Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 3274, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37280238

RESUMO

SARS-CoV-2 has resulted in high levels of morbidity and mortality world-wide, and severe complications can occur in older populations. Humoral immunity induced by authorized vaccines wanes within 6 months, and frequent boosts may only offer transient protection. GRT-R910 is an investigational self-amplifying mRNA (samRNA)-based SARS-CoV-2 vaccine delivering full-length Spike and selected conserved non-Spike T cell epitopes. This study reports interim analyses for a phase I open-label dose-escalation trial evaluating GRT-R910 in previously vaccinated healthy older adults (NCT05148962). Primary endpoints of safety and tolerability were assessed. Most solicited local and systemic adverse events (AEs) following GRT-R910 dosing were mild to moderate and transient, and no treatment-related serious AEs were observed. The secondary endpoint of immunogenicity was assessed via IgG binding assays, neutralization assays, interferon-gamma ELISpot, and intracellular cytokine staining. Neutralizing antibody titers against ancestral Spike and variants of concern were boosted or induced by GRT-R910 and, contrasting to authorized vaccines, persisted through at least 6 months after the booster dose. GRT-R910 increased and/or broadened functional Spike-specific T cell responses and primed functional T cell responses to conserved non-Spike epitopes. This study is limited due to small sample size, and additional data from ongoing studies will be required to corroborate these interim findings.


Assuntos
COVID-19 , RNA Mensageiro/genética , COVID-19/prevenção & controle , Humanos , Idoso , Masculino , Feminino , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Ensaios Clínicos como Assunto , Anticorpos Antivirais/imunologia , Anticorpos Neutralizantes/imunologia , Linfócitos T/imunologia
2.
Nat Med ; 28(8): 1619-1629, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35970920

RESUMO

Checkpoint inhibitor (CPI) therapies provide limited benefit to patients with tumors of low immune reactivity. T cell-inducing vaccines hold promise to exert long-lasting disease control in combination with CPI therapy. Safety, tolerability and recommended phase 2 dose (RP2D) of an individualized, heterologous chimpanzee adenovirus (ChAd68) and self-amplifying mRNA (samRNA)-based neoantigen vaccine in combination with nivolumab and ipilimumab were assessed as primary endpoints in an ongoing phase 1/2 study in patients with advanced metastatic solid tumors (NCT03639714). The individualized vaccine regimen was safe and well tolerated, with no dose-limiting toxicities. Treatment-related adverse events (TRAEs) >10% included pyrexia, fatigue, musculoskeletal and injection site pain and diarrhea. Serious TRAEs included one count each of pyrexia, duodenitis, increased transaminases and hyperthyroidism. The RP2D was 1012 viral particles (VP) ChAd68 and 30 µg samRNA. Secondary endpoints included immunogenicity, feasibility of manufacturing and overall survival (OS). Vaccine manufacturing was feasible, with vaccination inducing long-lasting neoantigen-specific CD8 T cell responses. Several patients with microsatellite-stable colorectal cancer (MSS-CRC) had improved OS. Exploratory biomarker analyses showed decreased circulating tumor DNA (ctDNA) in patients with prolonged OS. Although small study size limits statistical and translational analyses, the increased OS observed in MSS-CRC warrants further exploration in larger randomized studies.


Assuntos
Neoplasias Colorretais , Pan troglodytes , Adenoviridae/genética , Animais , Neoplasias Colorretais/tratamento farmacológico , Febre , Humanos , RNA Mensageiro/uso terapêutico
3.
Nat Commun ; 13(1): 3289, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35672369

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic continues to spread globally, highlighting the urgent need for safe and effective vaccines that could be rapidly mobilized to immunize large populations. We report the preclinical development of a self-amplifying mRNA (SAM) vaccine encoding a prefusion stabilized severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein and demonstrate strong cellular and humoral immune responses at low doses in mice and rhesus macaques. The homologous prime-boost vaccination regimen of SAM at 3, 10 and 30 µg induced potent neutralizing antibody (nAb) titers in rhesus macaques following two SAM vaccinations at all dose levels, with the 10 µg dose generating geometric mean titers (GMT) 48-fold greater than the GMT of a panel of SARS-CoV-2 convalescent human sera. Spike-specific T cell responses were observed with all tested vaccine regimens. SAM vaccination provided protective efficacy against SARS-CoV-2 challenge as both a homologous prime-boost and as a single boost following ChAd prime, demonstrating reduction of viral replication in both the upper and lower airways. The SAM vaccine is currently being evaluated in clinical trials as both a homologous prime-boost regimen at low doses and as a boost following heterologous prime.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Humanos , Macaca mulatta/genética , Camundongos , RNA Mensageiro , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Vacinação
4.
Nat Commun ; 9(1): 4671, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30405106

RESUMO

The epithelial-to-mesenchymal transition (EMT) is an important mechanism for cancer progression and metastasis. Numerous in vitro and tumor-profiling studies point to the miR-200-Zeb1 axis as crucial in regulating this process, yet in vivo studies involving its regulation within a physiological context are lacking. Here, we show that miR-200 ablation in the Rip-Tag2 insulinoma mouse model induces beta-cell dedifferentiation, initiates an EMT expression program, and promotes tumor invasion. Strikingly, disrupting the miR-200 sites of the endogenous Zeb1 locus causes a similar phenotype. Reexpressing members of the miR-200 superfamily in vitro reveals that the miR-200c family and not the co-expressed and closely related miR-141 family is responsible for regulation of Zeb1 and EMT. Our results thus show that disrupting the in vivo regulation of Zeb1 by miR-200c is sufficient to drive EMT, thus highlighting the importance of this axis in tumor progression and invasion and its potential as a therapeutic target.


Assuntos
Diferenciação Celular , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/genética , Proliferação de Células/genética , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Mutação/genética , Invasividade Neoplásica , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
6.
Nature ; 560(7717): E26, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29849139

RESUMO

In Fig. 4e of this Article, the labels for 'Control' and 'HFD' were reversed ('Control' should have been labelled blue rather than purple, and 'HFD' should have been labelled purple rather than blue). Similarly, in Fig. 4f of this Article, the labels for 'V' and 'GW' were reversed ('V' should have been labelled blue rather than purple, and 'GW' should have been labelled purple instead of blue). The original figure has been corrected online.

7.
Cell Rep ; 21(12): 3548-3558, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29262333

RESUMO

Isolated methylmalonic acidemia/aciduria (MMA) is a devastating metabolic disorder with poor outcomes despite current medical treatments. Like other mitochondrial enzymopathies, enzyme replacement therapy (ERT) is not available, and although promising, AAV gene therapy can be limited by pre-existing immunity and has been associated with genotoxicity in mice. To develop a new class of therapy for MMA, we generated a pseudoU-modified codon-optimized mRNA encoding human methylmalonyl-CoA mutase (hMUT), the enzyme most frequently mutated in MMA, and encapsulated it into biodegradable lipid nanoparticles (LNPs). Intravenous (i.v.) administration of hMUT mRNA in two different mouse models of MMA resulted in a 75%-85% reduction in plasma methylmalonic acid and was associated with increased hMUT protein expression and activity in liver. Repeat dosing of hMUT mRNA reduced circulating metabolites and dramatically improved survival and weight gain. Additionally, repeat i.v. dosing did not increase markers of liver toxicity or inflammation in heterozygote MMA mice.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/terapia , Terapia Genética/métodos , Metilmalonil-CoA Mutase/genética , Nanopartículas/administração & dosagem , RNA Mensageiro/genética , Administração Intravenosa , Animais , Feminino , Humanos , Lipídeos/química , Fígado/metabolismo , Masculino , Metilmalonil-CoA Mutase/metabolismo , Camundongos , Nanopartículas/química , RNA Mensageiro/metabolismo
8.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26935695

RESUMO

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias do Colo/patologia , Dieta Hiperlipídica/efeitos adversos , Intestinos/patologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Animais , Contagem de Células , Autorrenovação Celular/efeitos dos fármacos , Feminino , Genes APC , Humanos , Masculino , Camundongos , Obesidade/induzido quimicamente , Obesidade/patologia , Organoides/efeitos dos fármacos , Organoides/metabolismo , Organoides/patologia , PPAR delta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , beta Catenina/metabolismo
9.
Mol Cell ; 43(6): 993-1004, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21925387

RESUMO

The miR-16 family, which targets genes important for the G1-S transition, is a known modulator of the cell cycle, and members of this family are often deleted or downregulated in many types of cancers. Here, we report the reciprocal relationship-that of the cell cycle controlling the miR-16 family. Levels of this family increase rapidly as cells are arrested in G0. Conversely, as cells are released from G0 arrest, levels of the miR-16 family rapidly decrease. Such rapid changes are made possible by the unusual instabilities of several family members. The repression mediated by the miR-16 family is sensitive to these cell-cycle changes, which suggests that the rapid upregulation of the miR-16 family reinforces cell-cycle arrest in G0. Upon cell-cycle re-entry, the rapid decay of several members allows levels of the family to decrease, alleviating repression of target genes and allowing proper resumption of the cell cycle.


Assuntos
Ciclo Celular/genética , MicroRNAs/metabolismo , Estabilidade de RNA , Animais , Fase G1/genética , Camundongos , Células NIH 3T3 , Fase S/genética
10.
Proc Natl Acad Sci U S A ; 104(43): 17128-33, 2007 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-17940001

RESUMO

The tmRNA-SmpB system releases ribosomes stalled on truncated mRNAs and tags the nascent polypeptides to target them for proteolysis. In many species, mutations that disrupt tmRNA activity cause defects in growth or development. In Caulobacter crescentus cells lacking tmRNA activity there is a delay in the initiation of DNA replication, which disrupts the cell cycle. To understand the molecular basis for this phenotype, 73 C. crescentus proteins were identified that are tagged by tmRNA under normal growth conditions. Among these substrates, proteins involved in DNA replication, recombination, and repair were overrepresented, suggesting that misregulation of these factors in the absence of tmRNA activity might be responsible for the delay in initiation of DNA replication. Analysis of the tagging sites within these substrates revealed a conserved nucleotide motif 5' of the tagging site, which is required for wild-type tmRNA tagging.


Assuntos
Caulobacter/metabolismo , Proteômica , RNA Bacteriano/metabolismo , Proteínas de Bactérias/metabolismo , Sequência de Bases , Histidina/metabolismo , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Oligopeptídeos/metabolismo , RNA Bacteriano/química , RNA Bacteriano/genética , Sequências Reguladoras de Ácido Ribonucleico/genética , Especificidade por Substrato
11.
Protein Sci ; 16(8): 1535-42, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17600141

RESUMO

A method to rapidly screen libraries of cyclic peptides in vivo for molecules with biological activity has been developed and used to isolate cyclic peptide inhibitors of the ClpXP protease. Fluorescence activated cell sorting was used in conjunction with a fluorescent reporter to isolate cyclic peptides that inhibit the proteolysis of tmRNA-tagged proteins in Escherichia coli. Inhibitors shared little sequence similarity and interfered with unexpected steps in the ClpXP mechanism in vitro. One cyclic peptide, IXP1, inhibited the degradation of unrelated ClpXP substrates and has bactericidal activity when added to growing cultures of Caulobacter crescentus, a model organism that requires ClpXP activity for viability. The screen used here could be adapted to identify cyclic peptide inhibitors of any enzyme that can be expressed in E. coli in conjunction with a fluorescent reporter.


Assuntos
Antibacterianos/isolamento & purificação , Proteínas de Bactérias/antagonistas & inibidores , Endopeptidase Clp/antagonistas & inibidores , Peptídeos Cíclicos/isolamento & purificação , Peptídeos Cíclicos/farmacologia , Inibidores de Proteases/isolamento & purificação , Inibidores de Proteases/farmacologia , Antibacterianos/química , Antibacterianos/farmacologia , Caulobacter crescentus/enzimologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Modelos Biológicos , Biblioteca de Peptídeos , Peptídeos Cíclicos/química , Inibidores de Proteases/química , Especificidade por Substrato
12.
Mol Microbiol ; 57(2): 565-75, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15978085

RESUMO

The production and removal of regulatory RNAs must be controlled to ensure proper physiological responses. SsrA RNA (tmRNA), a regulatory RNA conserved in all bacteria, is cell cycle regulated and is important for control of cell cycle progression in Caulobacter crescentus. We report that RNase R, a highly conserved 3' to 5' exoribonuclease, is required for the selective degradation of SsrA RNA in stalked cells. Purified RNase R degrades SsrA RNA in vitro, and is kinetically competent to account for all SsrA RNA turnover. SmpB, a tmRNA-binding protein, protects SsrA RNA from RNase R degradation in vitro, and the levels of SmpB protein during the cell cycle correlate with SsrA RNA stability. These results suggest that SmpB binding controls the timing of SsrA RNA degradation by RNase R. We propose a model for the regulated degradation of SsrA RNA in which RNase R degrades SsrA RNA from a non-tRNA-like 3' end, and SmpB specifically protects SsrA RNA from RNase R. This model explains the regulation of SsrA RNA in other bacteria, and suggests that a highly conserved regulatory mechanism controls SsrA activity.


Assuntos
Caulobacter crescentus/fisiologia , Endorribonucleases/metabolismo , Estabilidade de RNA , RNA Bacteriano/metabolismo , Proteínas de Ligação a RNA/metabolismo , Caulobacter crescentus/citologia , Caulobacter crescentus/metabolismo , Ciclo Celular , Exorribonucleases
13.
Int Rev Cytol ; 225: 33-89, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12696590

RESUMO

In organisms from Drosophila to mammals, the musculature is comprised of an elaborate array of distinct fibers that are generated by the fusion of committed myoblasts. These muscle fibers differ from each other in features that include location, pattern of innervation, site of attachment, and size. The sizes of the newly formed muscles of an embryo are controlled in large part by the number of cells that form the syncitial fiber. Over the past few decades, an extensive body of literature has described the process of myoblast fusion in vertebrates, relying primarily on the strengths of tissue culture model systems. More recently, genetic studies in Drosophila embryos have provided new insights into the process. Together, these studies define the steps necessary for myoblast differentiation, the acquisition of fusion competence, the recognition and adhesion between myoblasts, and the fusion of two lipid bilayers into one. In this review, we have attempted to combine insights from both Drosophila and vertebrate studies to trace the processes and molecules involved in myoblast fusion. Implicit in this approach is the assumption that fundamental aspects of myoblast fusion will be similar, independent of the organism in which it is occurring.


Assuntos
Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Membrana Celular/metabolismo , Fusão de Membrana/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/embriologia , Mioblastos Esqueléticos/metabolismo , Animais , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestrutura , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Embrião não Mamífero/ultraestrutura , Humanos , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestrutura , Mioblastos Esqueléticos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA