Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cancer Metastasis Rev ; 41(2): 335-346, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35588337

RESUMO

The identification of microbes enriched in the healthy lung has led to the compelling discovery that microbes may contribute to lung cancer pathogenesis. Here, we review the recent literature showing microbial associations with lung cancer as well as the functional features that have been identified in human and murine studies. Most biomarker data remain limited due to variable findings. However, multiple studies have found that lung tumors or ipsilateral airway samples have decreased α diversity compared to normal tissue. Specific genera, such as Veillonella and Streptococcus, were also found in association with lung tumors using multiple sampling methodologies. These microbes, which are generally found in the upper respiratory track, are associated with an IL-17 signature in the lung, potentially resulting in a pro-tumorigenic environment. Studies detailing these immune mechanisms are limited, and further investigation is necessary to delineate how these bacteria, their metabolites, and potentially tumor-associated neoantigens modulate the immune response in cancer.


Assuntos
Neoplasias Pulmonares , Microbiota , Animais , Bactérias , Biomarcadores , Humanos , Imunidade , Neoplasias Pulmonares/patologia , Camundongos , Microbiota/fisiologia
2.
Cancer Immunol Immunother ; 71(10): 2405-2420, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35217892

RESUMO

Human gut microbial species found to associate with clinical responses to immune checkpoint inhibitors (ICIs) are often tested in mice using fecal microbiota transfer (FMT), wherein tumor responses in recipient mice may recapitulate human responses to ICI treatment. However, many FMT studies have reported only limited methodological description, details of murine cohorts, and statistical methods. To investigate the reproducibility and robustness of gut microbial species that impact ICI responses, we performed human to germ-free mouse FMT using fecal samples from patients with non-small cell lung cancer who had a pathological response or nonresponse after neoadjuvant ICI treatment. R-FMT mice yielded greater anti-tumor responses in combination with anti-PD-L1 treatment compared to NR-FMT, although the magnitude varied depending on mouse cell line, sex, and individual experiment. Detailed investigation of post-FMT mouse microbiota using 16S rRNA amplicon sequencing, with models to classify and correct for biological variables, revealed a shared presence of the most highly abundant taxa between the human inocula and mice, though low abundance human taxa colonized mice more variably after FMT. Multiple Clostridium species also correlated with tumor outcome in individual anti-PD-L1-treated R-FMT mice. RNAseq analysis revealed differential expression of T and NK cell-related pathways in responding tumors, irrespective of FMT source, with enrichment of these cell types confirmed by immunohistochemistry. This study identifies several human gut microbial species that may play a role in clinical responses to ICIs and suggests attention to biological variables is needed to improve reproducibility and limit variability across experimental murine cohorts.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Transplante de Microbiota Fecal , Humanos , Camundongos , Terapia Neoadjuvante , RNA Ribossômico 16S/genética , Reprodutibilidade dos Testes
3.
Clin Cancer Res ; 27(9): 2571-2583, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33593881

RESUMO

PURPOSE: While immune checkpoint inhibitors (ICI) have revolutionized the treatment of cancer by producing durable antitumor responses, only 10%-30% of treated patients respond and the ability to predict clinical benefit remains elusive. Several studies, small in size and using variable analytic methods, suggest the gut microbiome may be a novel, modifiable biomarker for tumor response rates, but the specific bacteria or bacterial communities putatively impacting ICI responses have been inconsistent across the studied populations. EXPERIMENTAL DESIGN: We have reanalyzed the available raw 16S rRNA amplicon and metagenomic sequencing data across five recently published ICI studies (n = 303 unique patients) using a uniform computational approach. RESULTS: Herein, we identify novel bacterial signals associated with clinical responders (R) or nonresponders (NR) and develop an integrated microbiome prediction index. Unexpectedly, the NR-associated integrated index shows the strongest and most consistent signal using a random effects model and in a sensitivity and specificity analysis (P < 0.01). We subsequently tested the integrated index using validation cohorts across three distinct and diverse cancers (n = 105). CONCLUSIONS: Our analysis highlights the development of biomarkers for nonresponse, rather than response, in predicting ICI outcomes and suggests a new approach to identify patients who would benefit from microbiome-based interventions to improve response rates.


Assuntos
Biomarcadores , Biologia Computacional , Inibidores de Checkpoint Imunológico/farmacologia , Microbiota/efeitos dos fármacos , Bactérias/classificação , Bactérias/genética , Biologia Computacional/métodos , Microbioma Gastrointestinal , Genoma Bacteriano , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Metagenômica/métodos , Microbiota/genética , RNA Ribossômico 16S , Curva ROC , Reprodutibilidade dos Testes , Sequenciamento Completo do Genoma
4.
EBioMedicine ; 48: 642-647, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31597596

RESUMO

The microbiome is increasingly recognized for its role in multiple aspects of cancer development and treatment, specifically in response to checkpoint inhibitors. While checkpoint inhibitors have revolutionized cancer treatment by producing durable anti-tumor responses, only a minority of patients respond to the available immunotherapy drugs and accurate, sensitive and specific microbiome predictors of response to treatment remain elusive. Additionally, the specific mechanisms linking the microbiome and host immunological responses remain unclear. In this review, we examine the evidence for the gut microbiome's association with anti-tumor responses to checkpoint inhibitors in the treatment of melanoma, non-small cell lung cancer, and renal cell carcinoma. Furthermore, we discuss the current evidence available from murine models seeking to explain the immunological mechanisms that may drive this process. While this work is promising in defining the impact of gut microbiota in cancer treatment, many unanswered questions indicate the need for additional human and experimental studies.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Biomarcadores Tumorais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Melanoma/tratamento farmacológico , Microbiota , Animais , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/etiologia , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Humanos , Imunomodulação/efeitos dos fármacos , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Melanoma/etiologia , Microbiota/efeitos dos fármacos , Microbiota/imunologia , Terapia de Alvo Molecular , Prognóstico , Resultado do Tratamento
5.
Science ; 369(6510): 1427-1428, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32943510
6.
Nanomedicine (Lond) ; 9(11): 1613-24, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24195674

RESUMO

AIMS: Antibodies are the principal mediator of immunity against reinfection with viruses. Antibodies typically neutralize viruses by binding to virion particles in solution prior to attachment to susceptible cells. Once viruses enter cells, conventional antibodies cannot inhibit virus infection or replication. It is desirable to develop an efficient and nontoxic method for the introduction of virus-inhibiting antibodies into cells. MATERIALS & METHODS: In this article, we report a new method for the delivery of small recombinant antibody fragments into virus-infected cells using a dendrimer-based molecular transporter. RESULTS & CONCLUSION: The construct penetrated virus-infected cells efficiently and inhibited virus replication. This method provides a novel approach for the immediate delivery of inhibitory antibodies directed to virus proteins that are exposed only in the intracellular environment. This approach circumvents the current and rather complicated expression of inhibitory antibodies in cells following gene transfer.


Assuntos
Anticorpos/química , Nanomedicina/métodos , Vírion/química , Animais , Anticorpos Monoclonais/química , Transporte Biológico , Proteínas do Capsídeo/química , Citoplasma/metabolismo , Dendrímeros/química , Ensaio de Imunoadsorção Enzimática , Técnicas de Transferência de Genes , HIV-1/imunologia , Humanos , Fragmentos de Imunoglobulinas/química , Rim , Macaca mulatta , Espectroscopia de Ressonância Magnética , Microscopia Confocal , Testes de Neutralização , Peptídeos/química , RNA Interferente Pequeno/metabolismo , Rotavirus/metabolismo , Vírus/química
7.
Future Microbiol ; 8(1): 123-31, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23252497

RESUMO

Respiratory syncytial virus is a single-stranded RNA virus in the Paramyxoviridae family that preferentially assembles and buds from the apical surface of polarized epithelial cells, forming filamentous structures that contain both viral proteins and the genomic RNA. Recent studies have described both viral and host factors that are involved in ribonucleoprotein assembly and trafficking of viral proteins to the cell surface. At the cell surface, viral proteins assemble into filaments that probably require interactions between viral proteins, host proteins and the cell membrane. Finally, a membrane scission event must occur to release the free virion. This article will review the recent literature describing the mechanisms that drive respiratory syncytial virus assembly and budding.


Assuntos
Vírus Sinciciais Respiratórios/fisiologia , Montagem de Vírus , Animais , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Transporte Proteico , Ribonucleoproteínas/metabolismo , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/metabolismo , Liberação de Vírus
8.
PLoS One ; 7(7): e40826, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22808269

RESUMO

Respiratory syncytial virus (RSV) is a single-stranded RNA virus that assembles into viral filaments at the cell surface. Virus assembly often depends on the ability of a virus to use host proteins to accomplish viral tasks. Since the fusion protein cytoplasmic tail (FCT) is critical for viral filamentous assembly, we hypothesized that host proteins important for viral assembly may be recruited by the FCT. Using a yeast two-hybrid screen, we found that filamin A interacted with FCT, and mammalian cell experiments showed it localized to viral filaments but did not affect viral replication. Furthermore, we found that a number of actin-associated proteins also were excluded from viral filaments. Actin or tubulin cytoskeletal rearrangement was not necessary for F trafficking to the cell surface or for viral assembly into filaments, but was necessary for optimal viral replication and may be important for anchoring viral filaments. These findings suggest that RSV assembly into filaments occurs independently of actin polymerization and that viral proteins are the principal drivers for the mechanical tasks involved with formation of complex, structured RSV filaments at the host cell plasma membrane.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Interações Hospedeiro-Patógeno , Vírus Sinciciais Respiratórios/fisiologia , Vírion/metabolismo , Montagem de Vírus/fisiologia , Animais , Linhagem Celular , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Filaminas , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Transporte Proteico , Técnicas do Sistema de Duplo-Híbrido
9.
Virology ; 434(1): 129-36, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23062737

RESUMO

We describe the first example of combining bacterial artificial chromosome (BAC) recombination-mediated mutagenesis with reverse genetics for a negative strand RNA virus. A BAC-based respiratory syncytial virus (RSV) rescue system was established. An important advantage of this system is that RSV antigenomic cDNA was stabilized in the BAC vector. The RSV genotype chosen was A2-line19F, a chimeric strain previously shown to recapitulate in mice key features of RSV pathogenesis. We recovered two RSV reporter viruses, one expressing the red fluorescent protein monomeric Katushka 2 (A2-K-line19F) and one expressing Renilla luciferase (A2-RL-line19F). As proof of principle, we efficiently generated a RSV gene deletion mutant (A2-line19FΔNS1/NS2) and a point mutant (A2-K-line19F-I557V) by recombination-mediated BAC mutagenesis. Together with sequence-optimized helper expression plasmids, BAC-RSV is a stable, versatile, and efficient reverse genetics platform for generation of a recombinant Pneumovirus.


Assuntos
Mutagênese , Recombinação Genética , Vírus Sinciciais Respiratórios/genética , Genética Reversa/métodos , Animais , Fusão Gênica Artificial , Linhagem Celular , Cromossomos Artificiais Bacterianos , Genes Reporter , Vetores Genéticos , Humanos , Luciferases/análise , Luciferases/genética , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Proteína Vermelha Fluorescente
10.
mBio ; 3(1)2012.
Artigo em Inglês | MEDLINE | ID: mdl-22318318

RESUMO

UNLABELLED: Respiratory syncytial virus (RSV) is a single-stranded RNA virus in the Paramyxoviridae family that assembles into filamentous structures at the apical surface of polarized epithelial cells. These filaments contain viral genomic RNA and structural proteins, including the fusion (F) protein, matrix (M) protein, nucleoprotein (N), and phosphoprotein (P), while excluding F-actin. It is known that the F protein cytoplasmic tail (FCT) is necessary for filament formation, but the mechanism by which the FCT mediates assembly into filaments is not clear. We hypothesized that the FCT is necessary for interactions with other viral proteins in order to form filaments. In order to test this idea, we expressed the F protein with cytoplasmic tail (CT) truncations or specific point mutations and determined the abilities of these variant F proteins to form filaments independent of viral infection when coexpressed with M, N, and P. Deletion of the terminal three FCT residues (amino acids Phe-Ser-Asn) or mutation of the Phe residue resulted in a loss of filament formation but did not affect F-protein expression or trafficking to the cell surface. Filament formation could be restored by addition of residues Phe-Ser-Asn to an FCT deletion mutant and was unaffected by mutations to Ser or Asn residues. Second, deletion of residues Phe-Ser-Asn or mutation of the Phe residue resulted in a loss of M, N, and P incorporation into virus-like particles. These data suggest that a C-terminal Phe residue in the FCT mediates assembly through incorporation of internal virion proteins into virus filaments at the cell surface. IMPORTANCE: Respiratory syncytial virus (RSV) is a leading cause of bronchiolitis and pneumonia in infants and the elderly worldwide. There is no licensed RSV vaccine and only limited therapeutics for use in infected patients. Many aspects of the RSV life cycle have been studied, but the mechanisms that drive RSV assembly at the cell surface are not well understood. This study provides evidence that a specific residue in the RSV fusion protein cytoplasmic tail coordinates assembly into viral filaments by mediating the incorporation of internal virion proteins. Understanding the mechanisms that drive RSV assembly could lead to targeted development of novel antiviral drugs. Moreover, since RSV exits infected cells in an ESCRT (endosomal sorting complexes required for transport)-independent manner, these studies may contribute new knowledge about a general strategy by which ESCRT-independent viruses mediate outward bud formation using viral protein-mediated mechanisms during assembly and budding.


Assuntos
Fenilalanina , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Proteínas Virais de Fusão/genética , Montagem de Vírus , Sequência de Aminoácidos , Linhagem Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Citoesqueleto/ultraestrutura , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação Puntual , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Recombinantes , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/ultraestrutura , Alinhamento de Sequência , Proteínas Virais de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA