0.02 units per milliliter in the enoxaparin group. The primary efficacy outcome occurred in 36 of 134 patients (27%) who received the 200-mg dose of FXI-ASO and in 3 of 71 patients (4%) who received the 300-mg dose of FXI-ASO, as compared with 21 of 69 patients (30%) who received enoxaparin. The 200-mg regimen was noninferior, and the 300-mg regimen was superior, to enoxaparin (P<0.001). Bleeding occurred in 3%, 3%, and 8% of the patients in the three study groups, respectively. CONCLUSIONS: This study showed that factor XI contributes to postoperative venous thromboembolism; reducing factor XI levels in patients undergoing elective primary unilateral total knee arthroplasty was an effective method for its prevention and appeared to be safe with respect to the risk of bleeding. (Funded by Isis Pharmaceuticals; FXI-ASO TKA ClinicalTrials.gov number, NCT01713361.).


Assuntos
Anticoagulantes/administração & dosagem , Artroplastia do Joelho , Fator XI/antagonistas & inibidores , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos/administração & dosagem , Complicações Pós-Operatórias/prevenção & controle , Trombose Venosa/prevenção & controle , Adulto , Idoso , Anticoagulantes/efeitos adversos , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Protocolos Clínicos , Enoxaparina/administração & dosagem , Enoxaparina/efeitos adversos , Fator XI/análise , Feminino , Hemorragia/induzido quimicamente , Humanos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Oligonucleotídeos/efeitos adversos , Oligonucleotídeos Antissenso/efeitos adversos , Tempo de Tromboplastina Parcial
11.
Mol Vis ; 23: 561-571, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28855795

RESUMO

PURPOSE: Age-related macular degeneration (AMD) is the leading cause of permanent vision loss among the elderly in many industrialized countries, and the complement system plays an important role in the pathogenesis of AMD. Inhibition of complement factor B, a key regulator of the alternative pathway, is implicated as a potential therapeutic intervention for AMD. Here we investigated the effect of liver factor B reduction on systemic and ocular factor B levels. METHODS: Second-generation antisense oligonucleotides (ASOs) targeting mouse and monkey factor B mRNA were administered by subcutaneous injection to healthy mice or monkeys, and the level of factor B mRNA was assessed in the liver and the eye. In addition, the factor B protein level was determined in plasma and whole eyes from the treated animals. RESULTS: Mice and monkeys treated with factor B ASOs demonstrated a robust reduction in liver factor B mRNA levels with no change in ocular factor B mRNA levels. Plasma factor B protein levels were significantly reduced in mice and monkeys treated with factor B ASOs, leading to a dramatic reduction in ocular factor B protein, below the assay detection levels. CONCLUSIONS: The results add to the increasing evidence that the liver is the main source of plasma and ocular factor B protein, and demonstrate that reduction of liver factor B mRNA by an ASO results in a significant reduction in plasma and ocular factor B protein levels. The results suggest that inhibition of liver factor B mRNA by factor B ASOs would reduce systemic alternative complement pathway activation and has potential to be used as a novel therapy for AMD.


Assuntos
Fator B do Complemento/genética , Fator B do Complemento/metabolismo , Olho/metabolismo , Fígado/metabolismo , Oligonucleotídeos Antissenso/administração & dosagem , RNA Mensageiro/metabolismo , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Injeções Subcutâneas , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
12.
Blood ; 126(15): 1844-55, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26286849

RESUMO

Sickle cell disease (SCD) results in vascular occlusions, chronic hemolytic anemia, and cumulative organ damage. A conspicuous feature of SCD is chronic inflammation and coagulation system activation. Thrombin (factor IIa [FIIa]) is both a central protease in hemostasis and a key modifier of inflammatory processes. To explore the hypothesis that reduced prothrombin (factor II [FII]) levels in SCD will limit vaso-occlusion, vasculopathy, and inflammation, we used 2 strategies to suppress FII in SCD mice. Weekly administration of FII antisense oligonucleotide "gapmer" to Berkeley SCD mice to selectively reduce circulating FII levels to ∼10% of normal for 15 weeks significantly diminished early mortality. More comprehensive, long-term comparative studies were done using mice with genetic diminution of circulating FII. Here, cohorts of FII(lox/-) mice (constitutively carrying ∼10% normal FII) and FII(WT) mice were tracked in parallel for a year following the imposition of SCD via hematopoietic stem cell transplantation. This genetically imposed suppression of FII levels resulted in an impressive reduction in inflammation (reduction in leukocytosis, thrombocytosis, and circulating interleukin-6 levels), reduced endothelial cell dysfunction (reduced endothelial activation and circulating soluble vascular cell adhesion molecule), and a significant improvement in SCD-associated end-organ damage (nephropathy, pulmonary hypertension, pulmonary inflammation, liver function, inflammatory infiltration, and microinfarctions). Notably, all of these benefits were achieved with a relatively modest 1.25-fold increase in prothrombin times, and in the absence of hemorrhagic complications. Taken together, these data establish that prothrombin is a powerful modifier of SCD-induced end-organ damage, and present a novel therapeutic target to ameliorate SCD pathologies.


Assuntos
Anemia Falciforme/complicações , Terapia Genética , Hipertensão Pulmonar/prevenção & controle , Inflamação/prevenção & controle , Protrombina/fisiologia , Doenças Vasculares/prevenção & controle , Anemia Falciforme/mortalidade , Anemia Falciforme/fisiopatologia , Animais , Coagulação Sanguínea , Células Cultivadas , Hipertensão Pulmonar/etiologia , Técnicas Imunoenzimáticas , Inflamação/etiologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Knockout , Oligorribonucleotídeos Antissenso/farmacologia , Protrombina/antagonistas & inibidores , Taxa de Sobrevida , Trombina/metabolismo , Doenças Vasculares/etiologia
13.
Blood ; 125(17): 2712-9, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25691157

RESUMO

Factor (F) XII, a key component of the contact system, triggers clotting via the intrinsic pathway, and is implicated in propagating thrombosis. Although nucleic acids are potent activators, it is unclear how the contact system is regulated to prevent uncontrolled clotting. Previously, we showed that histidine-rich glycoprotein (HRG) binds FXIIa and attenuates its capacity to trigger coagulation. To investigate the role of HRG as a regulator of the intrinsic pathway, we compared RNA- and DNA-induced thrombin generation in plasma from HRG-deficient and wild-type mice. Thrombin generation was enhanced in plasma from HRG-deficient mice, and accelerated clotting was restored to normal with HRG reconstitution. Although blood loss after tail tip amputation was similar in HRG-deficient and wild-type mice, carotid artery occlusion after FeCl3 injury was accelerated in HRG-deficient mice, and HRG administration abrogated this effect. To confirm that HRG modulates the contact system, we used DNase, RNase, and antisense oligonucleotides to characterize the FeCl3 model. Whereas DNase or FVII knockdown had no effect, carotid occlusion was abrogated with RNase or FXII knockdown, confirming that FeCl3-induced thrombosis is triggered by RNA in a FXII-dependent fashion. Therefore, in a nucleic acid-driven model, HRG inhibits thrombosis by modulating the intrinsic pathway of coagulation.


Assuntos
Coagulação Sanguínea , Proteínas/genética , Trombose/sangue , Trombose/genética , Animais , Cloretos , Fator XII/genética , Fator XII/metabolismo , Feminino , Compostos Férricos , Deleção de Genes , Técnicas de Silenciamento de Genes , Hemostasia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas/análise , Proteínas/metabolismo , Trombina/metabolismo , Trombose/induzido quimicamente , Trombose/metabolismo
14.
Cardiovasc Drugs Ther ; 31(4): 445-458, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28735360

RESUMO

PURPOSE: The need for novel approaches to cardiovascular drug development served as the impetus to convene an open meeting of experts from the pharmaceutical industry and academia to assess the challenges and develop solutions for drug discovery in cardiovascular disease. METHODS: The Novel Cardiovascular Therapeutics Summit first reviewed recent examples of ongoing or recently completed programs translating basic science observations to targeted drug development, highlighting successes (protein convertase sutilisin/kexin type 9 [PCSK9] and neprilysin inhibition) and targets still under evaluation (cholesteryl ester transfer protein [CETP] inhibition), with the hope of gleaning key lessons to successful drug development in the current era. Participants then reviewed the use of innovative approaches being explored to facilitate rapid and more cost-efficient evaluations of drug candidates in a short timeframe. RESULTS: We summarize observations gleaned from this summit and offer insight into future cardiovascular drug development. CONCLUSIONS: The rapid development in genetic and high-throughput drug evaluation technologies, coupled with new approaches to rapidly evaluate potential cardiovascular therapies with in vitro techniques, offer opportunities to identify new drug targets for cardiovascular disease, study new therapies with better efficiency and higher throughput in the preclinical setting, and more rapidly bring the most promising therapies to human testing. However, there must be a critical interface between industry and academia to guide the future of cardiovascular drug development. The shared interest among academic institutions and pharmaceutical companies in developing promising therapies to address unmet clinical needs for patients with cardiovascular disease underlies and guides innovation and discovery platforms that are significantly altering the landscape of cardiovascular drug development.


Assuntos
Fármacos Cardiovasculares/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Desenho de Fármacos , Animais , Fármacos Cardiovasculares/farmacologia , Doenças Cardiovasculares/fisiopatologia , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Indústria Farmacêutica , Humanos
15.
Blood ; 123(13): 2102-7, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24501216

RESUMO

Central venous catheter thrombosis can cause venous obstruction and pulmonary embolism. To determine the extent to which catheter thrombosis is triggered by the contact or extrinsic pathway of coagulation, we used antisense oligonucleotides (ASOs) to selectively knock down factor (f)XII, fXI, or high-molecular-weight kininogen (HK), key components of the contact pathway, or fVII, which is essential for the extrinsic pathway. Knockdown of contact pathway components prolonged the activated partial thromboplastin time and decreased target protein activity levels by over 90%, whereas fVII knockdown prolonged the prothrombin time and reduced fVII activity to a similar extent. Using a rabbit model of catheter thrombosis, catheters implanted in the jugular vein were assessed daily until they occluded, up to a maximum of 35 days. Compared with control, fXII and fXI ASO treatment prolonged the time to catheter occlusion by 2.2- and 2.3-fold, respectively. In contrast, both HK and fVII knockdown did not significantly prolong the time to occlusion, and dual treatment with fVII- and fXI-directed ASOs produced a time to occlusion similar to that with the fXI ASO alone. These findings suggest that catheter thrombosis is triggered via the contact pathway and identify fXII and fXI as potential targets to attenuate this complication.


Assuntos
Catéteres/efeitos adversos , Fator XII/genética , Fator XI/genética , Oligonucleotídeos Antissenso/farmacologia , Interferência de RNA/fisiologia , Trombose/prevenção & controle , Animais , Obstrução do Cateter , Modelos Animais de Doenças , Fator XI/antagonistas & inibidores , Fator XII/antagonistas & inibidores , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética/métodos , Masculino , Coelhos , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/genética , Trombose/genética
16.
Nucleic Acids Res ; 42(13): 8796-807, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24992960

RESUMO

Triantennary N-acetyl galactosamine (GalNAc, GN3: ), a high-affinity ligand for the hepatocyte-specific asialoglycoprotein receptor (ASGPR), enhances the potency of second-generation gapmer antisense oligonucleotides (ASOs) 6-10-fold in mouse liver. When combined with next-generation ASO designs comprised of short S-cEt (S-2'-O-Et-2',4'-bridged nucleic acid) gapmer ASOs, ∼ 60-fold enhancement in potency relative to the parent MOE (2'-O-methoxyethyl RNA) ASO was observed. GN3: -conjugated ASOs showed high affinity for mouse ASGPR, which results in enhanced ASO delivery to hepatocytes versus non-parenchymal cells. After internalization into cells, the GN3: -ASO conjugate is metabolized to liberate the parent ASO in the liver. No metabolism of the GN3: -ASO conjugate was detected in plasma suggesting that GN3: acts as a hepatocyte targeting prodrug that is detached from the ASO by metabolism after internalization into the liver. GalNAc conjugation also enhanced potency and duration of the effect of two ASOs targeting human apolipoprotein C-III and human transthyretin (TTR) in transgenic mice. The unconjugated ASOs are currently in late stage clinical trials for the treatment of familial chylomicronemia and TTR-mediated polyneuropathy. The ability to translate these observations in humans offers the potential to improve therapeutic index, reduce cost of therapy and support a monthly dosing schedule for therapeutic suppression of gene expression in the liver using ASOs.


Assuntos
Galactosamina/análogos & derivados , Glicolipídeos/química , Hepatócitos/metabolismo , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/química , Animais , Apolipoproteína C-III/genética , Receptor de Asialoglicoproteína/metabolismo , Fator XI/antagonistas & inibidores , Galactosamina/química , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligonucleotídeos Antissenso/metabolismo , Pré-Albumina/antagonistas & inibidores , alfa 1-Antitripsina
17.
Arterioscler Thromb Vasc Biol ; 34(8): 1668-73, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24947525

RESUMO

OBJECTIVE: Coagulation factor XI is proposed as therapeutic target for anticoagulation. However, it is still unclear whether the antithrombotic properties of factor XI inhibitors influence atherosclerotic disease and atherothrombosis. Our aim is to investigate whether factor XI antisense oligonucleotides could prevent thrombus formation on acutely ruptured atherosclerotic plaques. APPROACH AND RESULTS: Atherosclerotic plaques in the carotid arteries of Apoe(-/-) mice were acutely ruptured using ultrasound. The subsequent thrombus formation was visualized and quantified by intravital microscopy and immunohistochemistry. Mice were pretreated with either factor XI antisense or nonsense oligonucleotides (50 mg/kg) to lower factor XI plasma levels. A tail bleeding assay was used to determine the safety. On plaque rupture, initial platelet adhesion and platelet plug formation were not impaired in animals treated with factor XI antisense oligonucleotides. However, the ensuing thrombus formation and fibrin deposition were significantly lower after 5 to 10 minutes (P<0.05) in factor XI antisense oligonucleotide-treated animals without inducing a bleeding tendency. Furthermore, thrombi from antisense-treated animals were less stable than thrombi from placebo-treated animals. Moreover, macrophage infiltration and collagen deposition were lower in the carotid arteries of factor XI antisense-treated animals. No neutrophils were present. CONCLUSIONS: Factor XI antisense oligonucleotides safely prevent thrombus formation on acutely ruptured atherosclerotic plaques in mice. Furthermore, perturbed carotid arteries from factor XI antisense-treated animals show a less severe inflammatory response.


Assuntos
Aterosclerose/complicações , Coagulação Sanguínea , Plaquetas/metabolismo , Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/complicações , Fator XI/metabolismo , Placa Aterosclerótica , Trombose/etiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Artérias Carótidas/imunologia , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/sangue , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/imunologia , Doenças das Artérias Carótidas/patologia , Colesterol na Dieta , Colágeno/metabolismo , Modelos Animais de Doenças , Fator XI/genética , Fibrina/metabolismo , Inflamação/sangue , Inflamação/imunologia , Inflamação/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligonucleotídeos Antissenso/administração & dosagem , Adesividade Plaquetária , Agregação Plaquetária , Ruptura Espontânea , Trombose/sangue , Trombose/genética , Trombose/patologia , Trombose/prevenção & controle , Fatores de Tempo
18.
Arterioscler Thromb Vasc Biol ; 33(7): 1670-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23559626

RESUMO

OBJECTIVE: During coagulation, factor IX (FIX) is activated by 2 distinct mechanisms mediated by the active proteases of either FVIIa or FXIa. Both coagulation factors may contribute to thrombosis; FXI, however, plays only a limited role in the arrest of bleeding. Therefore, therapeutic targeting of FXI may produce an antithrombotic effect with relatively low hemostatic risk. APPROACH AND RESULTS: We have reported that reducing FXI levels with FXI antisense oligonucleotides produces antithrombotic activity in mice, and that administration of FXI antisense oligonucleotides to primates decreases circulating FXI levels and activity in a dose-dependent and time-dependent manner. Here, we evaluated the relationship between FXI plasma levels and thrombogenicity in an established baboon model of thrombosis and hemostasis. In previous studies with this model, antibody-induced inhibition of FXI produced potent antithrombotic effects. In the present article, antisense oligonucleotides-mediated reduction of FXI plasma levels by ≥ 50% resulted in a demonstrable and sustained antithrombotic effect without an increased risk of bleeding. CONCLUSIONS: These results indicate that reducing FXI levels using antisense oligonucleotides is a promising alternative to direct FXI inhibition, and that targeting FXI may be potentially safer than conventional antithrombotic therapies that can markedly impair primary hemostasis.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator XI/metabolismo , Fibrinolíticos/administração & dosagem , Oligonucleotídeos Antissenso/administração & dosagem , Trombose/prevenção & controle , Animais , Anticorpos Monoclonais/administração & dosagem , Derivação Arteriovenosa Cirúrgica , Tempo de Sangramento , Colágeno , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo , Fator XI/antagonistas & inibidores , Fator XI/genética , Fibrinolíticos/toxicidade , Hemorragia/induzido quimicamente , Macaca fascicularis , Oligonucleotídeos Antissenso/toxicidade , Papio , Trombina/metabolismo , Trombose/sangue , Trombose/etiologia , Trombose/genética , Fatores de Tempo
19.
Nat Rev Cancer ; 5(6): 468-79, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15905854

RESUMO

Improved understanding of the molecular mechanisms that mediate cancer progression and therapeutic resistance has identified many therapeutic gene targets that regulate apoptosis, proliferation and cell signalling. Antisense oligonucleotides offer one approach to target genes involved in cancer progression, especially those that are not amenable to small-molecule or antibody inhibition. Better chemical modifications of antisense oligonucleotides increase resistance to nuclease digestion, prolong tissue half-lives and improve scheduling. Indeed, recent clinical trials confirm the ability of this class of drugs to significantly suppress target-gene expression. The current status and future directions of several antisense drugs that have potential clinical use in cancer are reviewed.


Assuntos
Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Apoptose , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Oligonucleotídeos Antissenso/farmacocinética , Oncogenes , Regulação para Cima
20.
Hepatology ; 55(6): 1746-53, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22234891

RESUMO

UNLABELLED: Type 2 diabetes mellitus (T2DM) impairs hepatic clearance of atherogenic postprandial triglyceride-rich lipoproteins (TRLs). We recently reported that livers from T2DM db/db mice markedly overexpress the heparan sulfate glucosamine-6-O-endosulfatase-2 (SULF2), an enzyme that removes 6-O sulfate groups from heparan sulfate proteoglycans (HSPGs) and suppresses uptake of TRLs by cultured hepatocytes. In the present study, we evaluated whether Sulf2 inhibition in T2DM mice in vivo could correct their postprandial dyslipidemia. Selective second-generation antisense oligonucleotides (ASOs) targeting Sulf2 were identified. Db/db mice were treated for 5 weeks with Sulf2 ASO (20 or 50 mg/kg per week), nontarget (NT) ASO, or phosphate-buffered saline (PBS). Administration of Sulf2 ASO to db/db mice suppressed hepatic Sulf2 messenger RNA expression by 70%-80% (i.e., down to levels in nondiabetic db/m mice) and increased the ratio of tri- to disulfated disaccharides in hepatic HSPGs (P < 0.05). Hepatocytes isolated from db/db mice on NT ASO exhibited a significant impairment in very-low-density lipoprotein (VLDL) binding that was entirely corrected in db/db mice on Sulf2 ASO. Sulf2 ASO lowered the random, nonfasting plasma triglyceride (TG) levels by 50%, achieving nondiabetic values. Most important, Sulf2 ASO treatment flattened the plasma TG excursions in db/db mice after corn-oil gavage (iAUC, 1,500 ± 470 mg/dL·h for NT ASO versus 160 ± 40 mg/dL · h for Sulf2 ASO\P < 0.01). CONCLUSIONS: Despite extensive metabolic derangements in T2DM mice, inhibition of a single dys-regulated molecule, SULF2, normalizes the VLDL-binding capacity of their hepatocytes and abolishes postprandial hypertriglyceridemia. These findings provide a key proof of concept in vivo to support Sulf2 inhibition as an attractive strategy to improve metabolic dyslipidemia.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Dislipidemias/tratamento farmacológico , Fígado/enzimologia , Oligonucleotídeos Antissenso/uso terapêutico , Sulfatases/antagonistas & inibidores , Animais , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/metabolismo , Hepatócitos/metabolismo , Humanos , Lipoproteínas VLDL/metabolismo , Masculino , Camundongos , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA