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.
MAbs ; 15(1): 2273018, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38050985

RESUMO

To exploit highly conserved and difficult drug targets, including multipass membrane proteins, monoclonal antibody discovery efforts increasingly rely on the advantages offered by divergent species such as rabbits, camelids, and chickens. Here, we provide an overview of antibody discovery technologies, analyze gaps in therapeutic antibodies that stem from the historic use of mice, and examine opportunities to exploit previously inaccessible targets through discovery now possible in alternate species. We summarize the clinical development of antibodies raised from divergent species, discussing how these animals enable robust immune responses against highly conserved binding sites and yield antibodies capable of penetrating functional pockets via long HCDR3 regions. We also discuss the value of pan-reactive molecules often produced by these hosts, and how these antibodies can be tested in accessible animal models, offering a faster path to clinical development.


Assuntos
Anticorpos Monoclonais , Galinhas , Animais , Camundongos , Coelhos , Epitopos , Anticorpos Monoclonais/uso terapêutico
2.
Comput Intell Neurosci ; 2022: 1391340, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36156969

RESUMO

In the current age of technology, various diseases in the body are also on the rise. Tumours that cause more discomfort in the body are set to increase the discomfort of most patients. Patients experience different effects depending on the tumour size and type. Future developments in the medical field are moving towards the development of tools based on IoT devices. These advances will in the future follow special features designed based on multiple machine learning developed by artificial intelligence. In that order, an improved algorithm named Internet of Things-based enhanced machine learning is proposed in this paper. What makes it special is that it involves separate functions to diagnose each type of tumour. It analyzes and calculates things like the size, shape, and location of the tumour. Cure from cancer is determined by the stage at which we find cancer. Early detection of cancer has the potential to cure quickly. At a saturation point, the proposed Internet of Things-based enhanced machine learning model achieved 94.56% of accuracy, 94.12% of precision, 94.98% of recall, 95.12% of F1-score, and 1856 ms of execution time. The simulation is conducted to test the efficacy of the model, and the results of the simulation show that the proposed Internet of Things-based enhanced machine learning obtains a higher rate of intelligence than other methods.


Assuntos
Internet das Coisas , Neoplasias , Algoritmos , Inteligência Artificial , Humanos , Internet , Aprendizado de Máquina , Neoplasias/diagnóstico , Neoplasias/terapia
3.
Biomed Res Int ; 2022: 7760734, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35978632

RESUMO

All organisms contain antimicrobial peptides (AMPs), which are a critical component of the innate immune system. These chemicals have the ability to suppress the growth of a variety of fungi, bacteria, and viruses. Because AMPs interact with structural components of the microbial cell membrane and have a wide range of cellular targets, bacteria are unlikely to be able to develop resistance to them in the short term. The underlying structure of AMPs is critical in determining the selectivity with which they target their respective targets. As far as we know, peptides have not been tested in a lab to see if they can fight bacteria, fungus, and viruses in real life. In this paper, we develop an artificial neural network (ANN) using a back propagation neural network (BPNN) that enables optimal classification of tendency of a peptide sequence that involves the activities of antifungal, antibacterial, or antiviral. The BPNN is trained on the datasets collected across different repositories and then the overfitting is avoided using particle swarm optimization (PSO) algorithm. Hence, at the time of testing, the BPNN clearly finds the predicted samples belonging to the same classes and this avoids the problem of finding the false positives. The simulation is conducted to test the efficacy of the model against various metrics that includes accuracy, precision, recall, and f1-measure. The effectiveness of the BPNN-PSO model in classifying instances at a faster rate than other techniques is demonstrated by its performance. The principle is straightforward, it is not difficult to programme, it converges more quickly, and it generally offers a superior solution.


Assuntos
Algoritmos , Redes Neurais de Computação , Antifúngicos , Simulação por Computador , Peptídeos
4.
J Virol ; 88(24): 14364-79, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25275138

RESUMO

UNLABELLED: Chikungunya virus (CHIKV) is a reemerging alphavirus that causes a debilitating arthritic disease and infects millions of people and for which no specific treatment is available. Like many alphaviruses, the structural targets on CHIKV that elicit a protective humoral immune response in humans are poorly defined. Here we used phage display against virus-like particles (VLPs) to isolate seven human monoclonal antibodies (MAbs) against the CHIKV envelope glycoproteins E2 and E1. One MAb, IM-CKV063, was highly neutralizing (50% inhibitory concentration, 7.4 ng/ml), demonstrated high-affinity binding (320 pM), and was capable of therapeutic and prophylactic protection in multiple animal models up to 24 h postexposure. Epitope mapping using a comprehensive shotgun mutagenesis library of 910 mutants with E2/E1 alanine mutations demonstrated that IM-CKV063 binds to an intersubunit conformational epitope on domain A, a functionally important region of E2. MAbs against the highly conserved fusion loop have not previously been reported but were also isolated in our studies. Fusion loop MAbs were broadly cross-reactive against diverse alphaviruses but were nonneutralizing. Fusion loop MAb reactivity was affected by temperature and reactivity conditions, suggesting that the fusion loop is hidden in infectious virions. Visualization of the binding sites of 15 different MAbs on the structure of E2/E1 revealed that all epitopes are located at the membrane-distal region of the E2/E1 spike. Interestingly, epitopes on the exposed topmost and outer surfaces of the E2/E1 trimer structure were neutralizing, whereas epitopes facing the interior of the trimer were not, providing a rationale for vaccine design and therapeutic MAb development using the intact CHIKV E2/E1 trimer. IMPORTANCE: CHIKV is the most important alphavirus affecting humans, resulting in a chronic arthritic condition that can persist for months or years. In recent years, millions of people have been infected globally, and the spread of CHIKV to the Americas is now beginning, with over 100,000 cases occurring in the Caribbean within 6 months of its arrival. Our study reports on seven human MAbs against the CHIKV envelope, including a highly protective MAb and rarely isolated fusion loop MAbs. Epitope mapping of these MAbs demonstrates how some E2/E1 epitopes are exposed or hidden from the human immune system and suggests a structural mechanism by which these MAbs protect (or fail to protect) against CHIKV infection. Our results suggest that the membrane-distal end of CHIKV E2/E1 is the primary target for the humoral immune response to CHIKV, and antibodies targeting the exposed topmost and outer surfaces of the E2/E1 trimer determine the neutralizing efficacy of this response.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus Chikungunya/imunologia , Epitopos/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Sítios de Ligação , Técnicas de Visualização da Superfície Celular , Febre de Chikungunya/prevenção & controle , Modelos Animais de Doenças , Mapeamento de Epitopos , Feminino , Humanos , Imunização Passiva , Camundongos Endogâmicos C57BL , Modelos Moleculares , Conformação Proteica , Análise de Sobrevida
5.
Antimicrob Agents Chemother ; 57(10): 4622-31, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23817385

RESUMO

The identification of novel antiretroviral agents is required to provide alternative treatment options for HIV-1-infected patients. The screening of a phenotypic cell-based viral replication assay led to the identification of a novel class of 4,5-dihydro-1H-pyrrolo[3,4-c]pyrazol-6-one (pyrrolopyrazolone) HIV-1 inhibitors, exemplified by two compounds: BI-1 and BI-2. These compounds inhibited early postentry stages of viral replication at a step(s) following reverse transcription but prior to 2 long terminal repeat (2-LTR) circle formation, suggesting that they may block nuclear targeting of the preintegration complex. Selection of viruses resistant to BI-2 revealed that substitutions at residues A105 and T107 within the capsid (CA) amino-terminal domain (CANTD) conferred high-level resistance to both compounds, implicating CA as the antiviral target. Direct binding of BI-1 and/or BI-2 to CANTD was demonstrated using isothermal titration calorimetry and nuclear magnetic resonance (NMR) chemical shift titration analyses. A high-resolution crystal structure of the BI-1:CANTD complex revealed that the inhibitor bound within a recently identified inhibitor binding pocket (CANTD site 2) between CA helices 4, 5, and 7, on the surface of the CANTD, that also corresponds to the binding site for the host factor CPSF-6. The functional consequences of BI-1 and BI-2 binding differ from previously characterized inhibitors that bind the same site since the BI compounds did not inhibit reverse transcription but stabilized preassembled CA complexes. Hence, this new class of antiviral compounds binds CA and may inhibit viral replication by stabilizing the viral capsid.


Assuntos
Fármacos Anti-HIV/farmacologia , Proteínas do Capsídeo/metabolismo , HIV-1/efeitos dos fármacos , Fármacos Anti-HIV/química , Linhagem Celular , Cristalografia por Raios X , HIV-1/fisiologia , Humanos , Espectroscopia de Ressonância Magnética , Reação em Cadeia da Polimerase , Replicação Viral/efeitos dos fármacos
6.
J Virol ; 87(19): 10679-86, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23885079

RESUMO

The influenza virus M2 protein is a well-validated yet underexploited proton-selective ion channel essential for influenza virus infectivity. Because M2 is a toxic viral ion channel, existing M2 inhibitors have been discovered through live virus inhibition or medicinal chemistry rather than M2-targeted high-throughput screening (HTS), and direct measurement of its activity has been limited to live cells or reconstituted lipid bilayers. Here, we describe a cell-free ion channel assay in which M2 ion channels are incorporated into virus-like particles (VLPs) and proton conductance is measured directly across the viral lipid bilayer, detecting changes in membrane potential, ion permeability, and ion channel function. Using this approach in high-throughput screening of over 100,000 compounds, we identified 19 M2-specific inhibitors, including two novel chemical scaffolds that inhibit both M2 function and influenza virus infectivity. Counterscreening for nonspecific disruption of viral bilayer ion permeability also identified a broad-spectrum antiviral compound that acts by disrupting the integrity of the viral membrane. In addition to its application to M2 and potentially other ion channels, this technology enables direct measurement of the electrochemical and biophysical characteristics of viral membranes.


Assuntos
Antivirais/farmacologia , Membrana Celular/virologia , Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Canais Iônicos/efeitos dos fármacos , Prótons , Proteínas da Matriz Viral/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Membrana Celular/metabolismo , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Concentração de Íons de Hidrogênio , Influenza Humana/tratamento farmacológico , Influenza Humana/patologia , Bicamadas Lipídicas/metabolismo , Bibliotecas de Moléculas Pequenas , Proteínas da Matriz Viral/metabolismo , Vírion
7.
J Virol ; 86(12): 6643-55, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22496222

RESUMO

The emergence of resistance to existing classes of antiretroviral drugs necessitates finding new HIV-1 targets for drug discovery. The viral capsid (CA) protein represents one such potential new target. CA is sufficient to form mature HIV-1 capsids in vitro, and extensive structure-function and mutational analyses of CA have shown that the proper assembly, morphology, and stability of the mature capsid core are essential for the infectivity of HIV-1 virions. Here we describe the development of an in vitro capsid assembly assay based on the association of CA-NC subunits on immobilized oligonucleotides. This assay was used to screen a compound library, yielding several different families of compounds that inhibited capsid assembly. Optimization of two chemical series, termed the benzodiazepines (BD) and the benzimidazoles (BM), resulted in compounds with potent antiviral activity against wild-type and drug-resistant HIV-1. Nuclear magnetic resonance (NMR) spectroscopic and X-ray crystallographic analyses showed that both series of inhibitors bound to the N-terminal domain of CA. These inhibitors induce the formation of a pocket that overlaps with the binding site for the previously reported CAP inhibitors but is expanded significantly by these new, more potent CA inhibitors. Virus release and electron microscopic (EM) studies showed that the BD compounds prevented virion release, whereas the BM compounds inhibited the formation of the mature capsid. Passage of virus in the presence of the inhibitors selected for resistance mutations that mapped to highly conserved residues surrounding the inhibitor binding pocket, but also to the C-terminal domain of CA. The resistance mutations selected by the two series differed, consistent with differences in their interactions within the pocket, and most also impaired virus replicative capacity. Resistance mutations had two modes of action, either directly impacting inhibitor binding affinity or apparently increasing the overall stability of the viral capsid without affecting inhibitor binding. These studies demonstrate that CA is a viable antiviral target and demonstrate that inhibitors that bind within the same site on CA can have distinct binding modes and mechanisms of action.


Assuntos
Fármacos Anti-HIV/farmacologia , Capsídeo/efeitos dos fármacos , Produtos do Gene gag/antagonistas & inibidores , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Benzimidazóis/farmacologia , Benzodiazepinas/farmacologia , Capsídeo/metabolismo , Linhagem Celular , Produtos do Gene gag/química , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Infecções por HIV/tratamento farmacológico , HIV-1/química , HIV-1/genética , HIV-1/fisiologia , Humanos , Estrutura Terciária de Proteína , Montagem de Vírus/efeitos dos fármacos
8.
Lancet ; 365(9477): 2122-4, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15964449

RESUMO

Tissue engineering has made considerable progress in the past decade, but advances have stopped short of clinical application for most tissues. We postulated that an obstacle in engineering human tissues is the limited replicative capacity of adult somatic cells. To test this hypothesis, the effectiveness of telomerase expression to extend cellular lifespan was assessed in a model of human vascular tissue engineering. Telomerase expression in vascular cells isolated from elderly patients enabled the successful culture of engineered autologous blood vessels. Engineered vessels may one day provide a source of bypass conduit for patients with atherosclerotic disease.


Assuntos
Vasos Sanguíneos , Engenharia Tecidual/métodos , Idoso , Vasos Sanguíneos/enzimologia , Técnicas de Cultura de Células , Proteínas de Ligação a DNA/biossíntese , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Veia Safena/citologia , Telomerase/biossíntese , Transfecção
9.
J Biol Chem ; 279(50): 51745-8, 2004 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-15381700

RESUMO

The addition of telomeric repeats to chromosome ends by the enzyme telomerase is a highly orchestrated process. Although much is known regarding telomerase catalytic activity in vitro, less is known about how this activity is regulated in vivo to ensure proper telomere elongation. One protein that appears to be involved in negatively regulating telomerase function in vivo is PinX1 because overexpression of PinX1 inhibits telomerase activity and causes telomere shortening. To understand the nature of this repression, we characterized the interactions among PinX1 and the core components of telomerase, the human telomerase reverse transcriptase (hTERT) and associated human telomerase RNA (hTR). We now show that in vitro PinX1 binds directly to the hTERT protein subunit, primarily to the hTR-binding domain, as well as to the hTR subunit. However, in a cellular context, the association of PinX1 with hTR is dependent on the presence of hTERT. Taken together, we suggest that PinX1 represses telomerase activity in vivo by binding to the assembled hTERT.hTR complex.


Assuntos
RNA não Traduzido/química , RNA não Traduzido/metabolismo , Telomerase/química , Telomerase/metabolismo , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas de Ligação a DNA , Humanos , Técnicas In Vitro , Ligação Proteica , Subunidades Proteicas , RNA/química , RNA/genética , RNA/metabolismo , RNA Longo não Codificante , RNA não Traduzido/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Telomerase/genética , Proteínas Supressoras de Tumor/genética
10.
EMBO Rep ; 4(6): 633-8, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12776184

RESUMO

There is a pressing need to develop methods to engineer small-calibre arteries for bypass surgery. We hypothesized that the rate-limiting step that has thwarted previous attempts to engineer such vessels from non-neonatal tissues is the limited proliferative capacity of smooth muscle cells (SMCs), which are the main cellular component of these vessels. Ectopic expression of the human telomerase reverse transcriptase subunit (hTERT) has been shown recently to extend the lifespan of certain human cells. We therefore introduced hTERT into human SMCs and found that the resulting cells proliferated far beyond their normal lifespan but retained characteristics of normal control SMCs. Importantly, using these non-neonatal SMCs, we were able to engineer mechanically robust human vessels, a crucial step towards creating arteries of clinical value for bypass surgery.


Assuntos
Artérias/fisiologia , Telomerase/metabolismo , Aorta/citologia , Artérias/metabolismo , Artérias/patologia , Western Blotting , Divisão Celular , Colágeno/metabolismo , Proteínas de Ligação a DNA , Endotélio Vascular/citologia , Humanos , Miócitos de Músculo Liso/citologia , Fenótipo , Retroviridae/genética , Fatores de Tempo , Engenharia Tecidual
11.
Oncogene ; 21(46): 7121-5, 2002 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-12370834

RESUMO

A hallmark of cancer cells is the ability to proliferate indefinitely. This acquisition of an immortal lifespan usually requires the activation of telomerase, the enzyme that elongates telomeres. Human telomerase is minimally composed of the reverse transcriptase subunit hTERT, and the RNA subunit hTR. While hTR is ubiquitously expressed in human cells, the hTERT subunit is generally transcriptionally repressed in most normal somatic cells, but is illegitimately activated to restore telomerase activity in cancer cells. Indeed, in the thousands of different human tumours assayed, 85% were scored positive for telomerase activity. However, the levels of telomerase activity detected in tumour samples can vary substantially and even some normal somatic cells have been found to have low levels of enzyme activity. As the functional significance of low levels of telomerase activity is unclear, we investigated whether there is a minimum level of telomerase activity required for tumourigenesis. Using mutants of hTERT that induce varying levels of telomerase activity, we show that there does indeed exist a threshold of activity required for the processes of immortalization, transformation and tumourigenesis. Thus, low levels of activity detected in certain somatic cells would not be expected to contribute to tumourigenesis, nor does the mere detection of telomerase in cancer cells necessarily signify an immortal lifespan.


Assuntos
Transformação Celular Neoplásica , Neoplasias/enzimologia , Telomerase/fisiologia , Antígenos Virais de Tumores/genética , Proteínas de Ligação a DNA , Genes ras , Humanos , Mutação , Neoplasias/patologia , Telomerase/química , Telomerase/genética
12.
Mol Cell Biol ; 22(17): 6234-46, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12167716

RESUMO

Most human cancer cells are thought to acquire the ability to divide beyond the capacity of normal somatic cells through illegitimately activating the gene hTERT, which encodes the catalytic subunit of telomerase. While telomerase reverse transcriptase (TERT) is conserved in most eukaryotes, mounting evidence suggests that the C terminus of the human protein may have functions unique to higher eukaryotes. To search for domains responsible for such functions, we assayed a panel of tandem substitution mutations encompassing this region of human TERT for in vitro and in vivo functionality. We found four clusters of mutations that inactivated the biochemical and biological functions of telomerase, separated by mutations that had little or no effect on enzyme activity. We also identified a region where mutations generate catalytically active but biologically inert proteins. This C-terminal region that dissociates activities of telomerase (C-DAT) does not appear to be involved in nuclear localization or protein multimerization. Instead, it appears that the C-DAT region is involved in a step of in vivo telomere synthesis after the assembly of a catalytically active enzyme. Intriguingly, all of the described regions reside in a portion of TERT that is dispensable for cellular viability in yeast, arguing for a divergent role of the C terminus in higher eukaryotes.


Assuntos
Telomerase/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Proteínas de Arabidopsis/química , Catálise , Domínio Catalítico , Linhagem Celular , Sequência Consenso , Proteínas de Ligação a DNA , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , RNA Mensageiro/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/fisiologia , Proteínas de Saccharomyces cerevisiae/química , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Relação Estrutura-Atividade , Telomerase/genética , Telomerase/fisiologia , Telômero/metabolismo
13.
J Biol Chem ; 277(27): 24764-70, 2002 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-11956201

RESUMO

Telomerase is the enzyme essential to complete the replication of the terminal DNA of most eukaryotic chromosomes. In humans, this enzyme is composed of the telomerase reverse transcriptase (hTERT) and telomerase RNA (hTR) subunits. hTR has been found in the nucleolus, a site of assembly of ribosomes as well as other ribonucleoproteins (RNPs). We therefore tested whether the hTERT component is also found in the nucleolus, where it could complex with the hTR RNA to form a functional enzyme. We report here that hTERT does indeed localize to the nucleolus, and we mapped the domain responsible for this localization to the hTR-binding region of the protein by deletion analysis. Substitution mutations in two of the three conserved hTR-binding domains in this nucleolar localization domain (NoLD) abolished nucleolar localization. However, another mutation that impeded hTR binding did not alter this subcellular localization. Additionally, wild type hTERT was detected in the nucleolus of cells that failed to express hTR. Taken together, we propose that the nucleolar localization of hTERT involves more than just the association with the hTR subunit. Furthermore, the coincidental targeting of both the hTR and hTERT subunits to the nucleolus supports the premise that the assembly of telomerase occurs in the nucleolus.


Assuntos
Telomerase/metabolismo , Adenocarcinoma , Sequência de Aminoácidos , Substituição de Aminoácidos , Neoplasias Ósseas , Domínio Catalítico , Primers do DNA , Proteínas de Ligação a DNA , Feminino , Humanos , Mutagênese Sítio-Dirigida , Osteossarcoma , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Telomerase/análise , Telomerase/química , Células Tumorais Cultivadas , Neoplasias do Colo do Útero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA